SINGLE-BUFFER COMPOSITIONS FOR NUCLEIC ACID DETECTION

Abstract
The present disclosure provides single-buffer systems that enable efficient and rapid amplification and programmable nuclease enzyme mediated reactions. The single-buffer systems and reagent compositions therein may be used for assaying for a nucleic acid sequence from a sample.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in XML format and is hereby incorporated by reference in its entirety. Said XML copy, created on Jul. 7, 2023, is named 203477-715301_US_SL.xml and is 640,567 bytes in size.


BACKGROUND

Detection of ailments, especially at the early stages of disease or infection, can provide guidance on treatment or intervention to reduce the progression or transmission of said ailments. Such ailments can be detected at the point of need by devices capable of running diagnostic assays. Various biological species associated with an organism, disease state, phenotype, or genotype can be detected by these devices. In particular, there is a need for rapid and low cost point-of-care testing for detection of viral infections, for example, including human immunodeficiency virus (HIV), SARS and MERS coronaviruses, influenza H1N1 virus, Ebola virus (EBOV), Zika virus (ZIKV), and SARS-COV-2.


Despite decades of continuous and rapid development in nucleic acid diagnostics, nucleic acid detection is typically time intensive, error prone, and susceptible to contamination. Many of these problems stem from the multi-condition requirements requisite for multi-reaction detection schemes. Nucleic acid targets are often present in low copy numbers and as minor constituents in complex samples. Therefore, detection of these targets often requires amplification prior to detection.


However, amplification and detection reagents often comprise stringent condition requirements that are not cross-compatible, necessitating buffer exchange and sample transfer steps that can increase time and user input requirements and result in contamination and sample volume loss.


SUMMARY

Described herein, in certain embodiments, is a system for detecting a target nucleic acid, comprising a buffer comprising: (i) reagents for an amplification reaction targeting the target nucleic acid; and (ii) reagents for a DETECTR reaction targeting the target nucleic acid, comprising a programmable nuclease, a non-naturally occurring guide nucleic acid, and a reporter, wherein the non-naturally occurring guide nucleic acid comprises a sequence that hybridizes to a segment of the target nucleic acid, wherein at least 1 nM of the reporter undergoes transcollateral cleavage within one hour of addition of at least 5000 copies of the target nucleic acid to the system. In some examples, at least 5 nM of the reporter undergoes transcollateral cleavage within one hour of addition of at least 5000 copies of the target nucleic acid to the system. In some examples, at least 10 nM of the reporter undergoes transcollateral cleavage within one hour of addition of at least 5000 copies of the target nucleic acid to the system. In some embodiments, at least 1 nM of the reporter undergoes transcollateral cleavage within one hour of addition of at least 1000 copies of the target nucleic acid to the system. In some embodiments, at least 5 nM of the reporter undergoes transcollateral cleavage within one hour of addition of at least 100 copies of the target nucleic acid to the system. In some embodiments, the time to completion for the amplification and DETECTR reactions is each less than 45 minutes when performed in the buffer. In some embodiments, the amplification and DETECTR reagents have half-lives of greater than 1 week at room temperature. In some embodiments, the activities of the amplification reagents and the DETECTR reagents diminishes by less than 10% following a freeze-thaw cycle. In some embodiments, the reagents for the amplification reaction comprise reagents for thermal cycling amplification. In some embodiments, the reagents for the amplification reaction comprise reagents for isothermal amplification. In some embodiments, the reagents for the amplification reaction comprise reagents for transcription mediated amplification (TMA), helicase dependent amplification (HDA), circular helicase dependent amplification (cHDA), strand displacement amplification (SDA), loop mediated amplification (LAMP), exponential amplification reaction (EXPAR), rolling circle amplification (RCA), ligase chain reaction (LCR), simple method amplifying RNA targets (SMART), single primer isothermal amplification (SPIA), multiple displacement amplification (MDA), nucleic acid sequence based amplification (NASBA), hinge-initiated primer-dependent amplification of nucleic acids (HIP), nicking enzyme amplification reaction (NEAR), or improved multiple displacement amplification (IMDA). In some embodiments, the reagents for the amplification reaction comprise reagents for loop mediated amplification (LAMP). In some embodiments, the system further comprises an activator for the amplification reaction. In some embodiments, the activator for the amplification reaction comprises a magnesium or calcium salt. In some embodiments, the programmable nuclease comprises at least 60% sequence identity to SEQ ID NO: 18-170 or 221-268. In some embodiments, the programmable nuclease comprises a RuvC catalytic domain. In some embodiments, the programmable nuclease is a type V CRISPR/Cas effector protein. In some embodiments, the type V CRISPR/Cas effector protein is a Cas12 protein. In some embodiments, the Cas12 protein comprises a Cas12a polypeptide, a Cas12b polypeptide, a Cas12c polypeptide, a Cas12d polypeptide, a Cas12e polypeptide, a C2c4 polypeptide, a C2c8 polypeptide, a C2c5 polypeptide, a C2c10 polypeptide, and a C2c9 polypeptide. In some embodiments, the Cas12 protein has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 18-SEQ ID NO: 60. In some embodiments, the Cas12 protein is selected from SEQ ID NO: 18-SEQ ID NO: 60. In some embodiments, the type V CRISPR/Cas effector protein is a Cas14 protein. In some embodiments, the Cas14 protein comprises a Cas14a polypeptide, a Cas14b polypeptide, a Cas14c polypeptide, a Cas14d polypeptide, a Cas14e polypeptide, a Cas14f polypeptide, a Cas14g polypeptide, a Cas14h polypeptide, a Cas14i polypeptide, a Cas14j polypeptide, or a Cas14k polypeptide. In some embodiments, the Cas14 protein has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 61-SEQ ID NO: 152. In some embodiments, the Cas14 protein is selected from SEQ ID NO: 61-SEQ ID NO: 152. In some embodiments, the type V CRISPR/Cas effector protein is a CasΦ protein. In some embodiments, the CasΦ protein has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 221-SEQ ID NO: 268. In some embodiments, the CasΦ protein is selected from SEQ ID NO: 221-SEQ ID NO: 268. In some embodiments, the system further comprises a reverse transcriptase, an oligonucleotide primer, and dNTPs for reverse transcribing the target nucleic acid. In some embodiments, the buffer is a lysis buffer. In some embodiments, the viscosity of the buffer is at least 5 centipoise (cP). In some embodiments, the buffer comprises a pH of 7.5 to 8.5, at least 10 mM of a buffering agent, at least 1 mM ammonium acetate, at least 10 mM potassium acetate, at least 2.5 mM magnesium acetate, and at least 0.5% glycerol. In some embodiments, the buffer comprises a pH of 7.7 to 8.3. In some embodiments, the buffer comprises a pH of 7.85 to 8.15. In some embodiments, the buffering agent comprises HEPES, imidazole, TRIS-HCl, or phosphate. In some embodiments, the buffer further comprises at least 0.05% by volume of a detergent. In some embodiments, the detergent comprises Tween 20. In some embodiments, the buffer comprises a pH of 7.5 to 8.5, at least 5 mM of a buffering agent, at least 20 mM potassium acetate, at least 2.5 mM magnesium acetate, and at least 0.5% glycerol. In some embodiments, the buffer comprises a pH of 7.7 to 8.3. In some embodiments, the buffer comprises a pH of 7.85 to 8.15. In some embodiments, the buffering agent comprises phosphate or TRIS-HCl. In some embodiments, the buffer comprises at least 1 mM ammonium sulfate. In some embodiments, the buffer comprises at least 0.05% by volume of a detergent. In some embodiments, the detergent comprises Tween 20. In some embodiments, the buffer comprises a pH of 7.25 to 8.75, at least 5 mM of a buffering agent, at least 7.5 mM potassium acetate, at least 1 mM magnesium acetate, and at least 0.5% glycerol. In some embodiments, the buffering agent comprises phosphate. In some embodiments, the buffer comprises a pH of 7.5 to 8.5. In some embodiments, the buffer comprises a pH of 7.75 to 8.25. In some embodiments, the buffer further comprises at least 1 mM ammonium sulfate. In some embodiments, the buffer further comprises at least 0.05% by volume of a detergent. In some embodiments, the detergent comprises Tween 20.


In some embodiments, described herein is a system for detecting a target nucleic acid, comprising a buffer comprising: (i) amplification reagents for an amplification reaction targeting the target nucleic acid; and (ii) detection reagents for a detection reaction targeting the target nucleic acid; wherein the amplification reagents comprise one or more oligonucleotide primers, and a DNA polymerase; wherein the detection reagents comprise a programmable nuclease, a non-naturally occurring guide nucleic acid, and reporters; wherein the non-naturally occurring guide nucleic acid comprises a sequence that hybridizes to a segment of the target nucleic acid or DNA amplicons thereof; wherein the amplification reagents are present in amounts effective to amplify the target nucleic acid in a test sample to produce DNA amplicons of the target nucleic acid; and wherein the programmable nuclease and non-naturally occurring guide nucleic acid form a complex in the buffer that is activated upon binding one of the DNA amplicons to induce detectable transcollateral cleavage of the reporters. In some embodiments, (a) at least 1 nM of the reporters undergo transcollateral cleavage within one hour of addition of at least 5000 copies of the target nucleic acid to the system; (b) at least 5 nM of the reporters undergo transcollateral cleavage within one hour of addition of at least 5000 copies of the target nucleic acid to the system; (c) at least 10 nM of the reporters undergo transcollateral cleavage within one hour of addition of at least 5000 copies of the target nucleic acid to the system; (d) at least 1 nM of the reporters undergo transcollateral cleavage within one hour of addition of at least 1000 copies of the target nucleic acid to the system; or (e) at least 1 nM of the reporters undergo transcollateral cleavage within one hour of addition of at least 1000 copies of the target nucleic acid to the system. In some embodiments, the amplification reagents and detection reagents are present in amounts effective to produce a detectable signal in less than 45 minutes in the presence of the target nucleic acid. In some embodiments, the amplification and detection reagents have half-lives of greater than 1 week at room temperature. In some embodiments, the activities of the amplification reagents and the detection reagents diminishes by less than 10% following a freeze-thaw cycle. In some embodiments, the amplification reagents comprise reagents for isothermal amplification. In some embodiments, the amplification reagents comprise reagents for transcription mediated amplification (TMA), helicase dependent amplification (HDA), circular helicase dependent amplification (cHDA), strand displacement amplification (SDA), loop mediated amplification (LAMP), exponential amplification reaction (EXPAR), rolling circle amplification (RCA), ligase chain reaction (LCR), simple method amplifying RNA targets (SMART), single primer isothermal amplification (SPIA), multiple displacement amplification (MDA), nucleic acid sequence based amplification (NASBA), hinge-initiated primer-dependent amplification of nucleic acids (HIP), nicking enzyme amplification reaction (NEAR), or improved multiple displacement amplification (IMDA). In some embodiments, the amplification reagents comprise reagents for loop mediated amplification (LAMP), and further wherein (a) the one or more primers comprise a first primer and a second primer targeted to the target nucleic acid; (b) the first primer comprises a 5′ region that is complementary to a sequence generated by extension of the first primer; (c) the second primer comprises a 5′ region that is complementary to a sequence generated by extension of the second primer; and (d) the DNA polymerase is a strand-displacing DNA polymerase. In some embodiments, the system further comprises an activator for the amplification reaction, such as a magnesium or calcium salt. In some embodiments, the programmable nuclease comprises at least 60% sequence identity to SEQ ID NO: 18-170 or 221-268. In some embodiments, the programmable nuclease comprises an RuvC catalytic domain. In some embodiments, the programmable nuclease is a type V CRISPR/Cas effector protein. In some embodiments, the type V CRISPR/Cas effector protein is a Cas12 protein, such as (a) a Cas12a polypeptide, a Cas12b polypeptide, a Cas12c polypeptide, a Cas12d polypeptide, a Cas12e polypeptide, a C2c4 polypeptide, a C2c8 polypeptide, a C2c5 polypeptide, a C2c10 polypeptide, and a C2c9 polypeptide; (b) a protein that has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 18-SEQ ID NO: 60; or (c) a protein having a sequence selected from SEQ ID NO: 18-SEQ ID NO: 60. In some embodiments, the type V CRISPR/Cas effector protein is a Cas14 protein, such as (a) a Cas14a polypeptide, a Cas14b polypeptide, a Cas14c polypeptide, a Cas14d polypeptide, a Cas14e polypeptide, a Cas14f polypeptide, a Cas14g polypeptide, a Cas14h polypeptide, a Cas14i polypeptide, a Cas14j polypeptide, or a Cas14k polypeptide; (b) a protein that has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 61-SEQ ID NO: 152; or (c) a protein having a sequence selected from SEQ ID NO: 61-SEQ ID NO: 152. In some embodiments, the type V CRISPR/Cas effector protein is a CasΦ protein, such as (a) a protein that has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 221-SEQ ID NO: 268; or (b) a protein having a sequence selected from SEQ ID NO: 221-SEQ ID NO: 268. In some embodiments, the system further comprises a reverse transcriptase, an oligonucleotide primer, and dNTPs for reverse transcribing the target nucleic acid. In some embodiments, the buffer is a lysis buffer. In some embodiments, the viscosity of the buffer is at least 5 centipoise (cP). In some embodiments, the buffer comprises a pH of 7.5 to 8.5, at least 10 mM of a buffering agent, at least 1 mM ammonium acetate, at least 10 mM potassium acetate, at least 2.5 mM magnesium acetate, and at least 0.5% glycerol; and optionally: (a) the buffer comprises a pH of 7.7 to 8.3, or a pH of 7.85 to 8.15; (b) the buffering agent comprises HEPES, imidazole, TRIS-HCl, or phosphate; and/or (c) the buffer further comprises at least 0.05% by volume of a detergent, optionally wherein the detergent comprises Tween 20. In some embodiments, the buffer comprises a pH of 7.5 to 8.5, at least 5 mM of a buffering agent, at least 20 mM potassium acetate, at least 2.5 mM magnesium acetate, and at least 0.5% glycerol; and optionally: (a) the buffer comprises a pH of 7.7 to 8.3, or a pH of 7.85 to 8.15; (b) the buffering agent comprises phosphate or TRIS-HCl; (c) the buffer comprises at least 1 mM ammonium sulfate; and/or (d) the buffer comprises at least 0.05% by volume of a detergent, optionally wherein the detergent comprises Tween 20. In some embodiments, the buffer comprises a pH of 7.25 to 8.75, at least 5 mM of a buffering agent, at least 7.5 mM potassium acetate, at least 1 mM magnesium acetate, and at least 0.5% glycerol; and optionally: (a) the buffering agent comprises phosphate; (b) the buffer comprises a pH of 7.5 to 8.5, or a pH of 7.75 to 8.25; (c) the buffer further comprises at least 1 mM ammonium sulfate; and/or (d) the buffer further comprises at least 0.05% by volume of a detergent, optionally wherein the detergent comprises Tween 20.


In some embodiments, the system further comprises a circular template with internal complementarity formed from a single polynucleotide strand, wherein: (a) the circular template comprises a first portion with complementarity to one of the one or more oligonucleotide primers and a second portion with complementarity to a portion of the target nucleic acid; (b) the internal complementarity comprises part of the first portion and part of the second portion; (c) the second portion has a total length that is longer than a combined length of the first portion and second portion that are within the internal complementarity; and (d) the circular template undergoes a conformational change upon hybridization to the target nucleic acid to expose the first portion to hybridization to the oligonucleotide primer. In some embodiments, the system further comprises a circular template, wherein: (a) the circular template comprises a first portion with complementarity to one of the one or more oligonucleotide primers and a second portion with complementarity to the target nucleic acid; (b) the oligonucleotide primer complementary to the first portion comprises a blocking motif at its 3′ end; and (c) the oligonucleotide primer complementary to the first portion undergoes cleavage to remove the blocking motif by the programmable nuclease in the presence of the target nucleic acid.


In some embodiments, the system further comprises a polymer matrix, wherein the polymer matrix is complexed with the reporters. In some embodiments, the polymer matrix is formed from copolymerization of at least a first plurality of monomers with the reporters. In some embodiments, the polymer matrix comprises a hydrogel.


In some embodiments, the buffer of system further comprises: (a) one or more of betaine monohydrate, acetamide, GABA, L-proline, beta-alanine, 6-aminohexanoic acid, urea, methylurea, ethylurea, hypotaurine, NDSB-256, and ammonium acetate; (b) one or more of trehalose, xylitol, D-sorbitol, sucrose, and trimethylamine N-oxide dihydrate; and/or (c) trimethylamine N-oxide dihydrate.


In some embodiments, described herein is a system for detecting a target nucleic acid, comprising reagents in a buffer, wherein (a) the reagents comprise hairpin polynucleotides, programmable nucleases, non-naturally occurring guide nucleic acids, and reporters; (b) each hairpin polynucleotide comprises one or more RNA loops, a first portion comprising DNA, and a second portion joined to the first portion by one of the one or more RNA loops; (c) each non-naturally occurring guide nucleic acid comprises a sequence that hybridizes to a segment of the target nucleic acid; (d) in each hairpin polynucleotide, the second portion of the hairpin polynucleotide hybridizes to a segment of the first portion; (e) the programmable nucleases and non-naturally occurring guide nucleic acids form complexes in the buffer that are activated upon binding the target nucleic acid; (f) an activated programmable nuclease is effective to induce (i) transcollateral cleavage of the one or more RNA loops, and (ii) detectable transcollateral cleavage of the reporters; and (g) cleavage of the one or more RNA loops of one of the hairpin polynucleotides is effective to release the first portion of the hairpin polynucleotide to hybridize with one of the non-naturally occurring guide nucleic acids and form a further activated programmable nuclease. In some embodiments, the one or more RNA loops comprise a first RNA loop joining the first portion and the second portion, and a second RNA loop joining the first portion and a third portion; wherein the third portion hybridizes to a different segment of the first portion. In some embodiments, the second portion comprises RNA, DNA, or both.


In some embodiments described herein is a method of assaying for a target nucleic acid in a sample, the method comprising: (a) amplifying a portion of the target nucleic acid; (b) performing a DETECTR reaction targeting the target nucleic acid, comprising contacting the target nucleic acid with a programmable nuclease, a non-naturally occurring guide nucleic acid that hybridizes to a segment of the target nucleic acid, and a reporter, wherein the contacting the sample to reagents for amplifying and the contacting the sample to reagents for the DETECTR reaction are performed in the same reaction volume; and (c) assaying for a change in a signal, wherein the change in the signal is produced by cleavage of the reporter. In some embodiments, the amplifying and the DETECTR reaction occur simultaneously. In some embodiments, the assaying comprises measuring the rate of the change in the signal. In some embodiments, the change in the signal identifies a concentration of the target nucleic acid in the sample. In some embodiments, the amplifying comprises thermal cycling amplification. In some embodiments, the amplifying comprises isothermal amplification. In some embodiments, the amplifying comprises transcription mediated amplification (TMA), helicase dependent amplification (HDA), circular helicase dependent amplification (cHDA), strand displacement amplification (SDA), loop mediated amplification (LAMP), exponential amplification reaction (EXPAR), rolling circle amplification (RCA), ligase chain reaction (LCR), simple method amplifying RNA targets (SMART), single primer isothermal amplification (SPIA), multiple displacement amplification (MDA), nucleic acid sequence based amplification (NASBA), hinge-initiated primer-dependent amplification of nucleic acids (HIP), nicking enzyme amplification reaction (NEAR), or improved multiple displacement amplification (IMDA). In some embodiments, the amplifying comprises loop mediated amplification (LAMP). In some embodiments, the amplifying comprises providing a plurality of primers that target different portions of the target nucleic acid. In some embodiments, the reagents for amplification comprise a forward inner primer (FIP) primer, a backward inner primer (BIP) primer, a forward loop primer (LF) primer, and a backward loop primer (LB primer). In some embodiments, the method comprises reverse transcribing the target nucleic acid. In some embodiments, the reverse transcribing comprises contacting the sample to reagents for reverse transcription, and wherein the reagents for reverse transcription comprise a reverse transcriptase, an oligonucleotide primer, and dNTPs. In some embodiments, the programmable nuclease comprises a RuvC catalytic domain. In some embodiments, the programmable nuclease has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 397-SEQ ID NO: 423; or is selected from SEQ ID NO: 397-SEQ ID NO: 423.


In some embodiments, the programmable nuclease is a type V CRISPR/Cas effector protein. In some embodiments, the type V CRISPR/Cas effector protein is a Cas12 protein. In some embodiments, the Cas12 protein comprises a Cas12a polypeptide, a Cas12b polypeptide, a Cas12c polypeptide, a Cas12d polypeptide, a Cas12e polypeptide, a C2c4 polypeptide, a C2c8 polypeptide, a C2c5 polypeptide, a C2c10 polypeptide, and a C2c9 polypeptide. In some embodiments, the Cas12 protein has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 18-SEQ ID NO: 60. In some embodiments, the Cas12 protein is selected from SEQ ID NO: 18-SEQ ID NO: 60. In some embodiments, the type V CRISPR/Cas effector protein is a Cas14 protein. In some embodiments, the Cas14 protein comprises a Cas14a polypeptide, a Cas14b polypeptide, a Cas14c polypeptide, a Cas14d polypeptide, a Cas14e polypeptide, a Cas14f polypeptide, a Cas14g polypeptide, a Cas14h polypeptide, a Cas14i polypeptide, a Cas14j polypeptide, or a Cas14k polypeptide. In some embodiments, the Cas14 protein has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 61-SEQ ID NO: 152. In some embodiments, the Cas14 protein is selected from SEQ ID NO: 61-SEQ ID NO: 152. In some embodiments, the type V CRISPR/Cas effector protein is a CasΦ protein. In some embodiments, the CasΦ protein has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 221-SEQ ID NO: 268. In some embodiments, the Cas protein is selected from SEQ ID NO: 221-SEQ ID NO: 268. In some embodiments, the method further comprises lysing a cell or virus comprising the target nucleic acid. In some embodiments, the lysing is performed in the same reaction volume as the amplifying and the DETECTR reaction. In some embodiments, the change in the signal comprises a change in a calorimetric, potentiometric, amperometric, or piezo-electric signal. In some embodiments, the change in the signal comprises a change in a colorimetric signal (e.g., an increase in intensity, a decrease in intensity, a change in color, etc.). In some embodiments, the change in the signal comprises a fluorescence signal (e.g., an increase in intensity, a decrease in intensity, a change in phase or wavelength, etc.). In some embodiments, the assaying comprises detecting the change in a signal with a smartphone. In some embodiments, the reaction volume comprises a viscosity of at least 5 cP. In some embodiments, the reaction volume comprises a total dissolved solids concentration of at least 200 mM.


In some embodiments, described herein is a method of assaying for a target nucleic acid in a sample, the method comprising: (a) amplifying a portion of the target nucleic acid with a DNA polymerase to produce DNA amplicons of the target nucleic acid; (b) forming a complex comprising one of the DNA amplicons, a programmable nuclease, and a non-naturally occurring guide nucleic acid that hybridizes to a segment of the DNA amplicon, thereby activating the programmable nuclease; (c) cleaving reporters with the activated programmable nuclease; and (d) detecting a change in a signal, wherein the change in the signal is produced by cleavage of the reporters; wherein the target nucleic acid and reagents for the amplifying and cleaving are present in the same reaction volume. In some embodiments, the amplifying and the cleaving occur simultaneously. In some embodiments, the method further comprises measuring the rate of the change in the signal. In some embodiments, the method further comprises measuring a concentration of the target nucleic acid in the sample based on the change in the signal. In some embodiments, the amplifying comprises isothermal amplification. In some embodiments, the amplifying comprises transcription mediated amplification (TMA), helicase dependent amplification (HDA), circular helicase dependent amplification (cHDA), strand displacement amplification (SDA), loop mediated amplification (LAMP), exponential amplification reaction (EXPAR), rolling circle amplification (RCA), ligase chain reaction (LCR), simple method amplifying RNA targets (SMART), single primer isothermal amplification (SPIA), multiple displacement amplification (MDA), nucleic acid sequence based amplification (NASBA), hinge-initiated primer-dependent amplification of nucleic acids (HIP), nicking enzyme amplification reaction (NEAR), or improved multiple displacement amplification (IMDA). In some embodiments, the amplifying comprises loop mediated amplification (LAMP), wherein the LAMP comprises amplification with a first primer and a second primer targeted to the target nucleic acid and a strand-displacing polymerase, wherein the first primer comprises a 5′ region that is complementary to a sequence generated by extension of the first primer, and wherein the second primer comprises a 5′ region that is complementary to a sequence generated by extension of the second primer. In some embodiments, the amplifying comprises providing a plurality of primers that target different portions of the target nucleic acid. In some embodiments, reagents for the amplification comprise a FIP primer, a BIP primer, a LF primer, and a LB primer. In some embodiments, the amplifying comprises reverse transcribing the target nucleic acid. In some embodiments, the programmable nuclease comprises a RuvC catalytic domain. In some embodiments, the programmable nuclease has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 397-SEQ ID NO: 423; or is selected from SEQ ID NO: 397-SEQ ID NO: 423.


In some embodiments, the programmable nuclease is a type V CRISPR/Cas effector protein. In some embodiments, the type V CRISPR/Cas effector protein is a Cas12 protein, such as (a) a Cas12a polypeptide, a Cas12b polypeptide, a Cas12c polypeptide, a Cas12d polypeptide, a Cas12e polypeptide, a C2c4 polypeptide, a C2c8 polypeptide, a C2c5 polypeptide, a C2c10 polypeptide, and a C2c9 polypeptide; (b) a protein that has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 18-SEQ ID NO: 60; or (c) a protein having a sequence selected from SEQ ID NO: 18-SEQ ID NO: 60. In some embodiments, the type V CRISPR/Cas effector protein is a Cas14 protein, such as (a) a Cas14a polypeptide, a Cas14b polypeptide, a Cas14c polypeptide, a Cas14d polypeptide, a Cas14e polypeptide, a Cas14f polypeptide, a Cas14g polypeptide, a Cas14h polypeptide, a Cas14i polypeptide, a Cas14j polypeptide, or a Cas14k polypeptide; (b) a protein that has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 61-SEQ ID NO: 152; or (c) a protein having a sequence selected from SEQ ID NO: 61-SEQ ID NO: 152. In some embodiments, the type V CRISPR/Cas effector protein is a CasΦ protein, such as (a) a protein that has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, or at least 99% sequence identity to any one of SEQ ID NO: 221-SEQ ID NO: 268; or (b) a protein having a sequence selected from SEQ ID NO: 221-SEQ ID NO: 268. In some embodiments, the method further comprises lysing a cell or virus comprising the target nucleic acid. In some embodiments, the lysing is performed in the same reaction volume as the amplifying and the cleaving. In some embodiments, the change in the signal comprises a change in a calorimetric, potentiometric, amperometric, or piezo-electric signal. In some embodiments, the change in the signal comprises a change in a colorimetric signal. In some embodiments, the change in the signal comprises a fluorescence signal. In some embodiments, the assaying comprises detecting the change in a signal with a smartphone. In some embodiments, the reaction volume comprises a viscosity of at least 5 cP. In some embodiments, the reaction volume comprises a total dissolved solids concentration of at least 200 mM. In some embodiments, the system further comprises Thermostable inorganic pyrophosphatase (TIPP). In some embodiments, the method further comprises signal enhancement via hydrolysis of inorganic pyrophosphates, such as by enzymatic hydrolysis. In some embodiments, the amplification and the cleaving are carried out in the presence of TIPP, and wherein the signal is enhanced by the hydrolysis of inorganic pyrophosphates by TIPP, as compared to the same reaction carried out without TIPP. In some embodiments, the reaction volume further comprises Thermostable inorganic pyrophosphatase (TIPP).


In some embodiments, the amplifying comprises amplification of a circular template with an oligonucleotide primer, and further wherein: (a) the circular template comprises a single polynucleotide strand having internal complementarity; (b) the circular template comprises a first portion with complementarity to the primer and a second portion with complementarity to a portion of the target nucleic acid; (c) the internal complementarity comprises part of the first portion and part of the second portion; (d) the second portion has a total length that is longer than a combined length of the first portion and second portion that are within the internal complementarity; (e) the circular template undergoes a conformational change upon hybridization to the target nucleic acid to expose the first portion to hybridization to the oligonucleotide primer; and (f) extension of the oligonucleotide primer along the circular template produces the DNA amplicons. In some embodiments, the amplifying comprises amplification of a circular template with an oligonucleotide primer, and further wherein: (a) the circular template comprises a first portion with complementarity to the oligonucleotide primer and a second portion with complementarity to the target nucleic acid; (b) the oligonucleotide primer complementary to the first portion comprises a blocking motif at its 3′ end; (c) the oligonucleotide primer undergoes cleavage to remove the blocking motif by the programmable nuclease in the presence of the target nucleic acid; and (d) extension of the oligonucleotide primer along the circular template produces the DNA amplicons. In some embodiments, the amplification is isothermal. In some embodiments, the DNA polymerase is a strand-displacing polymerase.


In some embodiments of the methods, the reporters are complexed with a polymer matrix. In some embodiments, the polymer matrix is formed from copolymerization of a plurality of monomers with the reporters. In some embodiments, the polymer matrix comprises a hydrogel. In some embodiments, (i) the step of cleaving the reporters releases detectable moieties from the polymer matrix, and (ii) the step of detecting comprises capturing and detecting the released detectable moieties at a capture region of a support medium.


In some embodiments, said same reaction volume comprises one or more additives comprising: (a) one or more of betaine monohydrate, acetamide, GABA, L-proline, beta-alanine, 6-aminohexanoic acid, urea, methylurea, ethylurea, hypotaurine, NDSB-256, and ammonium acetate; (b) one or more of trehalose, xylitol, D-sorbitol, sucrose, and trimethylamine N-oxide dihydrate; and/or (c) trimethylamine N-oxide dihydrate.


In some embodiments, described herein is a method of assaying for a target nucleic acid in a sample, the method comprising the following steps in a single reaction volume: (a) forming a complex comprising the target nucleic acid, a first programmable nuclease, and a first non-naturally occurring guide nucleic acid that hybridizes to a segment of the target nucleic acid, thereby activating the programmable nuclease; (b) cleaving a hairpin polynucleotide of a plurality of hairpin polynucleotides with the activated programmable nuclease, wherein each hairpin polynucleotide comprises (i) one or more RNA loops that are cleaved, (i) a first portion comprising DNA, and (iii) a second portion joined to the first portion by one of the one or more RNA loops, wherein the second portion is hybridized to a segment of the first portion; (c) forming a second complex comprising the first portion of the cleaved hairpin polynucleotide, a second programmable nuclease, and a second non-naturally occurring guide nucleic acid that hybridizes to the first portion of the cleaved hairpin, thereby activating the second programmable nuclease; (d) cleaving reporters with the activated first or second programmable nuclease; and (e) detecting a change in a signal, wherein the change in the signal is produced by cleavage of the reporters. In some embodiments, the first programmable nuclease and the second programmable nuclease are the same, such as any of the programmable nucleases disclosed herein. In some embodiments, the one or more RNA loops comprise a first RNA loop joining the first portion and the second portion, and a second RNA loop joining the first portion and a third portion; wherein the third portion hybridizes to a different segment of the first portion. In some embodiments, the second portion comprises RNA, DNA, or both.


Additional aspects and advantages of the present disclosure will become readily apparent to those skilled in this art from the following detailed description, wherein only illustrative embodiments of the present disclosure are shown and described. As will be realized, the present disclosure is capable of other and different embodiments, and its several details are capable of modifications in various obvious respects, all without departing from the disclosure. Accordingly, the drawings and description are to be regarded as illustrative in nature, and not as restrictive.


INCORPORATION BY REFERENCE

All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. To the extent publications and patents or patent applications incorporated by reference contradict the disclosure contained in the specification, the specification is intended to supersede and/or take precedence over any such contradictory material.





BRIEF DESCRIPTION OF THE DRAWINGS

The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.


The novel features of the disclosure are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings (also “figure” and “FIG.” herein), of which:



FIG. 1 shows the mean time to result for RT-LAMP reactions performed in a variety of buffers.



FIG. 2 provides summary effect sizes for different buffer constituents on the mean time to result for RT-LAMP reactions. The values on the right of the table provides the statistical significance for each variable or set of variables on the time to result relative to a commercial buffer control.



FIG. 3 provides a graphical representation of measured effect sizes for various buffer components on RT-LAMP reaction times. The final column shows the weighting function used to generate the bottom (‘Desirability’) row of the chart, which shows whether a variable correlates positively or negatively with RT-LAMP reaction time.



FIG. 4 shows the mean reaction times for DETECTR reactions performed with the buffers listed in TABLE 2, SEQ ID NO: 28 targeting SARS-COV-2 N-gene, and two separate dilutions of SARS-COV N-gene LAMP amplicons (‘Dil 1’ & ‘Dil 2’).



FIG. 5 provides effect summary sizes for the buffer constituents in the DETECTR assay summarized in FIG. 4.



FIG. 6 provides a graphical representations of measured effect sizes for various buffer components on the DETECTR reaction times shown in FIG. 4.



FIGS. 7A-7C graphically depicts RT-LAMP, DETECTR reaction times, and total reaction times (RT-LAMP+DETECTR reaction times) performed in a variety of buffers.



FIG. 8 shows the mean reaction times for a variety of RT-LAMP assays performed in different buffers over seven (Dil-1 through Dil-7) replicates. NTC denotes controls lacking the target nucleic acid.



FIG. 9 displays combined RT-LAMP and DETECTR assay times for 4 different buffer formulations.



FIG. 10 provides the rates of RT-LAMP reactions performed in 10 different buffers.



FIG. 11 provides combined RT-LAMP and DETECTR assay times for 4 different buffer systems and 3 different initial target nucleic acid copy numbers.



FIGS. 12A-12B panel A displays reaction times for 4 different buffer systems and 3 different initial copy numbers for the target nucleic acid. Panel A provides reaction times for RT-LAMP reactions, while panel B provides reaction times for DETECTR assays.



FIGS. 13A-13B shows the effect size and statistical significance of different buffer parameters on the rates of RT-LAMP reactions. The far right column shows the weighting function used to generate the bottom (‘Desirability’) row of the chart, which depicts whether a variable correlates positively or negatively with RT-LAMP reaction time.



FIGS. 14A-14B shows the effect size and statistical significance of buffer parameters on the rates of DETECTR reactions. The far right column shows the weighting function used to generate the bottom (‘Desirability’) row of the chart, which depicts whether a variable correlates positively or negatively with RT-LAMP reaction time.



FIG. 15A presents results for RT-LAMP amplification with Cas14a DETECTR in single reaction volume (one-pot).



FIG. 15B shows a DETECTR assay screening buffers and alternative polymerases that enable RT-LAMP to work at lower temperatures (normally at 65° C.).



FIG. 16 shows the layout of the LAMP plate used in an RT-LAMP assay screening buffers and alternative polymerases that enable RT-LAMP to work at lower temperatures (normally at 65 C).



FIGS. 17A-17B show assays evaluating the performance of Cas variants on a putative target site in SARS-COV-2.



FIG. 18 shows a strategy to simply DETECTR workflow for CRISPR diagnostics into a one-pot assay.



FIGS. 19A-19B present results for HotPot involving LAMP amplification with Cas14a DETECTR in single reaction volume (one-pot).



FIG. 20 presents results for identifying buffers that are compatible with Cas14a and low temperature RT-LAMP (LowLAMP).



FIG. 21 presents results involving the impact of individual components on the performance of Cas14 at low temperature RT-LAMP conditions.



FIG. 22 presents results for LAMP amplification with Cas14a DETECTR in single reaction volume (one-pot).



FIGS. 23A-23B presents results for one-pot Cas14 with LowLAMP at 50° C.



FIG. 24 presents results for one-pot Cas14 with Bsm DNA polymerase at 55° C.



FIG. 25 presents results for a limit of detection study involving one-pot DETECTR (HotPot).



FIG. 26 presents results for a limit of detection study involving one-pot DETECTR (HotPot), where two different DNA polymerases at 55 C were tested.



FIG. 27 shows the results of using thermostable tracrRNA in the HotPot assays.



FIG. 28 shows the results of using sgRNA in the HotPot assays.



FIG. 29 presents results for a study involving replacing Bst polymerase in the NEAR assay, showing enablement for SARS-COV-2 detection at lower temperatures. FIG. 30 presents results for NEAR assay amplification functions in Cas14a optimal buffers.



FIG. 31 presents results for Cas14a functions in a range of KOAc salt concentrations.



FIG. 32 presents results for a study involving increasing concentrations of KOAc to improve NEAR performance in Cas14a optimal buffers.



FIG. 33 presents results for a study involving increasing concentrations of KOAc to improve NEAR performance in Cas14a optimal buffers.



FIGS. 34A-34B present sequences and results for performance of Cas14a. 1 crRNAs on SARS-COV-2 E-gene amplicon, respectively. FIG. 34A discloses SEQ ID NO: 431.



FIG. 35 presents results for the evaluation of the performance of Klenow (exo-) NEAR assay in IB13 buffer at decreasing salt concentrations.



FIG. 36 presents an overview of rolling circle amplification (sRCA)



FIG. 37 presents results from screening dumbbell DNA templates for sRCA.



FIG. 38 presents results from a study involving the ability of Cas14a to detect product of RCA reaction across increasing temperatures.



FIG. 39 presents results from a study involving the effects of trigger oligos.



FIG. 40 presents results from a study involving a titration of trigger oligos for Cas14 one-pot sRCA.



FIG. 41 presents results from evaluating of a Cas12 variant (SEQ ID NO: 28) in one-pot sRCA.



FIG. 42 presents an overview of RCA positive feedback for Cas13.



FIG. 43 presents results from evaluating Cas13-compatible DNA templates for RCA.



FIG. 44 presents results from a study evaluating whether a Cas13-compatible DNA template is functional in RCA.



FIG. 45 presents results from a study involving Cas13 functionality in a one-pot sRCA reaction across increasing temperatures.



FIG. 46 presents an overview of CasPin.



FIG. 47 presents potential hairpin structures for CasPin.



FIG. 48 presents results for an initial design using two hairpins. FIG. 48 discloses SEQ ID NO: 432.



FIG. 49 presents a schematic of combined gRNA and reporter immobilization on the left and results for immobilization of DETECTR components using NHS-Amine chemistries on the right.



FIG. 50 presents results from optimizing the conjugation buffer to reduce non-specific binding.



FIG. 51 presents results from a study involving immobilizing different combinations of reporter+guide+a Cas12 variant (SEQ ID NO: 28).



FIG. 52 presents results from a study optimizing gRNA and target concentrations to improve signal-to-noise ratio for immobilized DETECTR.



FIGS. 53A-53B present modifications and results from evaluating various amino modifications for DETECTR immobilization, respectively.



FIG. 54 presents results for the FASTR assay, involving detection of SARS-COV-2 with rapid thermocycling+CRISPR Dx.



FIG. 55 presents results from a study to determine top performing polymerases and buffers for the FASTR assay.



FIG. 56 presents results for single copy detection of SARS-COV-2 with FASTR.



FIG. 57 presents results for variations on rapid cycling times for denaturation and annealing/extension in FASTR.



FIG. 58 presents results for minimizing RT time for FASTR.



FIG. 59 presents results for higher pH buffers that improve FASTR performance.



FIG. 60 presents results for FASTR compatibility with crude lysis buffers.



FIG. 61 presents results for non-optimized multiplexing of FASTR.



FIG. 62 presents results for multiplex FASTR.



FIG. 63 presents results for the limit of detection of multiplex FASTR.



FIG. 64 presents key primers and gRNAs. FIG. 64 disclose SEQ ID NOS: 433-434, 318, 435-436, 326 and 437-439, respectively, in order of appearance.



FIG. 65 presents results depicting the enhancement of the signal generated in a Hotpot reaction with the addition of Thermostable Inorganic Pyrophosphatase (TIPP) in comparison to control conditions lacking TIPP, target RNA or both.



FIG. 66A and FIG. 66B present results from DETECTR lateral flow Hotpot reaction assay strips, depicting the enhancement of the signal generated in a Hotpot reaction with the addition of Thermostable Inorganic Pyrophosphatase (TIPP) in comparison to control conditions lacking TIPP, target RNA or both.



FIG. 67 shows an exemplary workflow for DETECTR-based HotPot reactions.



FIG. 68 shows fluorescence results of HotPot reactions with reporters immobilized on glass beads.



FIG. 69 shows lateral flow strip results using samples from the same experiments conducted to yield results illustrated in FIG. 68.



FIG. 70 shows fluorescence results of HotPot reactions with reporters immobilized on magnetic beads.



FIG. 71 shows fluorescence results of DETECTR-based HotPot assays for a variety of respiratory disease nucleic acid sequence targets.



FIGS. 72A-72C shows results of limit of detection experiments for initial HotPot assay testing.



FIG. 73 shows the fluorescence detected from HotPot assays in the presence of various additives.



FIGS. 74A-74B shows the influence of select additives that increase the speed and/or the signal strength of some HotPot assays.



FIG. 75 shows HotPot results from experiments conducted with various amounts of a few additives.



FIG. 76 shows HotPot results from experiments conducted with various amounts of a few additives with BSM DNA Polymerases.



FIGS. 77A-77B show lateral flow assay results of DETECTR-based OnePot and HotPot assays conducted with hydrogels comprising immobilized reporters.



FIG. 78 shows an exemplary hydrogel comprising immobilized reporters co-polymerized therein.



FIGS. 79A and 79B show exemplary multiplexing strategies for hydrogel immobilized DETECTR systems.



FIG. 80 shows a schematic of the NEAR reaction. A forward and reverse primer consisting of a nicking enzyme stabilization site and recognition region able amplify a target region of interest into a single-stranded DNA molecule. A guide RNA will bind a region complementary to the amplified ssDNA, allowing further detection by a DETECTR system.



FIGS. 81A-81B show an exemplary NEAR-DETECTR reaction. FIG. 81A show a forward and reverse primer flanking the target region. In addition, a panel of 19 guide RNAs are shown in comparison to the amplicon. FIG. 81A disclose SEQ ID NOS 395, 440 and 396, respectively, in order of appearance. FIG. 81B shows the detection of the amplicon using NEAR-DETECTR using the above mentioned guide RNAs.



FIGS. 82A-82B shows an example NEAR reaction with an exemplary guide RNA showing detection of the E-gene of SARS-COV2. FIG. 82A shows an example NEAR reaction with forward and reverse NEAR primers with a guide RNA. FIG. 82A discloses SEQ ID NO: 441. FIG. 82B shows the NEAR-DETECTR reaction following amplification, showing the resulting signal of 20,000 copies or 0 copies of the amplicon in solution.



FIG. 83 shows the resulting signal of a NEAR-DETECTR reaction in which the pre-amplification time is varied prior to DETECTR.



FIG. 84 shows the comparison of the NEAR-DETECTR reaction using orthogonal Cas systems (Cas12 Variant (SEQ ID NO: 28), Cas13 Variant (SEQ ID NO: 154), and Cas14 Variant (SEQ ID NO. 63)). The experiments were performed in the presence or absence of the target NEAR amplicon, showing different cleavage preferences for the reporter molecule.



FIG. 85 shows the optimization of the NEAR reaction using different magnesium (Mg2+) concentrations. This shows the time to result (in minutes) comparing different buffer compositions and different concentrations of added magnesium. The top panel shows the following conditions using Bst2.0, and the bottom panel shows the following experimental conditions using Bst.3.0. These results informed the following experimental conditions for the NEAR reaction: Bst2.0, 12 mM Mg2+, at 60 C, resulting in a less than 5 minute amplification time and approximately 20,000 copies.



FIGS. 86A-86B shows the experimental design of the primer and guide RNA design and the results of these designs. In FIG. 86A, eight primer pairs (R1763 F(1-8)/R(1-8)) and the guide RNA R1763. FIG. 86A discloses SEQ ID NO: 427. In FIG. 86B, the aforementioned primer pairs and guides R1763 and R1765 are used in a NEAR-DETECTR reaction to determine the efficacy of the reaction using different primer pairs. The raw fluorescence (AU) of the NEAR-DETECTR reaction is reported.



FIGS. 87A-87B describes the optimization of the hinge stabilization region in the NEAR primers to determine if the nicking enzyme activity can be modulated. In FIG. 87A, the hinge stabilization loop region is modified to alter the melting temperature of the stem loop. In FIG. 87B, the modified stem loops as shown in FIG. 87A were used for detection of the SARS-CoV-2 E-gene using different inputs of amplicons resulting from a NEAR amplification (1,000, 500, and 200 input amplicon copies).



FIG. 88 shows a comparison of a reverse transcription-NEAR-DETECTR (RT-NECTR) reaction using different reverse transcriptases. Wartmstart RTx (NEB), Bst 3.0, and Omniscript RT (Qiagen) were used on different amounts of input RNA in order to compare the limit of detection (LOD) of these reaction conditions.



FIG. 89 shows the experimental results of an RT-NECTR reaction using differing concentrations of NEAR primers.



FIG. 90 shows the LOD of the E-gene of SARS-COV-2 using RT-NECTR using different amplification times and input copies for the RT-NECTR reaction.



FIG. 91 shows detection of the SARS-COV-2 E gene using different Cas systems (Cas12 Variant (SEQ ID NO: 28), Cas13 Variant (SEQ ID NO: 154), and Cas14 Variant (SEQ ID NO. 63)) and a panel of different guides.





DETAILED DESCRIPTION

While various embodiments of the invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions may occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed.


Unless otherwise defined, all technical terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. As used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural references unless the context clearly dictates otherwise. Any reference to “or” herein is intended to encompass “and/or” unless otherwise stated.


Whenever the term “at least,” “greater than,” or “greater than or equal to” precedes the first numerical value in a series of two or more numerical values, the term “at least,” “greater than” or “greater than or equal to” applies to each of the numerical values in that series of numerical values. For example, greater than or equal to 1, 2, or 3 is equivalent to greater than or equal to 1, greater than or equal to 2, or greater than or equal to 3.


Whenever the term “no more than,” “less than,” “less than or equal to,” or “at most” precedes the first numerical value in a series of two or more numerical values, the term “no more than,” “less than,” “less than or equal to,” or “at most” applies to each of the numerical values in that series of numerical values. For example, less than or equal to 3, 2, or 1 is equivalent to less than or equal to 3, less than or equal to 2, or less than or equal to 1.


Where values are described as ranges, it will be understood that such disclosure includes the disclosure of all possible sub-ranges within such ranges, as well as specific numerical values that fall within such ranges irrespective of whether a specific numerical value or specific sub-range is expressly stated.


The present disclosure provides various compositions, methods, and devices of use thereof for assaying and detecting a nucleic acid. In many cases, nucleic acid detection comprises multiple steps (e.g., multiple reactions). For example, nucleic acid detection often comprises amplification of a target nucleic acid followed by detection. Augmenting the challenge of nucleic acid detection, multiple steps often require different conditions, which can necessitate buffer exchanges or sample transfers that increase the potential for contamination and sample loss. Furthermore, many single-buffer systems only support a single type of reaction, and drastically retard the rates of other reactions performed within them.


To address these challenges, aspects of the present disclosure provide buffers, systems, and compositions that can support multiple nucleic acid detection steps (e.g., multiple types of reactions) with optimal or close to optimal rates. Such buffers, systems, and compositions can enable nucleic acid detection without buffer exchanges or sample transfers, thereby diminishing total assay times, decreasing sample loss and contamination, and minimizing user input.


For example, particular aspects of the present disclosure provide containers comprising all of the reactants necessary for detection of a target nucleic acid from a sample. In such cases, no further user steps may be required once the sample is inserted into the container (e.g., through a pierceable film, membrane, or septum) and the container is placed within an instrument (e.g., a fluorimeter comprising a sample heater). In some aspects, a plurality of containers may be provided in a kit or on a single support (e.g., each container is a well on a multi-well plate). Two containers among the plurality of containers may comprise reagents targeting different nucleic acid sequences for detection to enable multiplexing.


The target nucleic acid can be a nucleic acid or a portion of a nucleic acid from a pathogen, virus, bacterium, fungi, protozoa, worm, or other agent(s) or organism(s) responsible for and/or related to a disease or condition in living organisms (e.g., humans, animals, plants, crops, and the like). The target nucleic acid can be a nucleic acid, or a portion thereof. The target nucleic acid can be a portion of a nucleic acid from a gene expressed in a cancer or genetic disorder in the sample. The target nucleic acid can be a portion of an RNA or DNA from any organism in the sample. In some embodiments, one or more programmable nucleases as disclosed herein can be activated to initiate trans cleavage activity of a reporter (also referred to herein as a detector nucleic acid). In general, the term “reporter” as used in this context refers to a reagent comprising a polynucleotide, wherein cleavage of the polynucleotide results in a change in a signal. For example, the reporter may comprise a fluorescent label joined to a quencher by a short polynucleotide sequence. Little to no fluorescence is detectable from the fluorescent label when joined to the quencher. However, upon cleavage of the polynucleotide, the fluorescent label is separated from the quencher, resulting in a significant and detectable increase in fluorescent signal upon excitation of the label. Alternative labels and arrangements for producing a change in signal upon cleavage of the polynucleotide portion of the reporter are possible, and illustrative examples are described herein. The polynucleotide of the reporter can comprise DNA, RNA, modified nucleotides, or a combination of two or more of these. A programmable nuclease as disclosed herein can, in some cases, bind to a target sequence or target nucleic acid to initiate trans cleavage of a reporter. The programmable nuclease can be referred to as an RNA-activated programmable RNA nuclease. In some instances, the programmable nuclease as disclosed herein can bind to a target DNA to initiate trans cleavage of an RNA reporter. Such a programmable nuclease can be referred to herein as a DNA-activated programmable RNA nuclease. In some cases, a programmable nuclease as described herein can be activated by a target RNA or a target DNA. For example, a programmable nuclease, e.g., a Cas enzyme, can be activated by a target RNA nucleic acid or a target DNA nucleic acid to cleave RNA reporters. In some embodiments, the Cas enzyme can bind to a target ssDNA which initiates trans cleavage of RNA reporters. In some instances, a programmable nuclease as disclosed herein can bind to a target DNA to initiate trans cleavage of a DNA reporter, and this programmable nuclease can be referred to as a DNA-activated programmable DNA nuclease.


The systems and methods of the present disclosure can be implemented using a device that is compatible with any type of programmable nuclease that is human-engineered or naturally occurring. The programmable nuclease can comprise a nuclease that is capable of being activated when complexed with a guide nucleic acid and a target nucleic acid segment or a portion thereof. A programmable nuclease can become activated when complexed with a guide nucleic acid and a target sequence of a target gene of interest. The programmable nuclease can be activated upon binding of a guide nucleic acid to a target nucleic acid and can exhibit or enable trans cleavage activity once activated. In any instances or embodiments where a CRISPR-based programmable nuclease is described or used, it is recognized herein that any other type of programmable nuclease can be used in addition to or in substitution of such a CRISPR-based programmable nuclease.


Various methods, reagents, compositions, systems, and devices disclosed herein use a programmable nuclease complexed with guide nucleic acid sequence to detect the presence or absence of, and/or quantify the amount of, a target nucleic acid sequence. Binding of a guide nucleic acid with a target nucleic acid may activate a programmable nuclease to cleave single stranded nucleotides in a sequence non-specific manner, hereinafter referred to as “transcollateral” cleavage or “trans cleavage”. Some assays of the present disclosure detect single-stranded, non-target nucleic acid cleavage to determine the presence and/or quantity of a target nucleic acid. Assays which leverage the transcollateral cleavage properties of programmable nuclease enzymes (e.g., CRISPR-Cas enzymes) are often referred to herein as DNA endonuclease targeted CRISPR trans reporter (DETECTR) reactions. Herein, detection of reporter cleavage (directly or indirectly) to determine the presence of a target nucleic acid sequence may be referred to as ‘DETECTR’. In some embodiments, described herein is a method of assaying for a target nucleic acid in a sample comprising contacting the target nucleic acid with a programmable nuclease, a non-naturally occurring guide nucleic acid that hybridizes to a segment of the target nucleic acid, and a reporter, and assaying for a change in a signal, wherein the change in the signal is produced by or indicative of cleavage of the reporter.


Assays disclosed herein may comprise amplification (e.g., loop-mediated amplification (LAMP) or recombinase polymerase amplification (RPA), rolling circle amplification (RCA), nicking enzyme amplification reaction (NEAR), etc.) of a target nucleic acid sequence (e.g., a viral nucleic acid extracted from a patient). A target nucleic acid may also be reverse transcribed. An assay targeting an RNA sequence may utilize reverse transcription (RT) and amplification to generate amplicons for a programmable nuclease-based detection (e.g., DETECTR) reaction. A target nucleic acid may be amplified by thermal amplification (e.g., PCR) or isothermal amplification (e.g., LAMP, RCA, NEAR). An assay may utilize an amplification reaction and a programmable nuclease-based detection (e.g., DETECTR) reaction.


Among the various aspects of the present disclosure are compositions and methods for rapid and accurate detection of a target nucleic acid. In many cases, target nucleic acids are present in low copy numbers or as small proportions of the total nucleic acid content from a sample. This is a particular challenge in pathogen (e.g., viral) diagnostics, as nucleic acid biomarkers are often present in copy numbers below 1000 in samples derived from patients. Owing to this challenge, nucleic acid detection typically requires multi-step and multi-reaction assays. However, the assays themselves often have strict and non-overlapping requirements for the physical and chemical conditions in which they can be performed. For example, conditions that support rapid amplification reactions are often unsuitable for programmable nuclease-mediated reactions (e.g., CRISPR-Cas enzyme-mediated reactions), such as DETECTR reactions. Thus, nucleic acid detection methods frequently require sample transfers and buffer exchanges, resulting in long assay times leading to low yields.


In various aspects, the present disclosure provides a range of compositions that facilitate rapid, single buffer nucleic acid detection assays. Among such compositions, the present disclosure provides buffers capable of supporting rapid amplification and programmable nuclease (e.g., CRISPR-Cas) enzyme mediated reactions (e.g., DETECTR reactions). In such cases, the buffer may not only enable fast reaction rates, but also high reaction yields. Furthermore, multi-reaction buffer compatibility can drastically diminish an assay's required user input. Further aspects of the disclosure build on these favorable characteristics, and provide methods for performing single buffer nucleic acid detection assays, as well as kits for performing such assays.


Assaying Methods

In some embodiments, a programmable nuclease can be used for detection of a target nucleic acid in a sample from a subject. The programmable nuclease may be provided in a buffer which enables fast (e.g., within 25%, 50%, or 75% of the fastest reported reaction rate for the programmable nuclease at a particular temperature) kinetics for the programmable nuclease and for reagents for other reactions (e.g., amplification, viral or cellular lysis, nucleic acid digestion, etc.). This can be particularly advantageous for low stability targets, such as RNA, which can be prone to rapid degradation, precipitation, denaturation, or side reactions. Whereas detecting a nucleic acid from a low titer sample (e.g., a sample comprising 1000, 500, 100, 50, 25, or 10 target nucleic acid molecules) may be unfeasible if a sample needs to be transferred between multiple reaction volumes, a single-buffer reaction system can enable detection of sparse nucleic acid targets by minimizing sample loss between steps.


The target nucleic acid may comprise a sequence associated with a pathogen (e.g., a virus), a human gene of interest (e.g., an oncogene such as BRCA1), a nucleic acid sequence for uniquely identifying an individual, or a fungal or bacterial nucleic acid sequence (e.g., for assessing skin health or a gut microbiome). For example, a programmable nuclease can be complexed with a guide nucleic acid that hybridizes to a target sequence of a target nucleic acid from coronavirus. The complex can be contacted to a sample from a subject. The target nucleic acid may or may not be present in the sample. If present, the target nucleic acid in the sample can optionally be reverse transcribed (RT). The target nucleic acid can be amplified by thermal amplification (e.g., PCR, FASTR) or isothermal amplification (e.g., LAMP, RPA, RT-RPA, or RT-LAMP). In some embodiments, reverse transcription and isothermal amplification may be performed simultaneously.


Upon activation, the programmable nuclease can cleave a reporter, which may comprise a detectable label attached to a polynucleotide (e.g., polydeoxyribonucleotide or polyribonucleotide). In some embodiments of the assay, upon cleavage of the polynucleotide, the detectable label emits a detectable signal, which is then detected and quantified (e.g., the detectable label may be a fluorophore and the detectable signal may be fluorescence). Upon detection of the detectable signal, it can be determined that the sample from the subject contained a target nucleic acid. In some embodiments, a programmable nuclease-based detection assay may detect multiple target nucleic acids or amplicons. For example, a programmable nuclease-based detection assay may target multiple non-overlapping or partially overlapping portions of a sequence of interest, while an amplification reaction may tile primers over a region of a target nucleic acid. Additionally, a programmable nuclease-based detection assay and/or amplification reaction may target multiple distinct sequences, such as sequences from separate portions of a genome or from separate genomes.


The compositions and methods of use thereof disclosed herein include using a programmable nuclease such as a Cas12 protein, a CasΦ protein, Cas14 protein, or a Cas13 protein to assay for, detect, and/or quantify a target nucleic acid. In some embodiments, a Cas12 protein, Cas13 protein, Cas14 protein, or a CasΦ protein is used for detection of a target nucleic acid in a sample from a subject. For use in an assay with a Cas12 protein, CasΦ protein, Cas14 protein, or a Cas13 protein, a target nucleic acid in a sample can be reverse transcribed and amplified by thermal (e.g., PCR, FASTR) or isothermal amplification (e.g., LAMP, RCA, NEAR). For use in an assay with a Cas13 protein, the amplified target nucleic acids can be transcribed back into RNA. If the subject is infected with coronavirus, the guide nucleic acid hybridizes to the target nucleic acid or amplicon thereof leading to activation of the Cas12 protein, Cas14 protein, CasΦ protein, or Cas13 protein. Upon activation, the Cas12 protein, the CasΦ protein, the Cas14 protein, or the Cas13 protein can cleave a reporter, wherein the reporter comprises a detectable label attached to the nucleic acid for cleavage by a Cas12 protein, the Cas14 protein a Cas13 protein, or a CasΦ protein. In some embodiments of the assay, upon cleavage of the reporter, the detectable label emits a detectable signal, which can then be captured and quantified (e.g., the detectable label may be a fluorophore and the detectable signal may be fluorescence). Upon detection of a detectable label, it can be determined that the sample comprised the target nucleic acid or target nucleic acids.


In some embodiments, a programmable nuclease having at least 60% sequence identity to SEQ ID NO: 18-170, 221-268, or 397-423 can be used for detection of a target nucleic acid (e.g., from a coronavirus such as SARS-COV-2) in a sample from a subject. For example, a programmable nuclease having at least 60% sequence identity to SEQ ID NO: 18 can be complexed with a guide nucleic acid that hybridizes to a target sequence of a target nucleic acid from coronavirus. For example, a programmable nuclease having at least 60% sequence identity to any one of SEQ ID NOs: 397, 398, 399, 400, 401, 402, 403, 404, 405, 406, 407, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, or 423 can be complexed with a guide nucleic acid that hybridizes to a target sequence of a target nucleic acid from coronavirus. For example, a programmable nuclease having at least 60% sequence identity to SEQ ID NO: 406 can be complexed with a guide nucleic acid that hybridizes to a target sequence of a target nucleic acid from coronavirus.


Also described herein are methods, reagents, and devices for detecting the presence of a target nucleic acid in a sample. The methods, reagents, and devices for detecting the presence of a target nucleic acid in a sample can be used in rapid lab tests for detection of a target nucleic acid of interest (e.g., target nucleic acids from a target population). In particular, provided herein are methods, reagents, and devices wherein the rapid lab tests can be performed in a single system. The target nucleic acid may be a portion of a nucleic acid from a virus (e.g., coronavirus) or other agents responsible for a disease in the sample.


In some embodiments, programmable nucleases disclosed herein are activated by RNA or DNA to initiate trans cleavage activity of a reporter. A programmable nuclease as disclosed herein, in some cases, binds to a target RNA to initiate trans cleavage of a reporter, and this programmable nuclease can be referred to as an RNA-activated programmable RNA nuclease. In some instances, a programmable nuclease as disclosed herein binds to a target DNA to initiate trans cleavage of a reporter, and this programmable nuclease can be referred to as a DNA-activated programmable RNA nuclease. In some cases, a programmable nuclease as described herein is capable of being activated by a target RNA or a target DNA. For example, a Cas13 protein, such as a Cas13a, disclosed herein is activated by a target RNA nucleic acid or a target DNA nucleic acid to transcollaterally cleave an RNA reporter. In some embodiments, the Cas13 binds to a target ssDNA which initiates trans cleavage of an RNA reporter.


The detection of the target nucleic acid in the sample may indicate the presence of the disease in the sample and may provide information for taking action to reduce the transmission of the disease to individuals in the disease-affected environment or near the disease-carrying individual. The detection of the target nucleic acid in the sample may indicate the presence of a disease mutation, such as a single nucleotide polymorphism (SNP) that provide antibiotic resistance to a disease-causing bacteria. The detection of the target nucleic acid is facilitated by a programmable nuclease. The programmable nuclease can become activated after binding of a guide nucleic acid with a target nucleic, in which the activated programmable nuclease can cleave the target nucleic acid and can have trans cleavage activity, which can also be referred to as “collateral” or “transcollateral” cleavage.


Trans cleavage activity can be non-specific cleavage of nearby single-stranded nucleic acids by the activated programmable nuclease, such as trans cleavage of reporters with a detection moiety. Once the reporter is cleaved by the activated programmable nuclease, the detection moiety is released from the reporter and generates a detectable signal. Often the detection moiety is at least one of a fluorophore, a dye, a polypeptide, or a nucleic acid. Sometimes the detection moiety binds to a capture molecule on the support medium to be immobilized. The detectable signal can be visualized on the support medium to assess the presence or level of the target nucleic acid associated with an ailment, such as a disease. The programmable nuclease can be a CRISPR-Cas (clustered regularly interspaced short palindromic repeats—CRISPR associated) nucleoprotein complex with trans cleavage activity, which can be activated by binding of a guide nucleic acid with a target nucleic acid. These assays, which leverage the transcollateral cleavage properties of programmable nucleases (e.g., CRISPR-Cas enzymes) are often referred to herein as DNA endonuclease targeted CRISPR trans reporter (DETECTR) reactions. A programmable nuclease-based detection (e.g., DETECTR) reaction can be performed in a fluidic device.


In some embodiments, the present disclosure provides for Cas12 detection of a target nucleic acid from a coronavirus. In this case, nucleic acids (e.g., RNA) from a sample may be optionally reverse transcribed and/or amplified into DNA. Any Cas12 protein disclosed herein may be complexed with a guide nucleic acid designed to hybridize to a nucleic acid sequence of the (optionally reverse transcribed and/or amplified) DNA. DETECTR reactions can then be carried out. In the presence of reverse transcribed and amplified DNA indicative of coronavirus, Cas12 is activated to transcollaterally cleave a reporter, emitting a detectable signal (e.g., fluorescence). In some embodiments, the present disclosure provides for Cas13 detection of a target nucleic acid from a coronavirus. In this case, RNA in a sample may be either directly detected by complexing a Cas13 enzyme with a guide nucleic acid designed to hybridize to a target RNA sequence from a coronavirus or, RNA may be reverse transcribed, amplified, and in vitro transcribed prior to contacting it with a Cas13 enzyme complexed with a guide nucleic acid designed to hybridize this amplified target RNA sequence from a coronavirus. In the presence of the RNA (unamplified or amplified), Cas13 may be activated to transcollaterally cleave a reporter, thereby emitting a detectable signal (e.g., fluorescence). In some embodiments, the present disclosure provides for Cas14 detection of a target nucleic acid from a coronavirus. In this case, nucleic acids (e.g., RNA) from a sample may be optionally reverse transcribed and/or amplified into DNA. Any Cas14 protein disclosed herein may be complexed with a guide nucleic acid designed to hybridize to a nucleic acid sequence of the (optionally reverse transcribed and/or amplified) DNA. DETECTR reactions can then be carried out. In the presence of reverse transcribed and amplified DNA indicative of coronavirus, Cas14 is activated to transcollaterally cleave a reporter, emitting a detectable signal (e.g., fluorescence). In some embodiments, the present disclosure provides for CasPhi detection of a target nucleic acid from a coronavirus. In this case, nucleic acids (e.g., RNA) from a sample may be optionally reverse transcribed and/or amplified into DNA. Any CasPhi protein disclosed herein may be complexed with a guide nucleic acid designed to hybridize to a nucleic acid sequence of the (optionally reverse transcribed and/or amplified) DNA. DETECTR reactions can then be carried out. In the presence of reverse transcribed and amplified DNA indicative of coronavirus, CasPhi is activated to transcollaterally cleave a reporter, emitting a detectable signal (e.g., fluorescence).


Also described herein is a kit for detecting a target nucleic acid (e.g., from a coronavirus such as SARS-COV-2). The kit may comprise a support medium; a guide nucleic acid sequences targeted to a target nucleic acid sequence; a programmable nuclease capable of being activated when complexed with a guide nucleic acid and a target nucleic acid; and a single-stranded reporter comprising a detection moiety, wherein the reporter is capable of being cleaved by the activated nuclease, thereby generating a first detectable signal. In some cases, the target nucleic acid for detecting a coronavirus may comprise a sequence with at least 60% sequence similarity to that of SEQ ID NOs: 179-184. In some cases, the guide nucleic acid for detection of a coronavirus may comprise a sequence with at least 60% sequence similarity to the sequences in SEQ ID NOs: 318-327.


A biological sample from an individual or an environmental sample can be tested for the presence of a particular nucleic acid sequence (e.g., whether a human sample comprises coronavirus). The detection of the target nucleic acid detected can also indicate that one or more of the target populations is wild-type or comprises a mutation, such as a mutation that confers resistance to treatment, such as antibiotic treatment. A sample from an individual or from an environment is applied to the reagents described herein. If the target nucleic acid is present in the sample, the target nucleic acid binds to the guide nucleic acid to activate the programmable nuclease. The activated programmable nuclease cleaves the reporter and generates a detectable signal that can be visualized, for example on a support medium. If the target nucleic acid is absent in the sample or below the threshold of detection, the guide nucleic acid remains unbound, the programmable nuclease remains inactivated, and the reporter remains uncleaved. Such methods, reagents, and devices described herein may allow for detection of target nucleic acid, and in turn the disease associated with the target nucleic acids (e.g., coronavirus such as SARS-CoV-2), in remote regions or low resource settings without specialized equipment. Also, such methods, reagents, and devices described herein may allow for detection of target nucleic acid, and in turn the disease associated with the target nucleic acids, in healthcare clinics or doctor offices without specialized equipment. In some cases, this provides a point of care testing for users to quickly and easily test for a disease or infection with high sensitivity at home or in an office of a healthcare provider. Assays that deliver results in under an hour, for example, in 15 to 60 minutes, are particularly desirable for at home testing for many reasons. For example, antivirals can be most effective when administered within the first 48 hours after disease exposure. Thus, the methods disclosed herein, which are capable of delivering results in under an hour, may allow for the delivery of anti-viral therapy during the first 48 hours after infection. Additionally, the systems and assays provided herein, which are capable of delivering quick diagnoses and results, can help keep or send a patient at home, improve comprehensive disease surveillance, and prevent the spread of an infection. In other cases, this provides a test, which can be used in a lab to detect one or more nucleic acid populations or varieties of interest in a sample from a subject. In particular, provided herein are methods, reagents, and devices, wherein the high sensitivity lab tests can be performed in a single assay. In some cases, this may be valuable in detecting diseases in a developing country and as a global healthcare tool to detect the spread of a disease or efficacy of a treatment or provide early detection of a disease.


Some methods as described herein use an editing technique, such as a technique using an editing enzyme or a programmable nuclease and guide nucleic acid, to detect a target nucleic acid. An editing enzyme or a programmable nuclease in the editing technique can be activated by one or more target nucleic acids, after which the activated editing enzyme or activated programmable nuclease can cleave nearby single-stranded nucleic acids, such reporters with a detection moiety. A target nucleic acid can be amplified by isothermal amplification or thermocycling amplification and then an editing technique can be used to detect the marker. In some instances, the editing technique can comprise an editing enzyme or programmable nuclease that, when activated, cleaves nearby RNA or DNA as the readout of the detection. The methods as described herein in some instances comprise obtaining a cell-free DNA sample, amplifying DNA from the sample, using an editing technique to cleave reporters, and reading the output of the editing technique. In other instances, the method comprises obtaining a fluid sample from a patient, and without amplifying a nucleic acid of the fluid sample, using an editing technique to cleave reporters, and detecting the nucleic acid. The method can also comprise using single-stranded detector DNA, cleaving the single-stranded detector DNA using an activated editing enzyme, wherein the editing enzyme cleaves at least 50% of a population of single-stranded detector DNA as measured by a change in color. A number of samples, guide nucleic acids, programmable nucleases or editing enzymes, support mediums, target nucleic acids, single-stranded reporters, and reagents are consistent with the devices, systems, fluidic devices, kits, and methods disclosed herein.


Also disclosed herein are reporters and methods detecting a target nucleic using the reporters. Often, the reporter is a protein-nucleic acid. For example, a method of assaying for a target nucleic acid in a sample comprises contacting the sample to a plurality of complexes comprising a guide nucleic acid, each guide nucleic acid sequence comprising a segment that is reverse complementary to a segment of a target nucleic acid sequence within a target nucleic acid population and programmable nucleases that exhibits sequence independent cleavage upon forming complexes comprising the segment of the guide nucleic acid binding to the segment of the target nucleic acid; and assaying for a signal indicating cleavage of at least some protein-nucleic acids of a population of protein-nucleic acids, wherein the signal indicates a presence of one or more of the target nucleic acid populations in the sample and wherein absence of the signal indicates an absence of the target nucleic acid population in the sample. Often, the protein-nucleic acid is an enzyme-nucleic acid or an enzyme substrate-nucleic acid. The nucleic acid can be DNA, RNA, or a DNA/RNA hybrid. The methods described herein use a programmable nuclease, such as the CRISPR/Cas system, to detect a target nucleic acid (e.g. from a coronavirus such as SARS-COV-2). A method of assaying for a target nucleic acid (e.g. from a coronavirus such as SARS-COV-2) in a sample, for example, comprises: a) contacting the sample to a plurality of complexes comprising a guide nucleic acid, each guide nucleic acid sequence comprising a segment that is reverse complementary to a segment of a nucleic acid target sequence within a target nucleic acid population, and programmable nucleases that exhibits sequence independent cleavage upon forming complexes comprising the segment of the guide nucleic acid binding to the segment of the target nucleic acid; b) contacting the complexes to a substrate; c) contacting the substrate to a reagent that differentially reacts with a cleaved substrate; and d) assaying for a signal indicating cleavage of the substrate, wherein the signal indicates a presence of one or more of the target nucleic acid populations in the sample and wherein absence of the signal indicates an absence of the target nucleic acid population in the sample. Often, the substrate is an enzyme-nucleic acid. Sometimes, the substrate is an enzyme substrate-nucleic acid.


Cleavage of the protein-nucleic acid produces a signal. For example, cleavage of the protein-nucleic acid produces a calorimetric signal, a potentiometric signal, an amperometric signal, an optical signal, or a piezo-electric signal. Various devices can be used to detect these different types of signals, which indicate whether a target nucleic acid is present in the sample.


The present disclosure provides buffers capable of supporting rapid amplification and programmable nuclease-based reactions (e.g., DETECTR).


In some assays, such a buffer comprising amplification and programmable nuclease-based detection reagents is contacted to a sample, thereby enabling simultaneous amplification and programmable nuclease-based detection reactions. For example, a buccal swab sample may be added to a buffer comprising amplification and programmable nuclease-based detection reagents targeting a particular oral bacterium gene, thereby enabling an assay for that particular oral bacterium. In some assays, a target nucleic acid is first contacted with amplification reagents, and later (e.g., after the amplification reaction has come to completion) contacted with reagents for a programmable nuclease-based detection reaction. In some assays, a target nucleic acid is first contacted with amplification reagents, and later sequentially contacted with different sets of programmable nuclease-based detection reagents targeting different nucleic acid sequences, wherein programmable nuclease-based detection reagent additions that lead to an increase in signal (e.g., fluorescence signal) may indicate the presence of a particular nucleic acid sequence.


Sample

A number of samples are consistent with the methods, reagents, and devices disclosed herein.


These samples can comprise a target nucleic acid for detection of an ailment, such as a disease, pathogen, or virus, such as influenza. Generally, a sample from an individual or an animal or an environmental sample can be obtained to test for presence of a disease, or any mutation of interest. A biological sample from the individual may be blood, serum, plasma, saliva, urine, mucosal sample, peritoneal sample, cerebrospinal fluid, gastric secretions, nasal secretions, sputum, pharyngeal exudates, urethral or vaginal secretions, an exudate, an effusion, or tissue. A tissue sample may be dissociated or liquified prior to application to the detection system of the present disclosure. A sample from an environment may be from soil, air, or water. In some instances, the environmental sample is taken as a swab from a surface of interest or taken directly from the surface of interest. In some instances, the raw sample is applied to the detection system. In some instances, the sample is diluted with a buffer or a fluid or concentrated prior to application to the detection system or be applied neat to the detection system. Sometimes, the sample is contained in no more 20 μL. The sample, in some cases, is contained in no more than 1, 5, 10, 15, 20, 25, 30, 35 40, 45, 50, 55, 60, 65, 70, 75, 80, 90, 100, 200, 300, 400, 500 L, or any of value from 1 μL to 500 μL. Sometimes, the sample is contained in more than 500 μL. The sample may be contained within a solid, membranous, or mesh material, such as a swab (e.g., a buccal or nasal swab).


In some instances, the sample is taken from single-cell eukaryotic organisms; a plant or a plant cell; an algal cell; a fungal cell; an animal cell, tissue, or organ; a cell, tissue, or organ from an invertebrate animal; a cell, tissue, fluid, or organ from a vertebrate animal such as fish, amphibian, reptile, bird, and mammal; a cell, tissue, fluid, or organ from a mammal such as a human, a non-human primate, an ungulate, a feline, a bovine, an ovine, and a caprine. In some instances, the sample is taken from nematodes, protozoans, helminths, or malarial parasites. In some cases, the sample comprises nucleic acids from a cell lysate from a eukaryotic cell, a mammalian cell, a human cell, a prokaryotic cell, or a plant cell. In some cases, the sample comprises nucleic acids expressed from a cell.


The sample used for disease testing may comprise at least one target sequence that can bind to a guide nucleic acid of the reagents described herein. A portion of a nucleic acid can be from a genomic locus, a transcribed mRNA, or a reverse transcribed cDNA. A portion of a nucleic acid can be from 5 to 100, 5 to 90, 5 to 80, 5 to 70, 5 to 60, 5 to 50, 5 to 40, 5 to 30, 5 to 25, 5 to 20, 5 to 15, or 5 to 10 nucleotides in length. A portion of a nucleic acid can be 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 45, 50, 60, 70, 80, 90, or 100 nucleotides in length. The target sequence can be reverse complementary to a guide nucleic acid. Each target sequence of the multiple target sequences can be reverse complementary to a distinct guide nucleic acid.


The systems and methods of the present disclosure can be used to detect one or more target sequences or nucleic acids in one or more samples. The one or more samples can comprise one or more target sequences or nucleic acids for detection of an ailment, such as a disease, cancer, or genetic disorder, or genetic information, such as for phenotyping, genotyping, or determining ancestry and are compatible with the reagents and support mediums as described herein. Generally, a sample can be taken from any place where a nucleic acid can be found. Samples can be taken from an individual/human, a non-human animal, or a crop, or an environmental sample can be obtained to test for presence of a disease, virus, pathogen, cancer, genetic disorder, or any mutation or pathogen of interest. A biological sample can be blood, serum, plasma, lung fluid, exhaled breath condensate, saliva, spit, urine, stool, feces, mucus, lymph fluid, peritoneal, cerebrospinal fluid, amniotic fluid, breast milk, gastric secretions, bodily discharges, secretions from ulcers, pus, nasal secretions, sputum, pharyngeal exudates, urethral secretions/mucus, vaginal secretions/mucus, anal secretion/mucus, semen, tears, an exudate, an effusion, tissue fluid, interstitial fluid (e.g., tumor interstitial fluid), cyst fluid, tissue, or, in some instances, any combination thereof. A sample can be an aspirate of a bodily fluid from an animal (e.g., human, animals, livestock, pet, etc.) or plant. A tissue sample can be from any tissue that can be infected or affected by a pathogen (e.g., a wart, lung tissue, skin tissue, and the like). A tissue sample (e.g., from animals, plants, or humans) can be dissociated or liquified prior to application to detection system of the present disclosure. A sample can be from a plant (e.g., a crop, a hydroponically grown crop or plant, and/or house plant). Plant samples can include extracellular fluid, from tissue (e.g., root, leaves, stem, trunk etc.). A sample can be taken from the environment immediately surrounding a plant, such as hydroponic fluid/water, or soil. A sample from an environment can be from soil, air, or water. In some instances, the environmental sample is taken as a swab from a surface of interest or taken directly from the surface of interest. In some instances, the raw sample is applied to the detection system. In some instances, the sample is diluted with a buffer or a fluid or concentrated prior to application to the detection system. In some cases, the sample is contained in no more than about 200 nanoliters (nL). In some cases, the sample is contained in about 200 nL. In some cases, the sample is contained in a volume that is greater than about 200 nL and less than about 20 microliters (μL). In some cases, the sample is contained in no more than 20 μl. In some cases, the sample is contained in no more than 1, 5, 10, 15, 20, 25, 30, 35 40, 45, 50, 55, 60, 65, 70, 75, 80, 90, 100, 200, 300, 400, 500 μl, or any of value from 1 μl to 500 μl. In some cases, the sample is contained in from 1 μL to 500 μL, from 10 μL to 500 μL, from 50 μL to 500 μL, from 100 μL to 500 μL, from 200 μL to 500 μL, from 300 μL to 500 μL, from 400 μL to 500 μL, from 1 μL to 200 μL, from 10 μL to 200 μL, from 50 μL to 200 μL, from 100 μL to 200 μL, from 1 μL to 100 μL, from 10 μL to 100 μL, from 50 μL to 100 μL, from 1 μL to 50 μL, from 10 μL to 50 μL, from 1 μL to 20 μL, from 10 μL to 20 μL, or from 1 μL to 10 μL. Sometimes, the sample is contained in more than 500 μl.


In some instances, the sample is taken from a single-cell eukaryotic organism; a plant or a plant cell; an algal cell; a fungal cell; an animal or an animal cell, tissue, or organ; a cell, tissue, or organ from an invertebrate animal; a cell, tissue, fluid, or organ from a vertebrate animal such as fish, amphibian, reptile, bird, and mammal; a cell, tissue, fluid, or organ from a mammal such as a human, a non-human primate, an ungulate, a feline, a bovine, an ovine, and a caprine. In some instances, the sample is taken from nematodes, protozoans, helminths, or malarial parasites. In some cases, the sample may comprise nucleic acids from a cell lysate from a eukaryotic cell, a mammalian cell, a human cell, a prokaryotic cell, or a plant cell. In some cases, the sample may comprise nucleic acids expressed from a cell.


The sample used for disease testing can comprise at least one target sequence that can bind to a guide nucleic acid of the reagents described herein. In some cases, the target sequence is a portion of a nucleic acid. A nucleic acid can be from a genomic locus, a transcribed mRNA, or a reverse transcribed cDNA. A nucleic acid can be from 5 to 100, 5 to 90, 5 to 80, 5 to 70, 5 to 60, 5 to 50, 5 to 40, 5 to 30, 5 to 25, 5 to 20, 5 to 15, or 5 to 10 nucleotides in length. A nucleic acid can be from 10 to 90, from 20 to 80, from 30 to 70, or from 40 to 60 nucleotides in length. A nucleic acid sequence can be from 10 to 95, from 20 to 95, from 30 to 95, from 40 to 95, from 50 to 95, from 60 to 95, from 10 to 75, from 20 to 75, from 30 to 75, from 40 to 75, from 50 to 75, from 5 to 50, from 15 to 50, from 25 to 50, from 35 to 50, or from 45 to 50 nucleotides in length. A nucleic acid can be 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 45, 50, 60, 70, 80, 90, or 100 nucleotides in length. The target nucleic acid can be reverse complementary to a guide nucleic acid. In some cases, at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 45, 50, 60, 70, 80, 90, or 100 nucleotides of a guide nucleic acid can be reverse complementary to a target nucleic acid.


In some cases, the target sequence is a portion of a nucleic acid from a virus or a bacterium or other agents responsible for a disease in the sample. The target sequence, in some cases, is a portion of a nucleic acid from a sexually transmitted infection or a contagious disease, in the sample. The target sequence, in some cases, is a portion of a nucleic acid from an upper respiratory tract infection, a lower respiratory tract infection, or a contagious disease, in the sample. The target sequence, in some cases, is a portion of a nucleic acid from a hospital acquired infection or a contagious disease, in the sample. The target sequence, in some cases, is a portion of a nucleic acid from sepsis, in the sample. These diseases can include but are not limited to respiratory viruses (e.g., SARS-COV-2 (i.e., a virus that causes COVID-19), SARS, MERS, influenza, Adenovirus, Coronavirus HKU1, Coronavirus NL63, Coronavirus 229E, Coronavirus OC43, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-COV-2), Human Metapneumovirus (hMPV), Human Rhinovirus/Enterovirus, Influenza A, Influenza A/H1, Influenza A/H3, Influenza A/H1-2009, Influenza B, Influenza C, Parainfluenza Virus 1, Parainfluenza Virus 2, Parainfluenza Virus 3, Parainfluenza Virus 4, Respiratory Syncytial Virus) and respiratory bacteria (e.g. Bordetella parapertussis, Bordetella pertussis, Chlamydia pneumoniae, Mycoplasma pneumoniae). Other viruses include human immunodeficiency virus (HIV), human papillomavirus (HPV), chlamydia, gonorrhea, syphilis, trichomoniasis, sexually transmitted infection, malaria, Dengue fever, Ebola, chikungunya, and leishmaniasis. Pathogens include viruses, fungi, helminths, protozoa, malarial parasites, Plasmodium parasites, Toxoplasma parasites, and Schistosoma parasites. Helminths include roundworms, heartworms, and phytophagous nematodes, flukes, Acanthocephala, and tapeworms. Protozoan infections include infections from Giardia spp., Trichomonas spp., African trypanosomiasis, amoebic dysentery, babesiosis, balantidial dysentery, Chaga's disease, coccidiosis, malaria and toxoplasmosis. Examples of pathogens such as parasitic/protozoan pathogens include, but are not limited to: Plasmodium falciparum, P. vivax, Trypanosoma cruzi and Toxoplasma gondii. Fungal pathogens include, but are not limited to Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis, Chlamydia pneumoniae, Chlamydia psittaci, and Candida albicans. Pathogenic viruses include but are not limited to: respiratory viruses (e.g., adenoviruses, parainfluenza viruses, severe acute respiratory syndrome (SARS), coronavirus, MERS), gastrointestinal viruses (e.g., noroviruses, rotaviruses, some adenoviruses, astroviruses), exanthematous viruses (e.g., the virus that causes measles, the virus that causes rubella, the virus that causes chickenpox/shingles, the virus that causes roseola, the virus that causes smallpox, the virus that causes fifth disease, chikungunya virus infection); hepatic viral diseases (e.g., hepatitis A, B, C, D, E); cutaneous viral diseases (e.g., warts (including genital, anal), herpes (including oral, genital, anal), molluscum contagiosum); hemmorhagic viral diseases (e.g. Ebola, Lassa fever, dengue fever, yellow fever, Marburg hemorrhagic fever, Crimean-Congo hemorrhagic fever); neurologic viruses (e.g., polio, viral meningitis, viral encephalitis, rabies), sexually transmitted viruses (e.g., HIV, HPV, and the like), immunodeficiency virus (e.g., HIV); influenza virus; dengue; West Nile virus; herpes virus; yellow fever virus; Hepatitis Virus C; Hepatitis Virus A; Hepatitis Virus B; papillomavirus; and the like. Pathogens include, e.g., HIV virus, Mycobacterium tuberculosis, Klebsiella pneumoniae, Acinetobacter baumannii, Bacillus anthracis, Bortadella pertussis, Burkholderia cepacia, Corynebacterium diphtheriae, Coxiella burnetii, Streptococcus agalactiae, methicillin-resistant Staphylococcus aureus, Legionella longbeachae, Legionella pneumophila, Leptospira interrogans, Moraxella catarrhalis, Streptococcus pyogenes, Escherichia coli, Neisseria gonorrhoeae, Neisseria meningitidis, Neisseria elongate, Neisseria gonorrhoeae, Parechovirus, Pneumococcus, Pneumocystis jirovecii, Cryptococcus neoformans, Histoplasma capsulatum, Haemophilus influenzae B, Treponema pallidum, Lyme disease spirochetes, Pseudomonas aeruginosa, Mycobacterium leprae, Brucella abortus, rabies virus, influenza virus, cytomegalovirus, herpes simplex virus I, herpes simplex virus II, human serum parvo-like virus, respiratory syncytial virus (RSV), M. genitalium, T. Vaginalis, varicella-zoster virus, hepatitis B virus, hepatitis C virus, measles virus, adenovirus, human T-cell leukemia viruses, Epstein-Barr virus, murine leukemia virus, mumps virus, vesicular stomatitis virus, Sindbis virus, lymphocytic choriomeningitis virus, wart virus, blue tongue virus, Sendai virus, feline leukemia virus, Reovirus, polio virus, simian virus 40, mouse mammary tumor virus, dengue virus, rubella virus, West Nile virus, Plasmodium falciparum, Plasmodium vivax, Toxoplasma gondii, Trypanosoma rangeli, Trypanosoma cruzi, Trypanosoma rhodesiense, Trypanosoma brucei, Schistosoma mansoni, Schistosoma japonicum, Babesia bovis, Eimeria tenella, Onchocerca volvulus, Leishmania tropica, Mycobacterium tuberculosis, Trichinella spiralis, Theileria parva, Taenia hydatigena, Taenia ovis, Taenia saginata, Echinococcus granulosus, Mesocestoides corti, Mycoplasma arthritidis, M. hyorhinis, M. orale, M. arginini, Acholeplasma laidlawii, M. salivarium, M. pneumoniae, Enterobacter cloacae, Kiebsiella aerogenes, Proteus vulgaris, Serratia macesens, Enterococcus faecalis, Enterococcus faecium, Streptococcus intermdius, Streptococcus pneumoniae, and Streptococcus pyogenes. Often the target nucleic acid may comprise a sequence from a virus or a bacterium or other agents responsible for a disease that can be found in the sample. In some cases, the target nucleic acid is a portion of a nucleic acid from a genomic locus, a transcribed mRNA, or a reverse transcribed cDNA from a gene locus in at least one of: human immunodeficiency virus (HIV), human papillomavirus (HPV), chlamydia, gonorrhea, syphilis, trichomoniasis, sexually transmitted infection, malaria, Dengue fever, Ebola, chikungunya, and leishmaniasis. Pathogens include viruses, fungi, helminths, protozoa, malarial parasites, Plasmodium parasites, Toxoplasma parasites, and Schistosoma parasites. Helminths include roundworms, heartworms, and phytophagous nematodes, flukes, Acanthocephala, and tapeworms. Protozoan infections include infections from Giardia spp., Trichomonas spp., African trypanosomiasis, amoebic dysentery, babesiosis, balantidial dysentery, Chaga's disease, coccidiosis, malaria and toxoplasmosis. Examples of pathogens such as parasitic/protozoan pathogens include, but are not limited to: Plasmodium falciparum, P. vivax, Trypanosoma cruzi and Toxoplasma gondii. Fungal pathogens include, but are not limited to Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis, and Candida albicans. Pathogenic viruses include but are not limited to immunodeficiency virus (e.g., HIV); influenza virus; dengue; West Nile virus; herpes virus; yellow fever virus; Hepatitis Virus C; Hepatitis Virus A; Hepatitis Virus B; papillomavirus; and the like. Pathogens include, e.g., HIV virus, Mycobacterium tuberculosis, Streptococcus agalactiae, methicillin-resistant Staphylococcus aureus, Staphylococcus epidermidis, Legionella pneumophila, Streptococcus pyogenes, Streptococcus salivarius, Escherichia coli, Neisseria gonorrhoeae, Neisseria meningitidis, Pneumococcus, Cryptococcus neoformans, Histoplasma capsulatum, Hemophilus influenzae B, Treponema pallidum, Lyme disease spirochetes, Pseudomonas aeruginosa, Mycobacterium leprae, Brucella abortus, rabies virus, influenza virus, cytomegalovirus, herpes simplex virus I, herpes simplex virus II, human serum parvo-like virus, respiratory syncytial virus (RSV), M. genitalium, T. vaginalis, varicella-zoster virus, hepatitis B virus, hepatitis C virus, measles virus, adenovirus, human T-cell leukemia viruses, Epstein-Barr virus, murine leukemia virus, mumps virus, vesicular stomatitis virus, Sindbis virus, lymphocytic choriomeningitis virus, wart virus, blue tongue virus, Sendai virus, feline leukemia virus, Reovirus, polio virus, simian virus 40, mouse mammary tumor virus, dengue virus, rubella virus, West Nile virus, Plasmodium falciparum, Plasmodium vivax, Toxoplasma gondii, Trypanosoma rangeli, Trypanosoma cruzi, Trypanosoma rhodesiense, Trypanosoma brucei, Schistosoma mansoni, Schistosoma japonicum, Babesia bovis, Eimeria tenella, Onchocerca volvulus, Leishmania tropica, Mycobacterium tuberculosis, Trichinella spiralis, Theileria parva, Taenia hydatigena, Taenia ovis, Taenia saginata, Echinococcus granulosus, Mesocestoides corti, Mycoplasma arthritidis, M. hyorhinis, M. orale, M. arginini, Acholeplasma laidlawii, M. salivarium and M. pneumoniae. In some cases, the target sequence is a portion of a nucleic acid from a genomic locus, a transcribed mRNA, or a reverse transcribed cDNA from a gene locus of bacterium or other agents responsible for a disease in the sample comprising a mutation that confers resistance to a treatment, such as a single nucleotide mutation that confers resistance to antibiotic treatment.


The sample used for cancer testing or cancer risk testing can comprise at least one target sequence or target nucleic acid segment that can bind to a guide nucleic acid of the reagents described herein. The target nucleic acid segment, in some cases, is a portion of a nucleic acid from a gene with a mutation associated with cancer, from a gene whose overexpression is associated with cancer, a tumor suppressor gene, an oncogene, a checkpoint inhibitor gene, a gene associated with cellular growth, a gene associated with cellular metabolism, or a gene associated with cell cycle. Sometimes, the target nucleic acid encodes for a cancer biomarker, such as a prostate cancer biomarker or non-small cell lung cancer. In some cases, the assay can be used to detect “hotspots” in target nucleic acids that can be predictive of cancer, such as lung cancer, cervical cancer, in some cases, the cancer can be a cancer that is caused by a virus. Some non-limiting examples of viruses that cause cancers in humans include Epstein-Barr virus (e.g., Burkitt's lymphoma, Hodgkin's Disease, and nasopharyngeal carcinoma); papillomavirus (e.g., cervical carcinoma, anal carcinoma, oropharyngeal carcinoma, penile carcinoma); hepatitis B and C viruses (e.g., hepatocellular carcinoma); human adult T-cell leukemia virus type 1 (HTLV-1) (e.g., T-cell leukemia); and Merkel cell polyomavirus (e.g., Merkel cell carcinoma). One skilled in the art will recognize that viruses can cause or contribute to other types of cancers. In some cases, the target nucleic acid is a portion of a nucleic acid that is associated with a blood fever. In some cases, the target nucleic acid segment is a portion of a nucleic acid from a genomic locus, a transcribed mRNA, or a reverse transcribed cDNA from a locus of at least one of: ALK, APC, ATM, AXIN2, BAP1, BARDI, BLM, BMPRIA, BRCA1, BRCA2, BRIP1, CASR, CDC73, CDHI, CDK4, CDKNIB, CDKNIC, CDKN2A, CEBPA, CHEK2, CTNNA1, DICER1, DIS3L2, EGFR, EPCAM, FH, FLCN, GATA2, GPC3, GREMI, HOXB13, HRAS, KIT, MAX, MENI, MET, MITF, MLH1, MSH2, MSH3, MSH6, MUTYH, NBN, NF1, NF2, NTHL1, PALB2, PDGFRA, PHOX2B, PMS2, POLDI, POLE, POTI, PRKARIA, PTCHI, PTEN, RAD50, RAD51C, RAD51D, RB1, RECQL4, RET, RUNX1, SDHA, SDHAF2, SDHB, SDHC, SDHD, SMAD4, SMARCA4, SMARCB1, SMARCE1, STK11, SUFU, TERC, TERT, TMEM127, TP53, TSC1, TSC2, VHL, WRN, and WT1.


The sample used for genetic disorder testing can comprise at least one target sequence or target nucleic acid segment that can bind to a guide nucleic acid of the reagents described herein. In some embodiments, the genetic disorder is hemophilia, sickle cell anemia, ß-thalassemia, Duchene muscular dystrophy, severe combined immunodeficiency, or cystic fibrosis. The target nucleic acid segment, in some cases, is a portion of a nucleic acid from a gene with a mutation associated with a genetic disorder, from a gene whose overexpression is associated with a genetic disorder, from a gene associated with abnormal cellular growth resulting in a genetic disorder, or from a gene associated with abnormal cellular metabolism resulting in a genetic disorder. In some cases, the target nucleic acid segment is a portion of a nucleic acid from a genomic locus, a transcribed mRNA, or a reverse transcribed cDNA from a locus of at least one of: CFTR, FMR1, SMNI, ABCB11, ABCC8, ABCDI, ACAD9, ACADM, ACADVL, ACATI, ACOXI, ACSF3, ADA, ADAMTS2, ADGRG1, AGA, AGL, AGPS, AGXT, AIRE, ALDH3A2, ALDOB, ALG6, ALMS1, ALPL, AMT, AQP2, ARGI, ARSA, ARSB, ASL, ASNS, ASPA, ASSI, ATM, ATP6VIB1, ATP7A, ATP7B, ATRX, BBS1, BBS10, BBS12, BBS2, BCKDHA, BCKDHB, BCSIL, BLM, BSND, CAPN3, CBS, CDH23, CEP290, CERKL, CHM, CHRNE, CIITA, CLN3, CLN5, CLN6, CLN8, CLRNI, CNGB3, COL27A1, COL4A3, COL4A4, COL4A5, COL7A1, CPS1, CPTIA, CPT2, CRB1, CTNS, CTSK, CYBA, CYBB, CYP11B1, CYP11B2, CYP17A1, CYP19A1, CYP27A1, DBT, DCLREIC, DHCR7, DHDDS, DLD, DMD, DNAH5, DNAI1, DNAI2, DYSF, EDA, EIF2B5, EMD, ERCC6, ERCC8, ESCO2, ETFA, ETFDH, ETHEI, EVC, EVC2, EYS, F9, FAH, FAM161A, FANCA, FANCC, FANCG, FH, FKRP, FKTN, G6PC, GAA, GALC, GALKI, GALT, GAMT, GBA, GBE1, GCDH, GFM1, GJB1, GJB2, GLA, GLB1, GLDC, GLE1, GNE, GNPTAB, GNPTG, GNS, GRHPR, HADHA, HAX1, HBA1, HBA2, HBB, HEXA, HEXB, HGSNAT, HLCS, HMGCL, HOGAI, HPS1, HPS3, HSD17B4, HSD3B2, HYALI, HYLSI, IDS, IDUA, IKBKAP, IL2RG, IVD, KCNJ11, LAMA2, LAMA3, LAMB3, LAMC2, LCA5, LDLR, LDLRAP1, LHX3, LIFR, LIPA, LOXHD1, LPL, LRPPRC, MAN2B1, MCOLN1, MED17, MESP2, MFSD8, MKS1, MLC1, MMAA, MMAB, MMACHC, MMADHC, MPI, MPL, MPV17, MTHFR, MTM1, MTRR, MTTP, MUT, MYO7A, NAGLU, NAGS, NBN, NDRG1, NDUFAF5, NDUFS6, NEB, NPC1, NPC2, NPHS1, NPHS2, NR2E3, NTRK1, OAT, OPA3, OTC, PAH, PC, PCCA, PCCB, PCDH15, PDHA1, PDHB, PEX1, PEX10, PEX12, PEX2, PEX6, PEX7, PFKM, PHGDH, PKHD1, PMM2, POMGNT1, PPTI, PROPI, PRPSI, PSAP, PTS, PUSI, PYGM, RAB23, RAG2, RAPSN, RARS2, RDH12, RMRP, RPE65, RPGRIPIL, RSI, RTELI, SACS, SAMHD1, SEPSECS, SGCA, SGCB, SGCG, SGSH, SLC12A3, SLC12A6, SLC17A5, SLC22A5, SLC25A13, SLC25A15, SLC26A2, SLC26A4, SLC35A3, SLC37A4, SLC39A4, SLC4A11, SLC6A8, SLC7A7, SMARCAL1, SMPD1, STAR, SUMF1, TAT, TCIRG1, TECPR2, TFR2, TGM1, TH, TMEM216, TPP1, TRMU, TSFM, TTPA, TYMP, USHIC, USH2A, VPS13A, VPS13B, VPS45, VRK1, VSX2, WNT10A, XPA, XPC, and ZFYVE26.


The method can comprise generating one or more droplets, aliquots, or subsamples from the sample. The one or more droplets, aliquots, or subsamples can correspond to a volumetric portion of the sample. The sample can be divided into 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or more droplets, aliquots, or subsamples. In some embodiments, the sample is not divided into subsamples. As used herein, a droplet can refer to a volumetric portion of the sample, a partitioned sub-sample of the sample, and/or an aliquot of the sample. In some cases, the detection chamber is configured to receive and contact the plurality of droplets with at least one programmable nuclease disposed on a surface of said detection chamber. The at least one programmable nuclease can comprise a guide nucleic acid complexed with a programmable nuclease. In some cases, the programmable nuclease may comprise a CRISPR/Cas enzyme. In some cases, the guide nucleic acid may comprise a guide RNA. In some embodiments, the device may comprise a plurality of programmable nuclease complexes comprising different guide RNAs.


The method can comprise amplifying one or more targets within each droplet, aliquot, or subsample. Amplification of the one or more targets within each droplet can be performed in parallel and/or simultaneously for each droplet. Dividing the sample into a plurality of droplets can enhance a speed and/or an efficiency of the amplification process (e.g., a thermocycling process) since the droplets comprise a smaller volume of material than the bulk sample introduced. Amplifying the one or more targets within each individual droplet can also permit effective amplification of various target nucleic acids that cannot be amplified as efficiently in a bulk sample containing the various target nucleic acids if the bulk sample were to undergo a singular amplification process. In some embodiments, amplification is performed on the bulk sample without first dividing the sample into subsamples. The method can further comprise using a CRISPR-based or programmable nuclease-based detection module to detect one or more targets (e.g., target sequences or target nucleic acids) in the sample. In some cases, the sample can be divided into a plurality of droplets, aliquots, or subsamples to facilitate sample preparation.


Described herein are various methods of sample preparation and reagent storage. Any of the devices described herein may comprise one or more sample preparation reagents. Any of the devices described herein may comprise sample preparation reagents as dried reagents. Dried reagents may comprise solids and/or semi-solids. In certain instances, dried reagents may comprise lyophilized reagents. Any of the devices described herein may comprise one or more lyophilized reagents (e.g., amplification reagents, programmable nucleases, buffers, excipients, etc.). In certain instances, methods include sample lysis, concentration, and/or filtration. In certain instances, methods include reconstitution of one or more lyophilized reagents. In some embodiments, lyophilized reagents may be in the form of lyophilized beads, spheres, and/or particulates. In some embodiments, the lyophilized bead, sphere, and/or particulate may comprise either single or multiple compounds. In some embodiments, the lyophilized bead, sphere, and/or particulate may be adjusted to various moisture levels or hygroscopy. In some embodiments, the lyophilized bead, sphere, and/or particulate may comprise assay internal standards. In some embodiments, the lyophilized bead, sphere, and/or particulate may have diameters between about 0.5 millimeters to about 5 millimeters in diameter.


The sample can be prepared before one or more targets are detected within the sample. The sample preparation steps described herein can process a crude sample to generate a pure or purer sample. Sample preparation can one or more physical or chemical processes, including, for example, nucleic acid purification, lysis, binding, washing, and/or eluting. In certain instances, sample preparation can comprise the following steps, in any order, including sample collection, nucleic acid purification, heat inactivation, and/or base/acid lysis.


In some embodiments, nucleic acid purification can be performed on the sample. Purification can comprise disrupting a biological matrix of a cell to release nucleic acids, denaturing structural proteins associated with the nucleic acids (nucleoproteins), inactivating nucleases that can degrade the isolated product (RNase and/or DNase), and/or removing contaminants (e.g., proteins, carbohydrates, lipids, biological or environmental elements, unwanted nucleic acids, and/or other cellular debris).


In some embodiments, lysis of a collected sample can be performed. Lysis can be performed using a protease (e.g., a Proteinase K or PK enzyme). In some cases, a solution of reagents can be used to lyse the cells in the sample and release the nucleic acids so that they are accessible to the programmable nuclease. Active ingredients of the solution can be chaotropic agents, detergents, salts, and can be of high osmolality, ionic strength, and pH. Chaotropic agents or chaotropes are substances that disrupt the three-dimensional structure in macromolecules such as proteins, DNA, or RNA. One example protocol may comprise a 4 M guanidinium isothiocyanate, 25 mM sodium citrate·2H20, 0.5% (w/v) sodium lauryl sarcosinate, and 0.1 M ß-mercaptoethanol), but numerous commercial buffers for different cellular targets can also be used. Alkaline buffers can also be used for cells with hard shells, particularly for environmental samples. Detergents such as sodium dodecyl sulphate (SDS) and cetyl trimethylammonium bromide (CTAB) can also be implemented to chemical lysis buffers. Cell lysis can also be performed by physical, mechanical, thermal or enzymatic means, in addition to chemically-induced cell lysis mentioned previously. In some cases, depending on the type of sample, nanoscale barbs, nanowires, acoustic generators, integrated lasers, integrated heaters, and/or microcapillary probes can be used to perform lysis.


In certain instances, heat inactivation can be performed on the sample. In some embodiments, a processed/lysed sample can undergo heat inactivation to inactivate, in the lysed sample, the proteins used during lysing (e.g., a PK enzyme or a lysing reagent). In some cases, a heating element integrated into the detection device can be used for heat-inactivation. The heating element can be powered by a battery or another source of thermal or electric energy that is integrated with the detection device.


In some cases, a target nucleic acid within the sample can undergo amplification before binding to a guide nucleic acid, for example a crRNA or sgRNA of a CRISPR enzyme. The target nucleic acid within a purified sample can be amplified. In some instances, amplification can be accomplished using loop mediated amplification (LAMP), isothermal recombinase polymerase amplification (RPA), rolling circle amplification (RCA), nicking enzyme amplification reaction (NEAR), FASTR, and/or polymerase chain reaction (PCR). In some instances, digital droplet amplification can used. Such nucleic acid amplification of the sample can improve at least one of a sensitivity, specificity, or accuracy of the detection of the target nucleic acid. The reagents for nucleic acid amplification can comprise a recombinase, an oligonucleotide primer, a single-stranded DNA binding (SSB) protein, and a polymerase. The nucleic acid amplification can be transcription mediated amplification (TMA). Nucleic acid amplification can be helicase dependent amplification (HDA) or circular helicase dependent amplification (cHDA). In additional cases, nucleic acid amplification is strand displacement amplification (SDA). The nucleic acid amplification can be recombinase polymerase amplification (RPA). The nucleic acid amplification can be at least one of loop mediated amplification (LAMP) or the exponential amplification reaction (EXPAR). Nucleic acid amplification is, in some cases, by rolling circle amplification (RCA), ligase chain reaction (LCR), simple method amplifying RNA targets (SMART), single primer isothermal amplification (SPIA), multiple displacement amplification (MDA), nucleic acid sequence-based amplification (NASBA), hinge-initiated primer-dependent amplification of nucleic acids (HIP), nicking enzyme amplification reaction (NEAR), or improved multiple displacement amplification (IMDA). The nucleic acid amplification can be performed for no greater than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 40, 50, or 60 minutes. Sometimes, the nucleic acid amplification is performed for from 1 to 60, from 5 to 55, from 10 to 50, from 15 to 45, from 20 to 40, or from 25 to 35 minutes. Sometimes, the nucleic acid amplification is performed for from 5 to 60, from 10 to 60, from 15 to 60, from 30 to 60, from 45 to 60, from 1 to 45, from 5 to 45, from 10 to 45, from 30 to 45, from 1 to 30, from 5 to 30, from 10 to 30, from 15 to 30, from 1 to 15, from 5 to 15, or from 10 to 15 minutes.


In some embodiments, amplification can comprise thermocycling of the sample. Thermocycling can be carried out for one or more droplets of the sample in parallel and/or independently in separate locations. This can be accomplished by methods such as (1) by holding droplets stationary in locations where a heating element is in close proximity to the droplet on one of the droplet sides and a heat sink element is in close proximity to the other side of the droplet, or (2) flowing the droplet through zones in a fluid channel where heat flows across it from a heating source to a heat sink. In some cases, one or more resistive heating elements can be used to perform thermocycling. In some cases, the thermocycling of the sample may comprise one or more reactions at different temperatures. In some cases, the reactions can include one or more reactions selected from an annealing reaction, a denaturation reaction, an extension reaction, reverse transcription reaction, and a detection reaction. In some cases, the thermocycling of the sample may comprise amplification of one or more nucleic acids in the sample.


In some cases, the annealing temperature of the reaction is performed at a temperature around 45° C. to 75° C. In some embodiments, the annealing temperature may be at a temperature of about 45° C., about 47° C., about 48° C., about 49° C., about 50° C., about 52° C., about 54° C., about 56° ° C., about 58° C., about 60° C., about 62° C., about 64° C., about 66° C., about 68° C., about 70° C., about 72° C., about 74° C., or about 76° C.


In some cases, the denaturation temperature of the reaction is performed at a temperature around 90° ° C. to about 110° C. In some embodiments, the denaturation temperature may be at a temperature of about 90° ° C., about 91° ° C., about 92° C., about 93° C., about 94° C., about 95° C., about 96° C., about 97° C., about 98° C., about 99° C., about 100° C., about 101° C., about 102° C., about 103° C., about 104° C., about 105° C., about 106° C., about 107° C., about 108° C., about 109° C., or about 110° C.


In some cases, the extension temperature of the reaction is performed at a temperature from around 55° C. to about 85° C. In some embodiments, the extension temperature may be at a temperature of about 55° C., about 57° C., about 59° C., about 60° C., about 61° C., about 62° C., about 63° C., about 64° C., about 65° C., about 66° C., about 68° C., about 70° C., about 71° C., about 72° C., about 73° C., about 75° C., about 76° C., about 78° C., about 80° C., about 81° C., about 82° C., about 83° C., about 84° C., or about 85° C.


In some cases, the reverse transcription step of the reaction is performed at a temperature of around 45° C. to about 75° C. In some embodiments, the reverse transcription may be at a temperature of about 45° C., about 47° C., about 48° C., about 49° C., about 50° C., about 52° C., about 54° C., about 55° C., about 57° C., about 59° C., about 60° ° C., about 61° C., about 63° C., about 65° C., about 66° C., about 68° C., about 70° C., about 72° C., about 73° C., or about 75° C.


In some cases, the detection step of the reaction is performed at a temperature of about 30° ° C. to about 50° C. In some embodiments, the detection step may be at a temperature of about 30° C., about 32° C., about 33° C., about 34° C., about 35° C., about 36° C., about 37° C., about 38° C., about 39° C., about 40° C., about 42° C., about 43° C., about 44° C., about 45° C., about 46° ° C., or about 50° C.


Sometimes, the nucleic acid amplification reaction is performed at a temperature of around 20-45° C. The nucleic acid amplification reaction can be performed at a temperature no greater than 20° C., 25° C., 30° C., 35° C., 37° C., 40° C., 45° C., 50° C., 55° C., 60° C., or 65° C. The nucleic acid amplification reaction can be performed at a temperature of at least 20° C., 25° C., 30° C., 35° C., 37° C., 40° ° C., 45° C., 50° C., 55° C., 60° C., or 65° C. In some cases, the nucleic acid amplification reaction is performed at a temperature of from 20° C. to 45° C., from 25° C. to 40° C., from 30° C. to 40° C., or from 35° C. to 40° C. In some cases, the nucleic acid amplification reaction is performed at a temperature of from 45° ° C. to 65° C., from 50° C. to 65° C., from 55° C. to 65° C., or from 60° C. to 65° C. In some cases, the nucleic acid amplification reaction can be performed at a temperature that ranges from about 20° C. to 45° C., from 25° C. to 45° C., from 30° C. to 45° C., from 35° C. to 45° C., from 40° ° C. to 45° C., from 20° C. to 37° C., from 25° C. to 37° C., from 30° C. to 37° C., from 35° C. to 37° C., from 20° ° C. to 30° C., from 25° C. to 30° C., from 20° C. to 25° C., or from about 22° ° C. to 25° C. In some cases, the nucleic acid amplification reaction can be performed at a temperature that ranges from about 40° ° C. to 65° C., from 45° C. to 65° C., from 50° C. to 65° C., from 55° C. to 65° C., from 60° ° C. to 65° C., from 40° C. to 60° C., from 45° C. to 60° C., from 50° C. to 60° C., from 55° C. to 60° C., from 40° ° C. to 55° C., from 45° C. to 55° C., from 50° C. to 55° C., from 40° ° C. to 50° C., or from about 45° C. to 50° C.


Additionally, target nucleic acid can optionally be amplified before binding to the guide nucleic acid (e.g., crRNA or sgRNA) of the programmable nuclease (e.g., CRISPR enzyme). This amplification can be PCR amplification or isothermal amplification. This nucleic acid amplification of the sample can improve at least one of sensitivity, specificity, or accuracy of the detection the target nucleic acid. The reagents for nucleic acid amplification can comprise a recombinase, a oligonucleotide primer, a single-stranded DNA binding (SSB) protein, and a polymerase. The nucleic acid amplification can be transcription mediated amplification (TMA). Nucleic acid amplification can be helicase dependent amplification (HDA) or circular helicase dependent amplification (cHDA). In additional cases, nucleic acid amplification is strand displacement amplification (SDA). The nucleic acid amplification can be recombinase polymerase amplification (RPA). The nucleic acid amplification can be at least one of loop mediated amplification (LAMP) or the exponential amplification reaction (EXPAR). Nucleic acid amplification is, in some cases, by rolling circle amplification (RCA), ligase chain reaction (LCR), simple method amplifying RNA targets (SMART), single primer isothermal amplification (SPIA), multiple displacement amplification (MDA), nucleic acid sequence based amplification (NASBA), hinge-initiated primer-dependent amplification of nucleic acids (HIP), nicking enzyme amplification reaction (NEAR), or improved multiple displacement amplification (IMDA). The nucleic acid amplification can be performed for no greater than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 40, 50, or 60 minutes. Sometimes, the nucleic acid amplification reaction is performed at a temperature of around 20-45° ° C. Sometimes, the nucleic acid amplification reaction is performed at a temperature of around 45-65° C. The nucleic acid amplification reaction can be performed at a temperature no greater than 20° C., 25° C., 30° C., 35° ° C., 37° ° C., 40° ° C., 45° C., 50° C., 55° C., 60° ° C., or 65° C. The nucleic acid amplification reaction can be performed at a temperature of at least 20° C., 25° ° C., 30° C., 35° C., 37° C., 40° ° C., 45° C., 50° C., 55° C., 60° C., or 65° C.


Devices of the present disclosure can be configured to perform droplet digitization or droplet generation. Droplet digitization or generation can comprise splitting a volume of the sample into multiple droplets, aliquots, or subsamples. The sample can have a volume that ranges from about 10 microliters to about 500 microliters. The plurality of droplets, aliquots, or subsamples can have a volume that ranges from about 0.01 microliters to about 100 microliters. The plurality of droplets, aliquots, or subsamples can have a same or substantially similar volume. In some cases, the plurality of droplets, aliquots, or subsamples can have different volumes. In some cases, the droplets, aliquots, or subsamples can be generated using a physical filter or one or more movable mechanisms (e.g., valves, etc.). In some cases, each droplet of the sample can undergo one or more sample preparation steps (e.g., nucleic acid purification, lysis, heat inactivation, amplification, etc.) independently and/or in parallel while the droplets are physically constrained or thermally isolated between two movable mechanisms.


The sample used for phenotyping testing can comprise at least one target nucleic acid segment that can bind to a guide nucleic acid of the reagents described herein. The target nucleic acid segment, in some cases, is a portion of a nucleic acid from a gene associated with a phenotypic trait.


The sample used for genotyping testing can comprise at least one target nucleic acid segment that can bind to a guide nucleic acid of the reagents described herein. The target nucleic acid segment, in some cases, is a portion of a nucleic acid from a gene associated with a genotype.


The sample used for ancestral testing can comprise at least one target nucleic acid segment that can bind to a guide nucleic acid of the reagents described herein. The target nucleic acid segment, in some cases, is a portion of a nucleic acid from a gene associated with a geographic region of origin or ethnic group.


The sample can be used for identifying a disease status. For example, a sample is any sample described herein, and is obtained from a subject for use in identifying a disease status of a subject. The disease can be a cancer or genetic disorder. Sometimes, a method may comprise obtaining a serum sample from a subject; and identifying a disease status of the subject. Often, the disease status is prostate disease status. In any of the embodiments described herein, the device can be configured for asymptomatic, pre-symptomatic, and/or symptomatic diagnostic applications, irrespective of immunity. In any of the embodiments described herein, the device can be configured to perform one or more serological assays on a sample (e.g., a sample comprising blood).


In some embodiments, the sample can be used to identify a mutation in a target nucleic acid of a plant or of a bacteria, virus, or microbe associated with a plant or soil. The devices and methods of the present disclosure can be used to identify a mutation of a target nucleic acid that affects the expression of a gene. A mutation that affects the expression of gene can be a mutation of a target nucleic acid within the gene, a mutation of a target nucleic acid comprising RNA associated with the expression of a gene, or a target nucleic acid comprising a mutation of a nucleic acid associated with regulation of expression of a gene, such as an RNA or a promoter, enhancer, or repressor of the gene. Often, the mutation is a single nucleotide mutation.


In some instances, the target nucleic acid is a single stranded nucleic acid. Alternatively, or in combination, the target nucleic acid is a double stranded nucleic acid and is prepared into single stranded nucleic acids before or upon contacting the reagents. The target nucleic acid can be a RNA, DNA, synthetic nucleic acids, or nucleic acids found in biological or environmental samples. The target nucleic acids include but are not limited to mRNA, rRNA, RNA, non-coding RNA, long non-coding RNA, and microRNA (miRNA). In some cases, the target nucleic acid is mRNA. In some cases, the target nucleic acid is from a virus, a parasite, or a bacterium described herein. In some cases, the target nucleic acid is transcribed from a gene as described herein.


A number of target nucleic acids are consistent with the systems and methods disclosed herein. Some methods described herein can detect a target nucleic acid present in the sample in various concentrations or amounts as a target nucleic acid population. In some cases, the sample has at least 2 target nucleic acids. In some cases, the sample has at least 3, 5, 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, or 10000 target nucleic acids. In some cases, the sample has from 1 to 10,000, from 100 to 8000, from 400 to 6000, from 500 to 5000, from 1000 to 4000, or from 2000 to 3000 target nucleic acids. In some cases, the sample has from 100 to 9500, from 100 to 9000, from 100 to 8500, from 100 to 8000, from 100 to 7500, from 100 to 7000, from 100 to 6500, from 100 to 6000, from 100 to 5500, from 100 to 5000, from 250 to 9500, from 250 to 9000, from 250 to 8500, from 250 to 8000, from 250 to 7500, from 250 to 7000, from 250 to 6500, from 250 to 6000, from 250 to 5500, from 250 to 5000, from 2500 to 9500, from 2500 to 9000, from 2500 to 8500, from 2500 to 8000, from 2500 to 7500, from 2500 to 7000, from 2500 to 6500, from 2500 to 6000, from 2500 to 5500, or from 2500 to 5000 target nucleic acids. In some cases, the method detects target nucleic acid present at least at one copy per 101 non-target nucleic acids, 102 non-target nucleic acids, 103 non-target nucleic acids, 104 non-target nucleic acids, f, 106 non-target nucleic acids, 107 non-target nucleic acids, 108 non-target nucleic acids, 109 non-target nucleic acids, or 1010 non-target nucleic acids.


In some cases, the target nucleic acid is present at less than 1 μg/ml in a sample. In some cases, the target nucleic acid is present at less than 100 ng/ml in a sample. In some cases, the target nucleic acid is present at less than 10 ng/ml in a sample. In some cases, the target nucleic acid is present at less than 1 ng/ml in a sample. In some cases, the target nucleic acid is present at less than 100 pg/ml in a sample. In some cases, the target nucleic acid is present at less than 10 pg/ml in a sample. In some cases, the target nucleic acid is present at less than 1 pg/ml in a sample. In some cases, the target nucleic acid is present at less than 100 fg/ml in a sample. In some cases, the target nucleic acid is present at less than 10 fg/ml in a sample. In some cases, the target nucleic acid is present at less than 1 fg/ml in a sample.


A number of target nucleic acid populations are consistent with the systems and methods disclosed herein. Some methods described herein can be implemented to detect two or more target nucleic acid populations present in the sample in various concentrations or amounts. In some cases, the sample has at least 2 target nucleic acid populations. In some cases, the sample has at least 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, or 50 target nucleic acid populations. In some cases, the sample has from 3 to 50, from 5 to 40, or from 10 to 25 target nucleic acid populations. In some cases, the sample has from 2 to 50, from 5 to 50, from 10 to 50, from 2 to 25, from 3 to 25, from 4 to 25, from 5 to 25, from 10 to 25, from 2 to 20, from 3 to 20, from 4 to 20, from 5 to 20, from 10 to 20, from 2 to 10, from 3 to 10, from 4 to 10, from 5 to 10, from 6 to 10, from 7 to 10, from 8 to 10, or from 9 to 10 target nucleic acid populations. In some cases, the methods of the present disclosure can be implemented to detect target nucleic acid populations that are present at least at one copy per 101 non-target nucleic acids, 102 non-target nucleic acids, 103 non-target nucleic acids, 104 non-target nucleic acids, 105 non-target nucleic acids, 106 non-target nucleic acids, 107 non-target nucleic acids, 108 non-target nucleic acids, 109 non-target nucleic acids, or 1010 non-target nucleic acids. The target nucleic acid populations can be present at different concentrations or amounts in the sample.


The compositions and methods of the present disclosure are compatible with intact and damaged nucleic acids. The target nucleic acid may be fragmented prior to amplification and/or detection (e.g., by a DETECTR assay). The target nucleic acid may comprise ordered or random fragmentation. The target nucleic acid may also comprise damage, such as oxidation or cross-linking. The target nucleic acid may comprise chemical, light-induced, and/or enzymatic cleavage. Conversely, the target nucleic acid may be unfragmented. A target nucleic acid sequence may be embedded within a longer nucleic acid. For example, a target nucleic acid may be a gene within an intact chromosome. A target nucleic acid may comprise a chemical modification, such as acetylation or methylation. A target nucleic acid may comprise a defined secondary, tertiary, or quaternary structure. A target nucleic acid may be complexed with another species, such as a histone.


Any of the above disclosed samples are consistent with the systems, assays, and programmable nucleases disclosed herein and can be used as a companion diagnostic with any of the diseases disclosed herein (e.g., a coronavirus infection), or can be used in reagent kits, point-of-care diagnostics, or over-the-counter diagnostics.


Reagents

A number of reagents are consistent with the methods, reagents, and devices disclosed herein. Many aspects of the present disclosure provide buffers, systems, and compositions that enable fast kinetics (e.g., within 25%, 50%, 75%, or 90% of the fastest reported rate for a reaction at a given temperature, or coming to completion within one hour) for multiple types of reactions or processes. Such buffers, systems, and compositions enable multiple reactions to be performed on a sample without intervening sample transfers, buffer exchanges, or reagent removal. In some cases, reagents (e.g., enzymes) for multiple reactions may be provided in a single solution. Such cases may require no more than sample addition to determine whether a nucleic acid (e.g., a particular nucleic acid sequence) is present in the sample.


These reagents are compatible with the samples, methods, and devices as described herein for detection of an ailment, such as a disease. The reagents described herein for detecting a disease, such as coronavirus, comprise multiple guide nucleic acids, each guide nucleic acid targeting a target nucleic acid segment indicative of the disease. Each guide nucleic acid binds to the target nucleic acid comprising a segment of a nucleic acid sequence (e.g., a nucleic acid from coronavirus) as described herein. Each guide nucleic acid can bind to the target nucleic acid comprising a portion of a nucleic acid (e.g., a target nucleic acid from coronavirus) as described herein and further comprising a mutation, such as a single nucleotide polymorphism (SNP), that can confer resistance to a treatment, such as antibiotic treatment. Each guide nucleic acid binds to the target nucleic acid comprising a portion of a nucleic acid. Each guide nucleic acid is complementary to a target nucleic acid. Often the guide nucleic acid binds specifically to the target nucleic acid. The target nucleic acid may be a RNA, DNA, or synthetic nucleic acids.


Disclosed herein are methods of assaying for a plurality of target nucleic acids (e.g., a plurality of nucleic acids from coronavirus) as described herein. For example, a method of assaying for a plurality of target nucleic acids in a sample comprises contacting the sample to a complex comprising a plurality guide nucleic acid sequences, each guide nucleic acid sequence comprising a segment that is reverse complementary to a segment of the target nucleic acid, and programmable nucleases that exhibits sequence independent cleavage upon forming a complex comprising the segment of the guide nucleic acid binding to the segment of the target nucleic acid; and assaying for a signal indicating cleavage of at least some protein-nucleic acids of a population of protein-nucleic acids, wherein the signal indicates a presence of one or more target nucleic acid of the plurality of target nucleic acids in the sample and wherein absence of the signal indicates an absence of the target nucleic acids in the sample. As another example, a method of assaying for a target nucleic acid in a sample, for example, comprises: a) contacting the sample to a plurality of complexes, each complex comprising a guide nucleic acid comprising a segment that is reverse complementary to a segment of the target nucleic acid and a programmable nuclease that exhibits sequence independent cleavage upon forming a complex comprising the segment of the guide nucleic acid binding to the segment of the target nucleic acid; b) contacting the plurality of complexes to a substrate; c) contacting the substrate to a reagent that differentially reacts with a cleaved substrate; and d) assaying for a signal indicating cleavage of the substrate, wherein the signal indicates a presence of the target nucleic acid in the sample and wherein absence of the signal indicates an absence of the target nucleic acid in the sample. Often, the substrate is an enzyme-nucleic acid. Sometimes, the substrate is an enzyme substrate-nucleic acid.


A programmable nuclease can comprise a programmable nuclease capable of being activated when complexed with a guide nucleic acid and target nucleic acid. The programmable nuclease can become activated after binding of a guide nucleic acid with a target nucleic acid, in which the activated programmable nuclease can cleave the target nucleic acid and can have trans cleavage activity. Trans cleavage activity can be non-specific cleavage of nearby single-stranded nucleic acids by the activated programmable nuclease, such as trans cleavage of reporters with a detection moiety. Once the reporter is cleaved by the activated programmable nuclease, the detection moiety can be released from the reporter and can generate a signal. A signal can be a calorimetric, potentiometric, amperometric, optical (e.g., fluorescent, colorimetric, etc.), or piezo-electric signal. Often, the signal is present prior to reporter cleavage and changes upon reporter cleavage. Sometimes, the signal is absent prior to reporter cleavage and is present upon reporter cleavage. The detectable moiety generating the detectable signal can be immobilized on a support medium for detection. The programmable nuclease can be a CRISPR-Cas (clustered regularly interspaced short palindromic repeats—CRISPR associated) nucleoprotein complex with trans cleavage activity, which can be activated by binding of a guide nucleic acid with a target nucleic acid. The CRISPR-Cas nucleoprotein complex can comprise a Cas protein (also referred to as a Cas nuclease) complexed with a guide nucleic acid, which can also be referred to as CRISPR enzyme. A guide nucleic acid can be a CRISPR RNA (crRNA). Sometimes, a guide nucleic acid comprises a crRNA and a trans-activating crRNA (tracrRNA).


The term, “guide nucleic acid,” as used herein refers to a nucleic acid comprising: a first nucleotide sequence that hybridizes to a target nucleic acid; and a second nucleotide sequence that capable of being non-covalently bound by an effector protein. The first sequence may be referred to herein as a spacer sequence. The second sequence may be referred to herein as a repeat sequence. In some embodiments, the first sequence is located 5′ of the second nucleotide sequence. In some embodiments, the first sequence is located 3′ of the second nucleotide sequence. Guide nucleic acids, when complexed with an effector protein, may bring the effector protein into proximity of a target nucleic acid. Sufficient conditions for hybridization of a guide nucleic acid to a target nucleic acid and/or for binding of a guide nucleic acid to an effector protein include in vivo physiological conditions of a desired cell type or in vitro conditions sufficient for assaying catalytic activity of a protein, polypeptide or peptide described herein, such as the nuclease activity of an effector protein. Guide nucleic acids may comprise DNA, RNA, or a combination thereof (e.g., RNA with a thymine base). Guide nucleic acids may include a chemically modified nucleobase or phosphate backbone. Guide nucleic acids may be referred to herein as a guide RNA (gRNA). However, a guide RNA is not limited to ribonucleotides, but may comprise deoxyribonucleotides and other chemically modified nucleotides.


A guide nucleic acid may comprise a CRISPR RNA (crRNA), a short-complementarity untranslated RNA (scoutRNA), an associated trans-activating RNA (tracrRNA) or a combination thereof. The combination of a crRNA with a tracrRNA may be referred to herein as a single guide RNA (sgRNA), wherein the crRNA and the tracrRNA are covalently linked. In some embodiments, the crRNA and tracrRNA are linked by a phosphodiester bond. In some instances, the crRNA and tracrRNA are linked by one or more linked nucleotides. A guide nucleic acid may comprise a naturally occurring guide nucleic acid. A guide nucleic acid may comprise a non-naturally occurring guide nucleic acid, including a guide nucleic acid that is designed to contain a chemical or biochemical modification.


Guide nucleic acids and portions thereof may be found in or identified from a CRISPR array present in the genome of a host organism. A crRNA may be the product of processing of a longer precursor CRISPR RNA (pre-crRNA) transcribed from the CRISPR array by cleavage of the pre-crRNA within each direct repeat sequence to afford shorter, mature crRNAs. A crRNA may be generated by a variety of mechanisms, including the use of dedicated endonucleases (e.g., Cas6 or Cas5d in Type I and III systems), coupling of a host endonuclease (e.g., RNase III) with tracrRNA (Type II systems), or a ribonuclease activity endogenous to the effector protein itself (e.g., Cpf1, from Type V systems). A crRNA may also be specifically generated outside of processing of a pre-crRNA and individually contacted to an effector protein in vivo or in vitro.


The CRISPR/Cas system used to detect modified target nucleic acids can comprise CRISPR RNAs (crRNAs), trans-activating crRNAs (tracrRNAs), Cas proteins, and reporters. The term, “CRISPR RNA (crRNA),” as used herein refers to a nucleic acid comprising a first sequence, often referred to as a “spacer sequence,” that hybridizes to a target sequence of a target nucleic acid, and a second sequence that either a) hybridizes to a portion of a tracrRNA or b) is capable of being non-covalently bound by an effector protein. In some embodiments, the crRNA is covalently linked to an additional nucleic acid (e.g., a tracrRNA), wherein the additional nucleic acid interacts with the effector protein.


A guide nucleic acid can comprise a sequence that is reverse complementary to the sequence of a target nucleic acid. A guide nucleic acid can be a crRNA. Sometimes, a guide nucleic acid comprises a crRNA and tracrRNA. The guide nucleic acid can bind specifically to the target nucleic acid. In some cases, the guide nucleic acid is not naturally occurring and made by artificial combination of otherwise separate segments of sequence. Often, the artificial combination is performed by chemical synthesis, by genetic engineering techniques, or by the artificial manipulation of isolated segments of nucleic acids. The target nucleic acid can be designed and made to provide desired functions. In some cases, the targeting region of a guide nucleic acid is 20 nucleotides in length. The targeting region of the guide nucleic acid may have a length of at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length. In some instances, the targeting region of the guide nucleic acid is 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length. In some cases, the targeting region of a guide nucleic acid has a length from exactly or about 12 nucleotides (nt) to about 80 nt, from about 12 nt to about 50 nt, from about 12 nt to about 45 nt, from about 12 nt to about 40 nt, from about 12 nt to about 35 nt, from about 12 nt to about 30 nt, from about 12 nt to about 25 nt, from about 12 nt to about 20 nt, from about 12 nt to about 19 nt, from about 19 nt to about 20 nt, from about 19 nt to about 25 nt, from about 19 nt to about 30 nt, from about 19 nt to about 35 nt, from about 19 nt to about 40 nt, from about 19 nt to about 45 nt, from about 19 nt to about 50 nt, from about 19 nt to about 60 nt, from about 20 nt to about 25 nt, from about 20 nt to about 30 nt, from about 20 nt to about 35 nt, from about 20 nt to about 40 nt, from about 20 nt to about 45 nt, from about 20 nt to about 50 nt, or from about 20 nt to about 60 nt. It is understood that the sequence of a polynucleotide need not be 100% complementary to that of its target nucleic acid to be specifically hybridizable or bind specifically. The guide nucleic acid can have a sequence comprising at least one uracil in a region from nucleic acid residue 5 to 20 that is reverse complementary to a modification variable region in the target nucleic acid. The guide nucleic acid, in some cases, has a sequence comprising at least one uracil in a region from nucleic acid residue 5 to 9, 10 to 14, or 15 to 20 that is reverse complementary to a modification variable region in the target nucleic acid. The guide nucleic acid can have a sequence comprising at least one uracil in a region from nucleic acid residue 5 to 20 that is reverse complementary to a methylation variable region in the target nucleic acid. The guide nucleic acid, in some cases, has a sequence comprising at least one uracil in a region from nucleic acid residue 5 to 9, 10 to 14, or 15 to 20 that is reverse complementary to a methylation variable region in the target nucleic acid.


The guide nucleic acid can be selected from a group of guide nucleic acids that have been tiled against the nucleic acid sequence of a strain of an infection or genomic locus of interest. The guide nucleic acid can be selected from a group of guide nucleic acids that have been tiled against the nucleic acid sequence of a strain of coronavirus. Often, guide nucleic acids that are tiled against the nucleic acid of a strain of an infection or genomic locus of interest can be pooled for use in a method described herein. Often, these guide nucleic acids are pooled for detecting a target nucleic acid in a single assay. The pooling of guide nucleic acids that are tiled against a single target nucleic acid can enhance the detection of the target nucleic using the methods described herein. The pooling of guide nucleic acids that are tiled against a single target nucleic acid can ensure broad coverage of the target nucleic acid within a single reaction using the methods described herein. The tiling, for example, is sequential along the target nucleic acid. Sometimes, the tiling is overlapping along the target nucleic acid. In some instances, the tiling comprises gaps between the tiled guide nucleic acids along the target nucleic acid. In some instances the tiling of the guide nucleic acids is non-sequential. Often, a method for detecting a target nucleic acid comprises contacting a target nucleic acid to a pool of guide nucleic acids and a programmable nuclease, wherein a guide nucleic acid of the pool of guide nucleic acids has a sequence selected from a group of tiled guide nucleic acid that correspond to nucleic acids of a target nucleic acid; and assaying for a signal produce by cleavage of at least some reporters of a population of reporters. Pooling of guide nucleic acids can ensure broad spectrum identification, or broad coverage, of a target species within a single reaction. This can be particularly helpful in diseases or indications, like sepsis, that may be caused by multiple organisms.


Described herein are reagents comprising a programmable nuclease capable of being activated when complexed with the guide nucleic acid and the target nucleic acid segment. A programmable nuclease can be capable of being activated when complexed with a guide nucleic acid and the target sequence. The programmable nuclease can be activated upon binding of the guide nucleic acid to its target nucleic acid and degrades non-specifically nucleic acid in its environment. The programmable nuclease has trans cleavage activity once activated. A programmable nuclease can be a Cas protein (also referred to, interchangeably, as a Cas nuclease). A crRNA and Cas protein can form a CRISPR enzyme.


Trans cleavage activity can be non-specific cleavage of nearby single-stranded nucleic acids by the activated programmable nuclease, such as trans cleavage of reporters with a detection moiety. Once the reporter is cleaved by the activated programmable nuclease, the detection moiety can be released from the reporter and can generate a signal. A signal can be a calorimetric, potentiometric, amperometric, optical (e.g., fluorescent, colorimetric, etc.), or piezo-electric signal. Often, the signal is present prior to reporter cleavage and changes upon reporter cleavage. Sometimes, the signal is absent prior to reporter cleavage and is present upon reporter cleavage. The detection moiety capable of generating the detectable signal can be immobilized on a support medium for detection. The programmable nuclease can be a CRISPR-Cas (clustered regularly interspaced short palindromic repeats—CRISPR associated) nucleoprotein complex with trans cleavage activity, which can be activated by binding of a guide nucleic acid with a target nucleic acid. The CRISPR-Cas nucleoprotein complex can comprise a Cas protein (also referred to as a Cas nuclease) complexed with a guide nucleic acid, which can also be referred to as CRISPR enzyme. A guide nucleic acid can be a CRISPR RNA (crRNA). Sometimes, a guide nucleic acid may comprise a crRNA and a trans-activating crRNA (tracrRNA).


The devices, systems, fluidic devices, kits, and methods for detecting the presence of a target nucleic acid in a sample described herein may comprise a generation of a signal indicative of the presence or absence of the target nucleic acid in the sample. The generation of a signal indicative of the presence or absence of the target nucleic acid in the sample as described herein is compatible with the methods and devices described herein (e.g., pneumatic valve devices, sliding valve devices, rotating valve devices, and lateral flow devices) and may result from the use of compositions disclosed herein (e.g. programmable nucleases, guide nucleic acids, reagents for in vitro transcription, reagents for amplification, reagents for reverse transcription, reporters, or any combination thereof) to carry out highly efficient, rapid, and accurate reactions for detecting whether a target nucleic acid is present in a sample (e.g., DETECTR reactions). As disclosed herein, in some embodiments, detecting the presence or absence of a target nucleic acid of interest involves measuring a signal emitted from a detection moiety present in a reporter, after cleavage of the reporter by an activated programmable nuclease. Alternatively, or in combination, in some embodiments, detecting the presence or absence of a target nucleic acid of interest involves measuring a signal emitted from a conjugate bound to a detection moiety present in a reporter, after cleavage of the reporter by an activated programmable nuclease. The conjugates may comprise a nanoparticle, a gold nanoparticle, a latex nanoparticle, a quantum dot, a chemiluminescent nanoparticle, a carbon nanoparticle, a selenium nanoparticle, a fluorescent nanoparticle, a liposome, or a dendrimer. The surface of the conjugate may be coated by a conjugate binding molecule that binds to the detection moiety or another affinity molecule of the cleaved detector molecule as described herein. Thus, the detecting steps disclosed herein involve indirectly (e.g., via a reporter) measuring the presence of a target nucleic acid, quantifying how much of the target nucleic acid is present, or, measuring a signal indicating that the target nucleic acid is absent in a sample. In some embodiments, a signal is generated upon cleavage of the reporter by the programmable nuclease. In other embodiments, the signal changes upon cleavage of the reporter by the programmable nuclease. In other embodiments, a signal may be present in the absence of reporter cleavage and disappear upon cleavage of the target nucleic acid by the programmable nuclease. For example, a signal may be produced in a microfluidic device or lateral flow device after contacting a sample with a composition comprising a programmable nuclease.


“Percent identity” and “% identity” can refer to the extent to which two sequences (nucleotide or amino acid) have the same residue at the same positions in an alignment. For example, “an amino acid sequence is X % identical to SEQ ID NO: Y” can refer to % identity of the amino acid sequence to SEQ ID NO: Y and is elaborated as X % of residues in the amino acid sequence are identical to the residues of sequence disclosed in SEQ ID NO: Y. Generally, computer programs can be employed for such calculations. Illustrative programs that compare and align pairs of sequences, include ALIGN (Myers and Miller, Comput Appl Biosci. 1988 March; 4(1): 11-7), FASTA (Pearson and Lipman, Proc Natl Acad Sci USA. 1988 April; 85(8):2444-8; Pearson, Methods Enzymol. 1990; 183:63-98) and gapped BLAST (Altschul et al., Nucleic Acids Res. 1997 Sep. 1; 25(17):3389-40), BLASTP, BLASTN, or GCG (Devereux et al., Nucleic Acids Res. 1984 Jan. 11; 12 (1 Pt 1):387-95).


An effector protein may be brought into proximity of a target nucleic acid in the presence of a guide nucleic acid when the guide nucleic acid includes a nucleotide sequence that is complementary with a target sequence in the target nucleic acid. The ability of an effector protein to detect a target nucleic acid and cleave a reporter may be dependent upon the effector protein being bound to a guide nucleic acid and the guide nucleic acid being hybridized to a target nucleic acid. An effector protein may also recognize a protospacer adjacent motif (PAM) sequence present in the target nucleic acid, which may direct the modification activity of the effector protein. An effector protein may modify a nucleic acid by cis cleavage or trans cleavage. An effector protein may be a CRISPR-associated (“Cas”) protein. An effector protein may function as a single protein, including a single protein that is capable of binding to a guide nucleic acid and modifying a target nucleic acid. Alternatively, an effector protein may function as part of a multiprotein complex, including, for example, a complex having two or more effector proteins, including two or more of the same effector proteins (e.g., dimer or multimer). An effector protein, when functioning in a multiprotein complex, may have only one functional activity (e.g., binding to a guide nucleic acid), while other effector proteins present in the multiprotein complex are capable of the other functional activity (e.g., modifying a target nucleic acid). An effector protein may be a modified effector protein having reduced modification activity (e.g., a catalytically defective effector protein) or no modification activity (e.g., a catalytically inactive effector protein). Accordingly, an effector protein as used herein encompasses a modified or programmable nuclease that does not have nuclease activity.


Several programmable nucleases are consistent with the methods and devices of the present disclosure. For example, CRISPR/Cas enzymes are programmable nucleases used in the methods and systems disclosed herein. CRISPR/Cas enzymes can include any of the known Classes and Types of CRISPR/Cas enzymes. Programmable nucleases disclosed herein include Class 1 CRISPR/Cas enzymes, such as the Type I, Type IV, or Type III CRISPR/Cas enzymes. Programmable nucleases disclosed herein also include the Class 2 CRISPR/Cas enzymes, such as the Type II, Type V, and Type VI CRISPR/Cas enzymes. Preferable programmable nucleases included in the several assays disclosed herein (e.g., for assaying for coronavirus in a device, such as a microfluidic device or a lateral flow assay) and methods of use thereof include a Type V or Type VI CRISPR/Cas enzyme.


The programmable nuclease system used to detect modified target nucleic acids can comprise CRISPR RNAs (crRNAs), trans-activating crRNAs (tracrRNAs), Cas proteins, and reporters.


Described herein are reagents comprising a programmable nuclease capable of being activated when complexed with the guide nucleic acid and the target nucleic acid segment or portion. A programmable nuclease can be capable of being activated when complexed with a guide nucleic acid and the target sequence. The programmable nuclease can be activated upon binding of the guide nucleic acid to its target nucleic acid and degrades non-specifically nucleic acid in its environment. The programmable nuclease has trans cleavage activity once activated. A programmable nuclease can be a Cas protein (also referred to, interchangeably, as a Cas nuclease). A crRNA and Cas protein can form a CRISPR enzyme.


Several programmable nucleases are consistent with the methods and devices of the present disclosure. For example, CRISPR/Cas enzymes are programmable nucleases used in the methods and systems disclosed herein.


The systems and methods of the present disclosure can be implemented using a device that is compatible with a plurality of programmable nucleases. The device can comprise a plurality of programmable nucleases and one or more corresponding guide nucleic acids. In some cases, the plurality of programmable nucleases are complexed with one or more corresponding guide nucleic acids. The plurality of programmable nucleases (and guide nucleic acids) can be the same. Alternatively, the plurality of programmable nuclease complexes can be different. For example, the plurality of programmable nuclease complexes can comprise different programmable nucleases and/or different guide nucleic acids associated with the programmable nucleases.


As used herein, a programmable nuclease generally refers to any enzyme that can cleave nucleic acid. The programmable nuclease can be any enzyme that can be or has been designed, modified, or engineered by human contribution so that the enzyme targets or cleaves the nucleic acid in a sequence-specific manner. Programmable nucleases can include, for example, zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and/or RNA-guided nucleases such as the bacterial clustered regularly interspaced short palindromic repeat (CRISPR)-Cas (CRISPR-associated) nucleases or Cpf1. Programmable nucleases can also include, for example, PfAgo and/or NgAgo.


ZFNs can cut genetic material in a sequence-specific matter and can be designed, or programmed, to target specific viral targets. A ZFN is composed of two domains: a DNA-binding zinc-finger protein linked to the Fokl nuclease domain. The DNA-binding zinc-finger protein is fused with the non-specific Fokl cleave domain to create ZFNs. The protein will typically dimerize for activity. Two ZFN monomers form an active nuclease; each monomer binds to adjacent half-sites on the target. The sequence specificity of ZFNs is determined by ZFPs. Each zinc-finger recognizes a 3-bp DNA sequence, and 3-6 zinc-fingers are used to generate a single ZFN subunit that binds to DNA sequences of 9-18 bp. The DNA-binding specificities of zinc-fingers is altered by mutagenesis. New ZFPs are programmed by modular assembly of pre-characterized zinc fingers.


Transcription activator-like effector nucleases (TALENs) can cut genetic material in a sequence-specific matter and can be designed, or programmed, to target specific viral targets. TALENs contain the Fokl nuclease domain at their carboxyl termini and a class of DNA binding domains known as transcription activator-like effectors (TALEs). TALENs are composed of tandem arrays of 33-35 amino acid repeats, each of which recognizes a single base-pair in the major groove of target viral DNA. The nucleotide specificity of a domain comes from the two amino acids at positions 12 and 13 where Asn-Asn, Asn-Ile, His-Asp and Asn-Gly recognize guanine, adenine, cytosine and thymine, respectively. That pattern allows one to program TALENs to target various nucleic acids.


The programmable nuclease can comprise any type of human engineered enzymes. Alternatively, the programmable nuclease can comprise CRISPR enzymes derived from naturally occurring bacteria or phage. A programmable nuclease can be a Cas protein (also referred to, interchangeably, as a Cas nuclease). A crRNA and Cas protein can form a CRISPR enzyme. The programmable nuclease can be a CRISPR-Cas (clustered regularly interspaced short palindromic repeats—CRISPR associated) nucleoprotein complex with trans cleavage activity, which can be activated by binding of a guide nucleic acid with a target nucleic acid. The programmable nuclease can comprise one or more amino acid modifications. The programmable nuclease be a nuclease derived from a CRISPR-Cas system. The programmable nuclease can be a nuclease derived from recombineering.


CRISPR/Cas enzymes can include any of the known Classes and Types of CRISPR/Cas enzymes. Programmable nucleases disclosed herein include Class 1 CRISPR/Cas enzymes, such as the Type I, Type IV, or Type III CRISPR/Cas enzymes. Programmable nucleases disclosed herein also include the Class 2 CRISPR/Cas enzymes, such as the Type II, Type V, and Type VI CRISPR/Cas enzymes. Preferable programmable nucleases included in the several devices disclosed herein (e.g., a microfluidic device such as a pneumatic valve device or a sliding valve device or a lateral flow assay) and methods of use thereof include a Type V or Type VI CRISPR/Cas enzyme.


In some embodiments, the Type V CRISPR/Cas enzyme is a programmable Cas12 nuclease. Type V CRISPR/Cas enzymes (e.g., Cas12 or Cas14) lack an HNH domain. A Cas12 nuclease of the present disclosure cleaves a nucleic acid via a single catalytic RuvC domain. The RuvC domain is within a nuclease, or “NUC” lobe of the protein, and the Cas12 nucleases further comprise a recognition, or “REC” lobe. The REC and NUC lobes are connected by a bridge helix and the Cas12 proteins additionally include two domains for PAM recognition termed the PAM interacting (PI) domain and the wedge (WED) domain. In some instances, a programmable Cas12 nuclease can be a Cas12a (also referred to as Cpf1) protein, a Cas12b protein, Cas12c protein, Cas12d protein, or a Cas12e protein.


In some embodiments, the programmable nuclease can be Cas13. Sometimes the Cas13 can be Cas13a, Cas13b, Cas13c, Cas13d, or Cas13e. In some cases, the programmable nuclease can be Mad7 or Mad2. In some cases, the programmable nuclease can be Cas12. Sometimes the Cas12 can be Cas12a, Cas12b, Cas12c, Cas12d, or Cas12e. In some cases, the Cas12 can be a Cas12 variant (SEQ ID NO: 28), which is a specific protein variant within the Cas12 protein family/classification). In some cases, the programmable nuclease can be Csm1, Cas9, C2c4, C2c8, C2c5, C2c10, C2c9, or CasZ. Sometimes, the Csm1 can also be also called smCms1, miCms1, obCms1, or suCms1. Sometimes Cas13a can also be also called C2c2. Sometimes CasZ can also be called Cas14a, Cas14b, Cas14c, Cas14d, Cas14e, Cas14f, Cas14g, or Cas14h. Sometimes, the programmable nuclease can be a type V CRISPR-Cas system. In some cases, the programmable nuclease can be a type VI CRISPR-Cas system. Sometimes the programmable nuclease can be a type III CRISPR-Cas system. Sometimes the programmable nuclease can be an engineered nuclease that is not from a naturally occurring CRISPR-Cas system. In some cases, the programmable nuclease can be from at least one of Leptotrichia shahii (Lsh), Listeria seeligeri (Lse), Leptotrichia buccalis (Lbu), Leptotrichia wadeu (Lwa), Rhodobacter capsulatus (Rca), Herbinix hemicellulosilytica (Hhe), Paludibacter propionicigenes (Ppr), Lachnospiraceae bacterium (Lba), [Eubacterium] rectale (Ere), Listeria newyorkensis (Lny), Clostridium aminophilum (Cam), Prevotella sp. (Psm), Capnocytophaga canimorsus (Cca, Lachnospiraceae bacterium (Lba), Bergeyella zoohelcum (Bzo), Prevotella intermedia (Pin), Prevotella buccae (Pbu), Alistipes sp. (Asp), Riemerella anatipestifer (Ran), Prevotella aurantiaca (Pau), Prevotella saccharolytica (Psa), Prevotella intermedia (Pin2), Capnocytophaga canimorsus (Cca), Porphyromonas gulae (Pgu), Prevotella sp. (Psp), Porphyromonas gingivalis (Pig), Prevotella intermedia (Pin3), Enterococcus italicus (Ei), Lactobacillus salivarius (Ls), or Thermus thermophilus (Tt). Sometimes the Cas13 is at least one of LbuCas13a, LwaCas13a, LbaCas13a, HheCas13a, PprCas13a, EreCas13a, CamCas13a, or LshCas13a. The trans cleavage activity of the CRISPR enzyme can be activated when the crRNA is complexed with the target nucleic acid. The trans cleavage activity of the CRISPR enzyme can be activated when the guide nucleic acid comprising a tracrRNA and crRNA are complexed with the target nucleic acid. The target nucleic acid can be RNA or DNA.


In some embodiments, a programmable nuclease as disclosed herein is an RNA-activated programmable RNA nuclease. In some embodiments, a programmable nuclease as disclosed herein is a DNA-activated programmable RNA nuclease. In some embodiments, a programmable nuclease is capable of being activated by a target RNA to initiate trans cleavage of an RNA reporter and is capable of being activated by a target DNA to initiate trans cleavage of an RNA reporter, such as a Type VI CRISPR/Cas enzyme (e.g., a Cas13 nuclease). For example, Cas13a of the present disclosure can be activated by a target RNA to initiate trans cleavage activity of the Cas13a for the cleavage of an RNA reporter and can be activated by a target DNA to initiate trans cleavage activity of the Cas13a for trans cleavage of an RNA reporter. An RNA reporter can be an RNA-based reporter. In some embodiments, the Cas13a recognizes and detects ssDNA to initiate transcleavage of RNA reporters. Multiple Cas13a isolates can recognize, be activated by, and detect target DNA, including ssDNA, upon hybridization of a guide nucleic acid with the target DNA. For example, Lbu-Cas13a and Lwa-Cas13a can both be activated to transcollaterally cleave RNA reporters by target DNA. Thus, Type VI CRISPR/Cas enzyme (e.g., a Cas13 nuclease, such as Cas13a) can be DNA-activated programmable RNA nucleases, and therefore can be used to detect a target DNA using the methods as described herein. DNA-activated programmable RNA nuclease detection of ssDNA can be robust at multiple pH values. For example, target ssDNA detection by Cas13 can exhibit consistent cleavage across a wide range of pH conditions, such as from a pH of 6.8 to a pH of 8.2. In contrast, target RNA detection by Cas13 can exhibit high cleavage activity of pH values from 7.9 to 8.2. In some embodiments, a DNA-activated programmable RNA nuclease that also is capable of being an RNA-activated programmable RNA nuclease, can have DNA targeting preferences that are distinct from its RNA targeting preferences. For example, the optimal ssDNA targets for Cas13a have different properties than optimal RNA targets for Cas13a. As one example, gRNA performance on ssDNA can not necessarily correlate with the performance of the same gRNAs on RNA. As another example, gRNAs can perform at a high level regardless of target nucleotide identity at a 3′ position on a target RNA sequence. In some embodiments, gRNAs can perform at a high level in the absence of a G at a 3′ position on a target ssDNA sequence. Furthermore, target DNA detected by Cas13 disclosed herein can be directly taken from organisms or can be indirectly generated by nucleic acid amplification methods, such as PCR and LAMP or any amplification method described herein. Key steps for the sensitive detection of a target DNA, such as a target ssDNA, by a DNA-activated programmable RNA nuclease, such as Cas13a, can include: (1) production or isolation of DNA to concentrations above about 0.1 nM per reaction for in vitro diagnostics, (2) selection of a target sequence with the appropriate sequence features to enable DNA detection as these features are distinct from those required for RNA detection, and (3) buffer composition that enhances DNA detection.


The detection of a target DNA by a DNA-activated programmable RNA nuclease can be connected to a variety of readouts including fluorescence, lateral flow, electrochemistry, or any other readouts described herein. Multiplexing of programmable DNA nuclease, such as a Type V CRISPR-Cas protein, with a DNA-activated programmable RNA nuclease, such as a Type VI protein, with a DNA reporter and an RNA reporter, can enable multiplexed detection of target ssDNAs or a combination of a target dsDNA and a target ssDNA, respectively. Multiplexing of different RNA-activated programmable RNA nucleases that have distinct RNA reporter cleavage preferences can enable additional multiplexing. Methods for the generation of ssDNA for DNA-activated programmable RNA nuclease-based diagnostics can include (1) asymmetric PCR, (2) asymmetric isothermal amplification, such as RPA, LAMP, SDA, etc. (3) NEAR for the production of short ssDNA molecules, and (4) conversion of RNA targets into ssDNA by a reverse transcriptase followed by RNase H digestion. Thus, DNA-activated programmable RNA nuclease detection of target DNA is compatible with the various systems, kits, compositions, reagents, and methods disclosed herein.


The programmable nuclease can comprise a programmable nuclease capable of being activated when complexed with a guide nucleic acid and target nucleic acid. The programmable nuclease can become activated after binding of a guide nucleic acid with a target nucleic acid, in which the activated programmable nuclease can cleave the target nucleic acid, which can initiate trans cleavage activity. In some cases, the trans cut or trans cleavage can cut and/or release a reporter. In other cases, the trans cut or trans cleavage can produce an analog of a target, which can be directly detected. Trans cleavage activity can be non-specific cleavage of nearby nucleic acids by the activated programmable nuclease, such as trans cleavage of reporters with a detection moiety. Once the reporter is cleaved by the activated programmable nuclease, the detection moiety can be released from the reporter and can generate a signal.


In some embodiments, the Type V CRISPR/Cas enzyme is a programmable Cas12 nuclease. Type V CRISPR/Cas enzymes (e.g., Cas12 or Cas14) lack an HNH domain. A Cas12 nuclease of the present disclosure cleaves a nucleic acids via a single catalytic RuvC domain. The RuvC domain is within a nuclease, or “NUC” lobe of the protein, and the Cas12 nucleases further comprise a recognition, or “REC” lobe. The REC and NUC lobes are connected by a bridge helix and the Cas12 proteins additionally include two domains for PAM recognition termed the PAM interacting (PI) domain and the wedge (WED) domain. (Murugan et al., Mol Cell. 2017 Oct. 5; 68(1): 15-25). A programmable Cas12 nuclease can be a Cas12a (also referred to as Cpf1) protein, a Cas12b protein, Cas12c protein, Cas12d protein, or a Cas12e protein. In some cases, a suitable Cas12 protein comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to any one of SEQ ID NO: 18-SEQ ID NO: 60.









TABLE 1







Cas12 Protein Sequences









SEQ




ID




NO
Description
Sequence





SEQ

Lachnospiraceae

MSKLEKFTNCYSLSKTLRFKAIPVGKTQENIDNKRLLVEDEKRAEDY


ID

bacterium

KGVKKLLDRYYLSFINDVLHSIKLKNLNNYISLFRKKTRTEKENKEL


NO:
ND2006
ENLEINLRKEIAKAFKGNEGYKSLFKKDIIETILPEFLDDKDEIALVNS


18
(LbCas12a)
FNGFTTAFTGFFDNRENMFSEEAKSTSIAFRCINENLTRYISNMDIFEK




VDAIFDKHEVQEIKEKILNSDYDVEDFFEGEFFNFVLTQEGIDVYNAII




GGFVTESGEKIKGLNEYINLYNQKTKQKLPKFKPLYKQVLSDRESLS




FYGEGYTSDEEVLEVFRNTLNKNSEIFSSIKKLEKLFKNFDEYSSAGIF




VKNGPAISTISKDIFGEWNVIRDKWNAEYDDIHLKKKAVVTEKYED




DRRKSFKKIGSFSLEQLQEYADADLSVVEKLKEIIIQKVDEIYKVYGS




SEKLFDADFVLEKSLKKNDAVVAIMKDLLDSVKSFENYIKAFFGEGK




ETNRDESFYGDFVLAYDILLKVDHIYDAIRNYVTQKPYSKDKFKLYF




QNPQFMGGWDKDKETDYRATILRYGSKYYLAIMDKKYAKCLQKID




KDDVNGNYEKINYKLLPGPNKMLPKVFFSKKWMAYYNPSEDIQKIY




KNGTFKKGDMFNLNDCHKLIDFFKDSISRYPKWSNAYDFNFSETEK




YKDIAGFYREVEEQGYKVSFESASKKEVDKLVEEGKLYMFQIYNKD




FSDKSHGTPNLHTMYFKLLFDENNHGQIRLSGGAELFMRRASLKKEE




LVVHPANSPIANKNPDNPKKTTTLSYDVYKDKRFSEDQYELHIPIAIN




KCPKNIFKINTEVRVLLKHDDNPYVIGIDRGERNLLYIVVVDGKGNIV




EQYSLNEIINNFNGIRIKTDYHSLLDKKEKERFEARQNWTSIENIKELK




AGYISQVVHKICELVEKYDAVIALEDLNSGFKNSRVKVEKQVYQKF




EKMLIDKLNYMVDKKSNPCATGGALKGYQITNKFESFKSMSTQNGF




IFYIPAWLTSKIDPSTGFVNLLKTKYTSIADSKKFISSFDRIMYVPEEDL




FEFALDYKNFSRTDADYIKKWKLYSYGNRIRIFRNPKKNNVFDWEE




VCLTSAYKELFNKYGINYQQGDIRALLCEQSDKAFYSSFMALMSLM




LQMRNSITGRTDVDFLISPVKNSDGIFYDSRNYEAQENAILPKNADA




NGAYNIARKVLWAIGQFKKAEDEKLDKVKIAISNKEWLEYAQTSVK




H





SEQ

Acidaminococcus

MTQFEGFTNLYQVSKTLRFELIPQGKTLKHIQEQGFIEEDKARNDHY


ID
sp.
KELKPIIDRIYKTYADQCLQLVQLDWENLSAAIDSYRKEKTEETRNA


NO:
BV316
LIEEQATYRNAIHDYFIGRTDNLTDAINKRHAEIYKGLFKAELFNGKV


19
(AsCas12a)
LKQLGTVTTTEHENALLRSFDKFTTYFSGFYENRKNVFSAEDISTAIP




HRIVQDNFPKFKENCHIFTRLITAVPSLREHFENVKKAIGIFVSTSIEEV




FSFPFYNQLLTQTQIDLYNQLLGGISREAGTEKIKGLNEVLNLAIQKN




DETAHIIASLPHRFIPLFKQILSDRNTLSFILEEFKSDEEVIQSFCKYKTL




LRNENVLETAEALFNELNSIDLTHIFISHKKLETISSALCDHWDTLRN




ALYERRISELTGKITKSAKEKVQRSLKHEDINLQEIISAAGKELSEAFK




QKTSEILSHAHAALDQPLPTTLKKQEEKEILKSQLDSLLGLYHLLDW




FAVDESNEVDPEFSARLTGIKLEMEPSLSFYNKARNYATKKPYSVEK




FKLNFQMPTLASGWDVNKEKNNGAILFVKNGLYYLGIMPKQKGRY




KALSFEPTEKTSEGFDKMYYDYFPDAAKMIPKCSTQLKAVTAHFQT




HTTPILLSNNFIEPLEITKEIYDLNNPEKEPKKFQTAYAKKTGDQKGY




REALCKWIDFTRDFLSKYTKTTSIDLSSLRPSSQYKDLGEYYAELNPL




LYHISFQRIAEKEIMDAVETGKLYLFQIYNKDFAKGHHGKPNLHTLY




WTGLFSPENLAKTSIKLNGQAELFYRPKSRMKRMAHRLGEKMLNK




KLKDQKTPIPDTLYQELYDYVNHRLSHDLSDEARALLPNVITKEVSH




EIIKDRRFTSDKFFFHVPITLNYQAANSPSKFNQRVNAYLKEHPETPII




GIDRGERNLIYITVIDSTGKILEQRSLNTIQQFDYQKKLDNREKERVA




ARQAWSVVGTIKDLKQGYLSQVIHEIVDLMIHYQAVVVLENLNFGF




KSKRTGIAEKAVYQQFEKMLIDKLNCLVLKDYPAEKVGGVLNPYQL




TDQFTSFAKMGTQSGFLFYVPAPYTSKIDPLTGFVDPFVWKTIKNHE




SRKHFLEGFDFLHYDVKTGDFILHFKMNRNLSFQRGLPGFMPAWDI




VFEKNETQFDAKGTPFIAGKRIVPVIENHRFTGRYRDLYPANELIALL




EEKGIVFRDGSNILPKLLENDDSHAIDTMVALIRSVLQMRNSNAATG




EDYINSPVRDLNGVCFDSRFQNPEWPMDADANGAYHIALKGQLLLN




HLKESKDLKLQNGISNQDWLAYIQELRN





SEQ

Francisella

MSIYQEFVNKYSLSKTLRFELIPQGKTLENIKARGLILDDEKRAKDYK


ID

novicida

KAKQIIDKYHQFFIEEILSSVCISEDLLQNYSDVYFKLKKSDDDNLQK


NO:
U112
DFKSAKDTIKKQISEYIKDSEKFKNLFNQNLIDAKKGQESDLILWLKQ


20
(FnCas12a)
SKDNGIELFKANSDITDIDEALEIIKSFKGWTTYFKGFHENRKNVYSS




NDIPTSIIYRIVDDNLPKFLENKAKYESLKDKAPEAINYEQIKKDLAEE




LTFDIDYKTSEVNQRVFSLDEVFEIANFNNYLNQSGITKFNTIIGGKFV




NGENTKRKGINEYINLYSQQINDKTLKKYKMSVLFKQILSDTESKSF




VIDKLEDDSDVVTTMQSFYEQIAAFKTVEEKSIKETLSLLFDDLKAQ




KLDLSKIYFKNDKSLTDLSQQVFDDYSVIGTAVLEYITQQIAPKNLDN




PSKKEQELIAKKTEKAKYLSLETIKLALEEFNKHRDIDKQCRFEEILA




NFAAIPMIFDEIAQNKDNLAQISIKYQNQGKKDLLQASAEDDVKAIK




DLLDQTNNLLHKLKIFHISQSEDKANILDKDEHFYLVFEECYFELANI




VPLYNKIRNYITQKPYSDEKFKLNFENSTLANGWDKNKEPDNTAILFI




KDDKYYLGVMNKKNNKIFDDKAIKENKGEGYKKIVYKLLPGANKM




LPKVFFSAKSIKFYNPSEDILRIRNHSTHTKNGSPQKGYEKFEFNIEDC




RKFIDFYKQSISKHPEWKDFGFRFSDTQRYNSIDEFYREVENQGYKLT




FENISESYIDSVVNQGKLYLFQIYNKDFSAYSKGRPNLHTLYWKALF




DERNLQDVVYKLNGEAELFYRKQSIPKKITHPAKEAIANKNKDNPK




KESVFEYDLIKDKRFTEDKFFFHCPITINFKSSGANKFNDEINLLLKEK




ANDVHILSIDRGERHLAYYTLVDGKGNIIKQDTFNIIGNDRMKTNYH




DKLAAIEKDRDSARKDWKKINNIKEMKEGYLSQVVHEIAKLVIEYN




AIVVFEDLNFGFKRGRFKVEKQVYQKLEKMLIEKLNYLVFKDNEFD




KTGGVLRAYQLTAPFETFKKMGKQTGIIYYVPAGFTSKICPVTGFVN




QLYPKYESVSKSQEFFSKFDKICYNLDKGYFEFSFDYKNFGDKAAKG




KWTIASFGSRLINFRNSDKNHNWDTREVYPTKELEKLLKDYSIEYGH




GECIKAAICGESDKKFFAKLTSVLNTILQMRNSKTGTELDYLISPVAD




VNGNFFDSRQAPKNMPQDADANGAYHIGLKGLMLLGRIKNNQEGK




KLNLVIKNEEYFEFVQNRNN





SEQ

Porphyromonas

MKTQHFFEDFTSLYSLSKTIRFELKPIGKTLENIKKNGLIRRDEQRLDD


ID

macacae

YEKLKKVIDEYHEDFIANILSSFSFSEEILQSYIQNLSESEARAKIEKTM


NO:
(PmCas12a)
RDTLAKAFSEDERYKSIFKKELVKKDIPVWCPAYKSLCKKFDNFTTS


21

LVPFHENRKNLYTSNEITASIPYRIVHVNLPKFIQNIEALCELQKKMG




ADLYLEMMENLRNVWPSFVKTPDDLCNLKTYNHLMVQSSISEYNRF




VGGYSTEDGTKHQGINEWINIYRQRNKEMRLPGLVFLHKQILAKVD




SSSFISDTLENDDQVFCVLRQFRKLFWNTVSSKEDDAASLKDLFCGL




SGYDPEAIYVSDAHLATISKNIFDRWNYISDAIRRKTEVLMPRKKESV




ERYAEKISKQIKKRQSYSLAELDDLLAHYSEESLPAGFSLLSYFTSLG




GQKYLVSDGEVILYEEGSNIWDEVLIAFRDLQVILDKDFTEKKLGKD




EEAVSVIKKALDSALRLRKFFDLLSGTGAEIRRDSSFYALYTDRMDK




LKGLLKMYDKVRNYLTKKPYSIEKFKLHFDNPSLLSGWDKNKELNN




LSVIFRQNGYYYLGIMTPKGKNLFKTLPKLGAEEMFYEKMEYKQIAE




PMLMLPKVFFPKKTKPAFAPDQSVVDIYNKKTFKTGQKGFNKKDLY




RLIDFYKEALTVHEWKLFNFSFSPTEQYRNIGEFFDEVREQAYKVSM




VNVPASYIDEAVENGKLYLFQIYNKDFSPYSKGIPNLHTLYWKALFS




EQNQSRVYKLCGGGELFYRKASLHMQDTTVHPKGISIHKKNLNKKG




ETSLFNYDLVKDKRFTEDKFFFHVPISINYKNKKITNVNQMVRDYIA




QNDDLQIIGIDRGERNLLYISRIDTRGNLLEQFSLNVIESDKGDLRTDY




QKILGDREQERLRRRQEWKSIESIKDLKDGYMSQVVHKICNMVVEH




KAIVVLENLNLSFMKGRKKVEKSVYEKFERMLVDKLNYLVVDKKN




LSNEPGGLYAAYQLTNPLFSFEELHRYPQSGILFFVDPWNTSLTDPST




GFVNLLGRINYTNVGDARKFFDRFNAIRYDGKGNILFDLDLSRFDVR




VETQRKLWTLTTFGSRIAKSKKSGKWMVERIENLSLCFLELFEQFNIG




YRVEKDLKKAILSQDRKEFYVRLIYLFNLMMQIRNSDGEEDYILSPA




LNEKNLQFDSRLIEAKDLPVDADANGAYNVARKGLMVVQRIKRGD




HESIHRIGRAQWLRYVQEGIVE





SEQ

Moraxella

MLFQDFTHLYPLSKTVRFELKPIDRTLEHIHAKNFLSQDETMADMHQ


ID

bovoculi

KVKVILDDYHRDFIADMMGEVKLTKLAEFYDVYLKFRKNPKDDEL


NO:
237
QKQLKDLQAVLRKEIVKPIGNGGKYKAGYDRLFGAKLFKDGKELG


22
(MbCas12a)
DLAKFVIAQEGESSPKLAHLAHFEKFSTYFTGFHDNRKNMYSDEDK




HTAIAYRLIHENLPRFIDNLQILTTIKQKHSALYDQIINELTASGLDVSL




ASHLDGYHKLLTQEGITAYNTLLGGISGEAGSPKIQGINELINSHHNQ




HCHKSERIAKLRPLHKQILSDGMSVSFLPSKFADDSEMCQAVNEFYR




HYADVFAKVQSLFDGFDDHQKDGIYVEHKNLNELSKQAFGDFALLG




RVLDGYYVDVVNPEFNERFAKAKTDNAKAKLTKEKDKFIKGVHSL




ASLEQAIEHYTARHDDESVQAGKLGQYFKHGLAGVDNPIQKIHNNH




STIKGFLERERPAGERALPKIKSGKNPEMTQLRQLKELLDNALNVAH




FAKLLTTKTTLDNQDGNFYGEFGVLYDELAKIPTLYNKVRDYLSQK




PFSTEKYKLNFGNPTLLNGWDLNKEKDNFGVILQKDGCYYLALLDK




AHKKVFDNAPNTGKSIYQKMIYKYLEVRKQFPKVFFSKEAIAINYHP




SKELVEIKDKGRQRSDDERLKLYRFILECLKIHPKYDKKFEGAIGDIQ




LFKKDKKGREVPISEKDLFDKINGIFSSKPKLEMEDFFIGEFKRYNPSQ




DLVDQYNIYKKIDSNDNRKKENFYNNHPKFKKDLVRYYYESMCKH




EEWEESFEFSKKLQDIGCYVDVNELFTEIETRRLNYKISFCNINADYID




ELVEQGQLYLFQIYNKDFSPKAHGKPNLHTLYFKALFSEDNLADPIY




KLNGEAQIFYRKASLDMNETTIHRAGEVLENKNPDNPKKRQFVYDII




KDKRYTQDKFMLHVPITMNFGVQGMTIKEFNKKVNQSIQQYDEVN




VIGIDRGERHLLYLTVINSKGEILEQCSLNDITTASANGTQMTTPYHKI




LDKREIERLNARVGWGEIETIKELKSGYLSHVVHQISQLMLKYNAIV




VLEDLNFGFKRGRFKVEKQIYQNFENALIKKLNHLVLKDKADDEIGS




YKNALQLTNNFTDLKSIGKQTGFLFYVPAWNTSKIDPETGFVDLLKP




RYENIAQSQAFFGKFDKICYNADKDYFEFHIDYAKFTDKAKNSRQIW




TICSHGDKRYVYDKTANQNKGAAKGINVNDELKSLFARHHINEKQP




NLVMDICQNNDKEFHKSLMYLLKTLLALRYSNASSDEDFILSPVAND




EGVFFNSALADDTQPQNADANGAYHIALKGLWLLNELKNSDDLNK




VKLAIDNQTWLNFAQNR





SEQ

Moraxella

MGIHGVPAALFQDFTHLYPLSKTVRFELKPIGRTLEHIHAKNFLSQDE


ID

bovoculi

TMADMYQKVKVILDDYHRDFIADMMGEVKLTKLAEFYDVYLKFRK


NO:
AAX08_00205
NPKDDGLQKQLKDLQAVLRKESVKPIGSGGKYKTGYDRLFGAKLFK


23
(Mb2Cas12a)
DGKELGDLAKFVIAQEGESSPKLAHLAHFEKFSTYFTGFHDNRKNM




YSDEDKHTAIAYRLIHENLPRFIDNLQILTTIKQKHSALYDQIINELTA




SGLDVSLASHLDGYHKLLTQEGITAYNRIIGEVNGYTNKHNQICHKS




ERIAKLRPLHKQILSDGMGVSFLPSKFADDSEMCQAVNEFYRHYTDV




FAKVQSLFDGFDDHQKDGIYVEHKNLNELSKQAFGDFALLGRVLDG




YYVDVVNPEFNERFAKAKTDNAKAKLTKEKDKFIKGVHSLASLEQA




IEHHTARHDDESVQAGKLGQYFKHGLAGVDNPIQKIHNNHSTIKGFL




ERERPAGERALPKIKSGKNPEMTQLRQLKELLDNALNVAHFAKLLTT




KTTLDNQDGNFYGEFGVLYDELAKIPTLYNKVRDYLSQKPFSTEKY




KLNFGNPTLLNGWDLNKEKDNFGVILQKDGCYYLALLDKAHKKVF




DNAPNTGKNVYQKMVYKLLPGPNKMLPKVFFAKSNLDYYNPSAEL




LDKYAKGTHKKGDNFNLKDCHALIDFFKAGINKHPEWQHFGFKFSP




TSSYRDLSDFYREVEPQGYQVKFVDINADYIDELVEQGKLYLFQIYN




KDFSPKAHGKPNLHTLYFKALFSEDNLADPIYKLNGEAQIFYRKASL




DMNETTIHRAGEVLENKNPDNPKKRQFVYDIIKDKRYTQDKFMLHV




PITMNFGVQGMTIKEFNKKVNQSIQQYDEVNVIGIDRGERHLLYLTVI




NSKGEILEQRSLNDITTASANGTQVTTPYHKILDKREIERLNARVGW




GEIETIKELKSGYLSHVVHQINQLMLKYNAIVVLEDLNFGFKRGRFK




VEKQIYQNFENALIKKLNHLVLKDKADDEIGSYKNALQLTNNFTDL




KSIGKQTGFLFYVPAWNTSKIDPETGFVDLLKPRYENIAQSQAFFGKF




DKICYNTDKGYFEFHIDYAKFTDKAKNSRQKWAICSHGDKRYVYDK




TANQNKGAAKGINVNDELKSLFARYHINDKQPNLVMDICQNNDKEF




HKSLMCLLKTLLALRYSNASSDEDFILSPVANDEGVFFNSALADDTQ




PQNADANGAYHIALKGLWLLNELKNSDDLNKVKLAIDNQTWLNFA




QNR





SEQ

Moraxella

MGIHGVPAALFQDFTHLYPLSKTVRFELKPIGKTLEHIHAKNFLNQD


ID

bovoculi

ETMADMYQKVKAILDDYHRDFIADMMGEVKLTKLAEFYDVYLKFR


NO:
AAX11_00205
KNPKDDGLQKQLKDLQAVLRKEIVKPIGNGGKYKAGYDRLFGAKLF


24
(Mb3Cas12a)
KDGKELGDLAKFVIAQEGESSPKLAHLAHFEKFSTYFTGFHDNRKN




MYSDEDKHTAIAYRLIHENLPRFIDNLQILATIKQKHSALYDQIINELT




ASGLDVSLASHLDGYHKLLTQEGITAYNTLLGGISGEAGSRKIQGINE




LINSHHNQHCHKSERIAKLRPLHKQILSDGMGVSFLPSKFADDSEVC




QAVNEFYRHYADVFAKVQSLFDGFDDYQKDGIYVEYKNLNELSKQ




AFGDFALLGRVLDGYYVDVVNPEFNERFAKAKTDNAKAKLTKEKD




KFIKGVHSLASLEQAIEHYTARHDDESVQAGKLGQYFKHGLAGVDN




PIQKIHNNHSTIKGFLERERPAGERALPKIKSDKSPEIRQLKELLDNAL




NVAHFAKLLTTKTTLHNQDGNFYGEFGALYDELAKIATLYNKVRDY




LSQKPFSTEKYKLNFGNPTLLNGWDLNKEKDNFGVILQKDGCYYLA




LLDKAHKKVFDNAPNTGKSVYQKMIYKLLPGPNKMLPKVFFAKSNL




DYYNPSAELLDKYAQGTHKKGDNFNLKDCHALIDFFKAGINKHPEW




QHFGFKFSPTSSYQDLSDFYREVEPQGYQVKFVDINADYINELVEQG




QLYLFQIYNKDFSPKAHGKPNLHTLYFKALFSEDNLVNPIYKLNGEA




EIFYRKASLDMNETTIHRAGEVLENKNPDNPKKRQFVYDIIKDKRYT




QDKFMLHVPITMNFGVQGMTIKEFNKKVNQSIQQYDEVNVIGIDRG




ERHLLYLTVINSKGEILEQRSLNDITTASANGTQMTTPYHKILDKREIE




RLNARVGWGEIETIKELKSGYLSHVVHQISQLMLKYNAIVVLEDLNF




GFKRGRFKVEKQIYQNFENALIKKLNHLVLKDKADDEIGSYKNALQ




LTNNFTDLKSIGKQTGFLFYVPAWNTSKIDPETGFVDLLKPRYENIAQ




SQAFFGKFDKICYNADRGYFEFHIDYAKFNDKAKNSRQIWKICSHGD




KRYVYDKTANQNKGATIGVNVNDELKSLFTRYHINDKQPNLVMDIC




QNNDKEFHKSLMYLLKTLLALRYSNASSDEDFILSPVANDEGVFFNS




ALADDTQPQNADANGAYHIALKGLWLLNELKNSDDLNKVKLAIDN




QTWLNFAQNR





SEQ

Thiomicrospira

MGIHGVPAATKTFDSEFFNLYSLQKTVRFELKPVGETASFVEDFKNE


ID
sp. XS5
GLKRVVSEDERRAVDYQKVKEIIDDYHRDFIEESLNYFPEQVSKDAL


NO:
(TsCas12a)
EQAFHLYQKLKAAKVEEREKALKEWEALQKKLREKVVKCFSDSNK


25

ARFSRIDKKELIKEDLINWLVAQNREDDIPTVETFNNFTTYFTGFHEN




RKNIYSKDDHATAISFRLIHENLPKFFDNVISFNKLKEGFPELKFDKV




KEDLEVDYDLKHAFEIEYFVNFVTQAGIDQYNYLLGGKTLEDGTKK




QGMNEQINLFKQQQTRDKARQIPKLIPLFKQILSERTESQSFIPKQFES




DQELFDSLQKLHNNCQDKFTVLQQAILGLAEADLKKVFIKTSDLNAL




SNTIFGNYSVFSDALNLYKESLKTKKAQEAFEKLPAHSIHDLIQYLEQ




FNSSLDAEKQQSTDTVLNYFIKTDELYSRFIKSTSEAFTQVQPLFELEA




LSSKRRPPESEDEGAKGQEGFEQIKRIKAYLDTLMEAVHFAKPLYLV




KGRKMIEGLDKDQSFYEAFEMAYQELESLIIPIYNKARSYLSRKPFKA




DKFKINFDNNTLLSGWDANKETANASILFKKDGLYYLGIMPKGKTFL




FDYFVSSEDSEKLKQRRQKTAEEALAQDGESYFEKIRYKLLPGASKM




LPKVFFSNKNIGFYNPSDDILRIRNTASHTKNGTPQKGHSKVEFNLND




CHKMIDFFKSSIQKHPEWGSFGFTFSDTSDFEDMSAFYREVENQGYV




ISFDKIKETYIQSQVEQGNLYLFQIYNKDFSPYSKGKPNLHTLYWKAL




FEEANLNNVVAKLNGEAEIFFRRHSIKASDKVVHPANQAIDNKNPHT




EKTQSTFEYDLVKDKRYTQDKFFFHVPISLNFKAQGVSKFNDKVNGF




LKGNPDVNIIGIDRGERHLLYFTVVNQKGEILVQESLNTLMSDKGHV




NDYQQKLDKKEQERDAARKSWTTVENIKELKEGYLSHVVHKLAHLI




IKYNAIVCLEDLNFGFKRGRFKVEKQVYQKFEKALIDKLNYLVFKEK




ELGEVGHYLTAYQLTAPFESFKKLGKQSGILFYVPADYTSKIDPTTGF




VNFLDLRYQSVEKAKQLLSDFNAIRFNSVQNYFEFEIDYKKLTPKRK




VGTQSKWVICTYGDVRYQNRRNQKGHWETEEVNVTEKLKALFASD




SKTTTVIDYANDDNLIDVILEQDKASFFKELLWLLKLTMTLRHSKIKS




EDDFILSPVKNEQGEFYDSRKAGEVWPKDADANGAYHIALKGLWN




LQQINQWEKGKTLNLAIKNQDWFSFIQEKPYQE





SEQ

Butyrivibrio

MGIHGVPAAYYQNLTKKYPVSKTIRNELIPIGKTLENIRKNNILESDV


ID
sp. NC3005
KRKQDYEHVKGIMDEYHKQLINEALDNYMLPSLNQAAEIYLKKHV


NO:
(BsCas12a)
DVEDREEFKKTQDLLRREVTGRLKEHENYTKIGKKDILDLLEKLPSIS


26

EEDYNALESFRNFYTYFTSYNKVRENLYSDEEKSSTVAYRLINENLP




KFLDNIKSYAFVKAAGVLADCIEEEEQDALFMVETFNMTLTQEGID




MYNYQIGKVNSAINLYNQKNHKVEEFKKIPKMKVLYKQILSDREEV




FIGEFKDDETLLSSIGAYGNVLMTYLKSEKINIFFDALRESEGKNVYV




KNDLSKTTMSNIVFGSWSAFDELLNQEYDLANENKKKDDKYFEKRQ




KELKKNKSYTLEQMSNLSKEDISPIENYIERISEDIEKICIYNGEFEKIV




VNEHDSSRKLSKNIKAVKVIKDYLDSIKELEHDIKLINGSGQELEKNL




VVYVGQEEALEQLRPVDSLYNLTRNYLTKKPFSTEKVKLNFNKSTLL




NGWDKNKETDNLGILFFKDGKYYLGIMNTTANKAFVNPPAAKTEN




VFKKVDYKLLPGSNKMLPKVFFAKSNIGYYNPSTELYSNYKKGTHK




KGPSFSIDDCHNLIDFFKESIKKHEDWSKFGFEFSDTADYRDISEFYRE




VEKQGYKLTFTDIDESYINDLIEKNELYLFQIYNKDFSEYSKGKLNLH




TLYFMMLFDQRNLDNVVYKLNGEAEVFYRPASIAENELVIHKAGEG




IKNKNPNRAKVKETSTFSYDIVKDKRYSKYKFTLHIPITMNFGVDEV




RRFNDVINNALRTDDNVNVIGIDRGERNLLYVVVINSEGKILEQISLN




SIINKEYDIETNYHALLDEREDDRNKARKDWNTIENIKELKTGYLSQ




VVNVVAKLVLKYNAIICLEDLNFGFKRGRQKVEKQVYQKFEKMLIE




KLNYLVIDKSREQVSPEKMGGALNALQLTSKFKSFAELGKQSGIIYY




VPAYLTSKIDPTTGFVNLFYIKYENIEKAKQFFDGFDFIRFNKKDDMF




EFSFDYKSFTQKACGIRSKWIVYTNGERIIKYPNPEKNNLFDEKVINV




TDEIKGLFKQYRIPYENGEDIKEIIISKAEADFYKRLFRLLHQTLQMRN




STSDGTRDYIISPVKNDRGEFFCSEFSEGTMPKDADANGAYNIARKG




LWVLEQIRQKDEGEKVNLSMTNAEWLKYAQLHLL





SEQ
AacCas12b
MAVKSIKVKLRLDDMPEIRAGLWKLHKEVNAGVRYYTEWLSLLRQ


ID

ENLYRRSPNGDGEQECDKTAEECKAELLERLRARQVENGHRGPAGS


NO:

DDELLQLARQLYELLVPQAIGAKGDAQQIARKFLSPLADKDAVGGL


27

GIAKAGNKPRWVRMREAGEPGWEEEKEKAETRKSADRTADVLRAL




ADFGLKPLMRVYTDSEMSSVEWKPLRKGQAVRTWDRDMFQQAIER




MMSWESWNQRVGQEYAKLVEQKNRFEQKNFVGQEHLVHLVNQLQ




QDMKEASPGLESKEQTAHYVTGRALRGSDKVFEKWGKLAPDAPFD




LYDAEIKNVQRRNTRRFGSHDLFAKLAEPEYQALWREDASFLTRYA




VYNSILRKLNHAKMFATFTLPDATAHPIWTRFDKLGGNLHQYTFLFN




EFGERRHAIRFHKLLKVENGVAREVDDVTVPISMSEQLDNLLPRDPN




EPIALYFRDYGAEQHFTGEFGGAKIQCRRDQLAHMHRRRGARDVYL




NVSVRVQSQSEARGERRPPYAAVFRLVGDNHRAFVHFDKLSDYLAE




HPDDGKLGSEGLLSGLRVMSVDLGLRTSASISVFRVARKDELKPNSK




GRVPFFFPIKGNDNLVAVHERSQLLKLPGETESKDLRAIREERQRTLR




QLRTQLAYLRLLVRCGSEDVGRRERSWAKLIEQPVDAANHMTPDW




REAFENELQKLKSLHGICSDKEWMDAVYESVRRVWRHMGKQVRD




WRKDVRSGERPKIRGYAKDVVGGNSIEQIEYLERQYKFLKSWSFFG




KVSGQVIRAEKGSRFAITLREHIDHAKEDRLKKLADRIIMEALGYVY




ALDERGKGKWVAKYPPCQLILLEELSEYQFNNDRPPSENNQLMQWS




HRGVFQELINQAQVHDLLVGTMYAAFSSRFDARTGAPGIRCRRVPA




RCTQEHNPEPFPWWLNKFVVEHTLDACPLRADDLIPTGEGEIFVSPFS




AEEGDFHQIHADLNAAQNLQQRLWSDFDISQIRLRCDWGEVDGELV




LIPRLTGKRTADSYSNKVFYTNTGVTYYERERGKKRRKVFAQEKLSE




EEAELLVEADEAREKSVVLMRDPSGIINRGNWTRQKEFWSMVNQRI




EGYLVKQIRSRVPLQDSACENTGDI





SEQ
Cas12
MKKIDNFVGCYPVSKTLRFKAIPIGKTQENIEKKRLVEEDEVRAKDY


ID
Variant
KAVKKLIDRYHREFIEGVLDNVKLDGLEEYYMLFNKSDREESDNKKI


NO:

EIMEERFRRVISKSFKNNEEYKKIFSKKIIEEILPNYIKDEEEKELVKGF


28

KGFYTAFVGYAQNRENMYSDEKKSTAISYRIVNENMPRFITNIKVFE




KAKSILDVDKINEINEYILNNDYYVDDFFNIDFFNYVLNQKGIDIYNAI




IGGIVTGDGRKIQGLNECINLYNQENKKIRLPQFKPLYKQILSESESMS




FYIDEIESDDMLIDMLKESLQIDSTINNAIDDLKVLFNNIFDYDLSGIFI




NNGLPITTISNDVYGQWSTISDGWNERYDVLSNAKDKESEKYFEKRR




KEYKKVKSFSISDLQELGGKDLSICKKINEIISEMIDDYKSKIEEIQYLF




DIKELEKPLVTDLNKIELIKNSLDGLKRIERYVIPFLGTGKEQNRDEVF




YGYFIKCIDAIKEIDGVYNKTRNYLTKKPYSKDKFKLYFENPQLMGG




WDRNKESDYRSTLLRKNGKYYVAIIDKSSSNCMMNIEEDENDNYEK




INYKLLPGPNKMLPKVFFSKKNREYFAPSKEIERIYSTGTFKKDTNFV




KKDCENLITFYKDSLDRHEDWSKSFDFSFKESSAYRDISEFYRDVEK




QGYRVSFDLLSSNAVNTLVEEGKLYLFQLYNKDFSEKSHGIPNLHTM




YFRSLFDDNNKGNIRLNGGAEMFMRRASLNKQDVTVHKANQPIKN




KNLLNPKKTTTLPYDVYKDKRFTEDQYEVHIPITMNKVPNNPYKINH




MVREQLVKDDNPYVIGIDRGERNLIYVVVVDGQGHIVEQLSLNEIIN




ENNGISIRTDYHTLLDAKERERDESRKQWKQIENIKELKEGYISQVVH




KICELVEKYDAVIALEDLNSGFKNSRVKVEKQVYQKFEKMLITKLN




YMVDKKKDYNKPGGVLNGYQLTTQFESFSKMGTQNGIMFYIPAWL




TSKMDPTTGFVDLLKPKYKNKADAQKFFSQFDSIRYDNQEDAFVFK




VNYTKFPRTDADYNKEWEIYTNGERIRVFRNPKKNNEYDYETVNVS




ERMKELFDSYDLLYDKGELKETICEMEESKFFEELIKLFRLTLQMRNS




ISGRTDVDYLISPVKNSNGYFYNSNDYKKEGAKYPKDADANGAYNI




ARKVLWAIEQFKMADEDKLDKTKISIKNQEWLEYAQTHCE





SEQ
Cas12
MATLVSFTKQYQVQKTLRFELIPQGKTQANIDAKGFINDDLKRDENY


ID
Variant
MKVKGVIDELHKNFIEQTLVNVDYDWRSLATAIKNYRKDRSDTNKK


NO:

NLEKTQEAARKEIIAWFEGKRGNSAFKNNQKSFYGKLFKKELFSEIL


29

RSDDLEYDEETQDAIACFDKFTTYFVGFHENRKNMYSTEAKSTSVA




YRVVNENFSKFLSNCEAFSVLEAVCPNVLVEAEQELHLHKAFSDLKL




SDVFKVEAYNKYLSQTGIDYYNQIIGGISSAEGVRKIRGVNEVVNNAI




QQNDELKVALRNKQFTMVQLFKQILSDRSTLSFVSEQFTSDQEVITV




VKQFNDDIVNNKVLAVVKTLFENFNSYDLEKIYINSKELASVSNALL




KDWSKIRNAVLENKIIELGANPPKTKISAVEKEVKNKDFSIAELASYN




DKYLDKEGNDKEICSIANVVLEAVGALEIMLAESLPADLKTLENKNK




VKGILDAYENLLHLLNYFKVSAVNDVDLAFYGAFEKVYVDISGVMP




LYNKVRNYATKKPYSVEKFKLNFAMPTLADGWDKNKERDNGSIILL




KDGQYYLGVMNPQNKPVIDNAVCNDAKGYQKMVYKMFPEISKMV




TKCSTQLNAVKAHFEDNTNDFVLDDTDKFISDLTITKEIYDLNNVLY




DGKKKFQIDYLRNTGDFAGYHKALETWIDFVKEFLSKYRSTAIYDLT




TLLPTNYYEKLDVFYSDVNNLCYKIDYENISVEQVNEWVEEGNLYL




FKIYNKDFATGSTGKPNLHTMYWNAVFAEENLHDVVVKLNGGAEL




FYRPKSNMPKVEHRVGEKLVNRKNVNGEPIADSVHKEIYAYANGKI




SKSELSENAQEELPLAIIKDVKHNITKDKRYLSDKYFFHVPITLNYKA




NGNPSAFNTKVQAFLKNNPDVNIIGIDRGERNLLYVVVIDQQGNIIDK




KQVSYNKVNGYDYYEKLNQREKERIEARQSWGAVGKIKELKEGYL




SLVVREIADMMVKYNAIVVMENLNAGFKRVRGGIAEKAVYQKFEK




MLIDKLNYLVFKDVEAKEAGGVLNAYQLTDKFDSFEKMGNQSGFLF




YVPAAYTSKIDPVTGFANVFSTKHITNTEAKKEFICSFNSLRYDEAKD




KFVLECDLNKFKIVANSHIKNWKFIIGGKRIVYNSKNKTYMEKYPCE




DLKATLNASGIDFSSSEIINLLKNVPANREYGKLFDETYWAIMNTLQ




MRNSNALTGEDYIISAVADDNEKVFDSRTCGAELPKDADANGAYHI




ALKGLYLLQRIDISEEGEKVDLSIKNEEWFKFVQQKEYAR





SEQ
Cas12
MKEQFINRYPLSKTLRFSLIPVGETENNFNKNLLLKKDKQRAENYEK


ID
Variant
VKCYIDRFHKEYIESVLSKARIEKVNEYANLYWKSNKDDSDIKAMES


NO:

LENDMRKQISKQLTSTEIYKKRLFGKELICEDLPSFLTDKDERETVEC


30

FRSFTTYFKGFNTNRENMYSSDGKSTAIAYRCINDNLPRFLDNVKSF




QKVFDNLSDETITKLNTDLYNIFGRNIEDIFSVDYFEFVLTQSGIEIYNS




MIGGYTCSDKTKIQGLNECINLYNQQVAKNEKSKKLPLMKPLYKQIL




SEKDSVSFIPEKFNSDNEVLHAIDDYYTGHIGDFDLLTELLQSLNTYN




ANGIFVKSGVAITDISNGAFNSWNVLRSAWNEKYEALHPVTSKTKID




KYIEKQDKIYKAIKSFSLFELQSLGNENGNEITDWYISSINESNSKIKE




AYLQAQKLLNSDYEKSYNKRLYKNEKATELVKNLLDAIKEFQKLIK




PLNGTGKEENKDELFYGKFTSYYDSIADIDRLYDKVRNYITQKPYSK




DKIKLNFDNPQLLGGWDKNKESDYRTVLLHKDGLYYLAVMDKSHS




KAFVDAPEITSDDKDYYEKMEYKLLPGPNKMLPKVFFASKNIDTFQP




SDRILDIRKRESFKKGATFNKAECHEFIDYFKDSIKKHDDWSQFGFKF




SPTESYNDISEFYREISDQGYSVRFNKISKNYIDGLVNNGYIYLFQIYN




KDFSKYSKGTPNLHTLYFKMLFDERNLSNVVYKLNGEAEMFYREAS




IGDKEKITHYANQPIKNKNPDNEKKESVFEYDIVKDKRFTKRQFSLH




LPITINFKAHGQEFLNYDVRKAVKYKDDNYVIGIDRGERNLIYISVIN




SNGEIVEQMSLNEIISDNGHKVDYQKLLDTKEKERDKARKNWTSVE




NIKELKEGYISQVVHKICELVIKYDAVIAMEDLNFGFKRGRFPVEKQ




VYQKFENMLISKLNLLIDKKAEPTEDGGLLRAYQLTNKFDGVNKAK




QNGIIFYVPAWDTSKIDPATGFVNLLKPKCNTSVPEAKKLFETIDDIK




YNANTDMFEFYIDYSKFPRCNSDFKKSWTVCTNSSRILTFRNKEKNN




KWDNKQIVLTDEFKSLFNEFGIDYKGNLKDSILSISNADFYRRLIKLL




SLTLQMRNSITGSTLPEDDYLISPVANKSGEFYDSRNYKGTNAALPC




DADANGAYNIARKALWAINVLKDTPDDMLNKAKLSITNAEWLEYT




QK





SEQ
Cas12
MNNPRGAFGGFTNLYSLSKTLRFELKPYLEIPEGEKGKLFGDDKEYY


ID
Variant
KNCKTYTEYYLKKANKEYYDNEKVKNTDLQLVNFLHDERIEDAYQ


NO:

VLKPVFDTLHEEFITDSLESAEAKKIDFGNYYGLYEKQKSEQNKDEK


31

KKIDKPLETERGKLRKAFTPIYEAEGKNLKNKAGKEKKDKDILKESG




FKVLIEAGILKYIKNNIDEFADKKLKNNEGKEITKKDIETALGAENIEG




IFDGFFTYFSGFNQNRENYYSTEEKATAVASRIVDENLSKFCDNILLY




RKNENDYLKIFNFLKNKGKDLKLKNSKFGKENEPEFIPAYDMKNDE




KSFSVADFVNCLSQGEIEKYNAKIANANYLINLYNQNKDGNSSKLSM




FKILYKQIGCGEKKDFIKTIKDNAELKQILEKACEAGKKYFIRGKSED




GGVSNIFDFTDYIQSHENYKGVYWSDKAINTISGKYFANWDTLKNK




LGDAKVFNKNTGEDKADVKYKVPQAVMLSELFAVLDDNAGEDWR




EKGIFFKASLFEGDQNKSEIIKNANRPSQALLKMICDDMESLAKNFID




SGDKILKISDRDYQKDENKQKIKNWLDNALWINQILKYFKVKANKI




KGDSIDARIDSGLDMLVFSSDNPAEDYDMIRNYLTQKPQDEINKLKL




NFENSSLAGGWDENKEKDNSCIILKDEQDKQYLAVMKYENTKVFEQ




KNSQLYIADNAAWKKMIYKLVPGASKTLPKVFFSKKWTANRPTPSD




IVEIYQKGSFKKENVDFNDKKEKDESRKEKNREKIIAELQKTCWMDI




RYNIDGKIESAKYVNKEKLAKLIDFYKENLKKYPSEEESWDRLFAFG




FSDTKSYKSIDQFYIEVDKQGYKLEFVTINKARLDEYVRDGKIYLFEI




RSRDNNLVNGEEKTSAKNLQTIYWNAAFGGDDNKPKLNGEAEIFYR




PAIAENKLNKKKDKNGKEIIDGYRFSKEKFIFHCPITLNFCLKETKIND




KLNAALAKPENGQGVYFLGIDRGEKHLAYYSLVNQKGEILEQGTLN




LPFLDKNGKSRSIKVEKKSFEKDSNGGIIKDKDGNDKIKIEFVECWNY




NDLLDARAGDRDYARKNWTTIGTIKELKDGYISQVVRKIVDLSIYKN




TETKEFREMPAFIVLEDLNIGFKRGRQKIEKQVYQKLELALAKKLNF




LVDKKADIGEIGSVTKAIQLTPPVNNFGDMENRKQFGNMLYIRADYT




SQTDPATGWRKSIYLKSGSESNVKEQIEKSFFDIRYESGDYCFEYRDR




HGKMWQLYSSKNGVSLDRFHGERNNSKNVWESEKQPLNEMLDILF




DEKRFDKSKSLYEQMFKGGVALTRLPKEINKKDKPAWESLRFVIILIQ




QIRNTGKNGDDRNGDFIQSPVRDEKTGEHFDSRIYLDKEQKGEKADL




PTSGDANGAYNIARKGIVVAEHIKRGFDKLYISDEEWDTWLAGDEI




WDKWLKENRESLTKTRK





SEQ
Cas12
MNGNRIIVYREFVGVTPVAKTLRNELRPIGHTQEHIIHNGLIQEDELR


ID
Variant
QEKSTELKNIMDDYYREYIDKSLSGVTDLDFTLLFELMNLVQSSPSK


NO:

DNKKALEKEQSKMREQICTHMQSDSNYKNIFNAKFLKEILPDFIKNY


32

NQYDAKDKAGKLETLALFNGFSTYFTDFFEKRKNVFTKEAVSTSIAY




RIVHENSLTFLANMTSYKKISEKALDEIEVIEKNNQDKMGDWELNQI




FNPDFYNMVLIQSGIDFYNEICGVVNAHMNLYCQQTKNNYNLFKMR




KLHKQILAYTSTSFEVPKMFEDDMSVYNAVNAFIDETEKGNIIGKLK




DIVNKYDELDEKRIYISKDFYETLSCFMSGNWNLITGCVENFYDENIH




AKGKSKEEKVKKAVKEDKYKSINDVNDLVEKYIDEKERNEFKNSNA




KQYIREISNIITDTETAHLEYDEHISLIESEEKADEMKKRLDMYMNMY




HWAKAFIVDEVLDRDEMFYSDIDDIYNILENIVPLYNRVRNYVTQKP




YNSKKIKLNFQSPTLANGWSQSKEFDNNAIILIRDNKYYLAIFNAKNK




PDKKIIQGNSDKKNDNDYKKMVYNLLPGANKMLPKVFLSKKGIETF




KPSDYIISGYNAHKHIKTSENFDISFCRDLIDYFKNSIEKHAEWRKYEF




KFSATDSYNDISEFYREVEMQGYRIDWTYISEADINKLDEEGKIYLFQ




IYNKDFAENSTGKENLHTMYFKNIFSEENLKDIIIKLNGQAELFYRRA




SVKNPVKHKKDSVLVNKTYKNQLDNGDVVRIPIPDDIYNEIYKMYN




GYIKENDLSEAAKEYLDKVEVRTAQKDIVKDYRYTVDKYFIHTPITI




NYKVTARNNVNDMAVKYIAQNDDIHVIGIDRGERNLIYISVIDSHGNI




VKQKSYNILNNYDYKKKLVEKEKTREYARKNWKSIGNIKELKEGYI




SGVVHEIAMLMVEYNAIIAMEDLNYGFKRGRFKVERQVYQKFESML




INKLNYFASKGKSVDEPGGLLKGYQLTYVPDNIKNLGKQCGVIFYVP




AAFTSKIDPSTGFISAFNFKSISTNASRKQFFMQFDEIRYCAEKDMFSF




GFDYNNFDTYNITMSKTQWTVYTNGERLQSEFNNARRTGKTKSINL




TETIKLLLEDNEINYADGHDVRIDMEKMDEDKNSEFFAQLLSLYKLT




VQMRNSYTEAEEQEKGISYDKIISPVINDEGEFFDSDNYKESDDKECK




MPKDADANGAYCIALKGLYEVLKIKSEWTEDGFDRNCLKLPHAEW




LDFIQNKRYE





SEQ
Cas12
MKKIDSFVNYYPLSKTLRFSLIPVGKTEDNFNAKLLLEEDEKRAIEYE


ID
Variant
KVKRYIDRYHKHFIETVLANFHLDDVNEYAELYYKAGKDDKDLKY


NO:

MEKLEGKMRKSISAAFTKDKKYKEIFGQEIIKNILPEFLENEDEKESV


33

KMFQGFFTYFTGFNDNRKNMYTHEAQTTAISYRCINENLPKFLDNV




QSFAKIKESISSDIMNKLDEVCMDLYGVYAQDMFCTDYFSFVLSQSG




IDRYNNIIGGYVDDKGVKIQGINEYINLYNQQVDEKNKRLPLMKKLY




KQILIEKESISFIPEKFESDNIVINAISDYYHNNVENLFDDFNKLFNEFS




EYDDNGIFVTSGLAVTDISNAVFGSWNIISDSWNEEYKDSHPMKKTT




NAEKYYEDMKKEYKKNLSFTIAELQRLGEAGCNDECKGDIKEYYKT




TVAEKIENIKNAYEISKDLLASDYEKSNDKKLCKNDSAISLLKNLLDS




IKDLEKTIKPLLGTGKEENKDDVFYGKFTNLYEMISEIDRLYDKVRN




YVTQKPYSKDKIKLNFENPQHLGGWDKNKERDYRSVLLKKEDKYY




LAIMDKSNNKAFIDFPDDGECYEKIEYKLLPGPNKMLPKVFFASSNIE




YFAPSKKILEIRSRESFKKGDMFNLKDCHEFIDFFKESIKKHEDWSQF




GFEFSPTEKYNDISEFYNEVKIQGYSLKYKNVSKKYIDELIECGQLYL




FQIYNKDFSVYAKGNPNLHTMYFKMLFDERNLANVVYQLNGGAEM




FYRKASIKDSEKIVHHANQPIKNKNADNVKKESVFEYDIIKDKRFTK




RQFSIHIPITLNFKAKGQNFINNDVRMALKKADENYVIGIDRGERNLL




YICVINSKGEIVEQKSLNEIIGDNGYRVDYHKLLDKKEAERDEARKS




WGTIENIKELKEGYLSQIVHEISKLVIKYDAVIAIEDLNSGFKKGRFKV




EKQVYQKFENMLCTKLNYLVDKNADANECGGLLKAYQLTNKEDG




ANRGRQNGIIFSVPAWLTSKIDPVTGFADLLRPKYKSVSESVEFISKID




NIRYNSKEDYFEFDIDYSKFPNSTASYKKKWTVCTYGERIINVRNKE




KNNMWDNKTIVLTDEFKKLFADFGVDVSKNIKESVLAIDSKDFYYR




FINLLANTLQLRNSEVGNVDVDYLISPVKGVDGSFYDSRLVKEKTLP




ENADANGAYNIARKALWAIDVLKQTKDEELKNANLSIKNAEWLEY




VQK





SEQ
Cas12
MRTMVTFEDFTKQYQVSKTLRFELIPQGKTLENMKRDGIISVDRQRN


ID
Variant
EDYQKAKGILDKLYKYILDFTMETVVIDWEALATATEEFRKSKDKK


NO:

TYEKVQSKIRTALLEHVKKQKVGTEDLFKGMFSSKIITGEVLAAFPEI


34

RLSDEENLILEKFKDFTTYFTGFFENRKNVFTDEALSTSFTYRLVNDN




FIKFFDNCIVFKNVVNISPHMAKSLETCASDLGIFPGVSLEEVFSISFY




NRLLTQTGIDQFNQLLGGISGKEGEHKQQGLNEIINLAMQQNLEVKE




VLKNKAHRFTPLFKQILSDRSTMSFIPDAFADDDEVLSAVDAYRKYL




SEKNIGDRAFQLISDMEAYSPELMRIGGKYVSVLSQLLFYSWSEIRDG




VKAYKESLITGKKTKKELENIDKEIKYGVTLQEIKEALPKKDIYEEVK




KYAMSVVKDYHAGLAEPLPEKIETDDERASIKHIMDSMLGLYRFLE




YFSHDSIEDTDPVFGECLDTILDDMNETVPLYNKVRNFSTRKVYSTE




KFKLNFNNSSLANGWDKNKEQANGAILLRKEGEYFLGIFNSKNKPK




LVSDGGAGIGYEKMIYKQFPDFKKMLPKCTISLKDTKAHFQKSDEDF




TLQTDKFEKSIVITKQIYDLGTQTVNGKKKFQVDYPRLTGDMEGYR




AALKEWIDFGKEFIQAYTSTAIYDTSLFRDSSDYPDLPSFYKDVDNIC




YKLTFEWIPDAVIDDCIDDGSLYLFKLHNKDFSSGSIGKPNLHTLYW




KALFEEENLSDVVVKLNGQAELFYRPKSLTRPVVHEEGEVIINKTTST




GLPVPDDVYVELSKFVRNGKKGNLTDKAKNWLDKVTVRKMPHAIT




KDRRFTVDKFFFHVPITLNYKADSSPYRFNDFVRQYIKDCSDVKIIGI




DRGERNLIYAVVIDGKGNIIEQRSFNTVGTYNYQEKLEQKEKERQTA




RQDWATVTKIKDLKKGYLSAVVHELSKMIVKYKAIVALENLNVGFK




RMRGGIAERSVYQQFEKALIDKLNYLVFKDEEQSGYGGVLNAYQLT




DKFESFSKMGQQTGFLFYVPAAYTSKIDPLTGFINPFSWKHVKNRED




RRNFLNLFSKLYYDVNTHDFVLAYHHSNKDSKYTIKGNWEIADWDI




LIQENKEVFGKTGTPYCVGKRIVYMDDSTTGHNRMCAYYPHTELKK




LLSEYGIEYTSGQDLLKIIQEFDDDKLVKGLFYIIKAALQMRNSNSET




GEDYISSPIEGRPGICFDSRAEADTLPYDADANGAFHIAMKGLLLTER




IRNDDKLAISNEEWLNYIQEMRG





SEQ
Cas12
MNKDIRKNFTDFVGISEIQKTLRFILIPIGKTAQNIDKYNMFEDDEIRH


ID
Variant
EYYPILKEACDDFYRNHIDQQFENLELDWSKLDEALASEDRDLINET


NO:

RATYRQVLFNRLKNSVDIKGDSKKNKTLSLESSDKNLGKKKTKNTF


35

QYNFNDLFKAKLIKAILPLYIEYIYEGEKLENAKKALKMYNRFTSRLS




NFWQARANIFTDDEISTGSPYRLVNDNFTIFRINNSIYTKNKPFIEEDIL




EFEKKLKSKKIIKDFESVDDYFTVNAFNKLCTQNGIDKYNSILGGFTT




KEREKVKGLNELFNLAQQSINKGKKGEYRKNIRLGKLTKLKKQILAI




SDSTSFLIEQIEDDQDLYNKIKDFFELLLKEEIENENIFTQYANLQKLIE




QADLSKIYINAKHLNKISHQVTGKWDSLNKGIALLLENININEESKEK




SEVISNGQTKDISSEAYKRYLQIQSEEKDIERLRTQIYFSLEDLEKALD




LVLIDENMDRSDKSILSYVQSPDLNVNFERDLTDLYSRIMKLEENNE




KLLANHSAIDLIKEFLDLIMLRYSRWQILFCDSNYELDQTFYPIYDAV




MEILSNIIRLYNLARNYLSRKPDRMKKKKINFNNPTLADGWSESKIPD




NSSMLFIKDGMYYLGIIKNRAAYSELLEAESLQSSEKKKSENSSYER




MNYHFLPDAFRSIPKSSIAMKAVKEHFEINQKTADLLLDTDKFSKPLR




ITKEIFDMQYVDLHKNKKKYQVDYLRDTGDKKGYRKALNTWLNFC




KDFISKYKGRNLFDYSKIKDADHYETVNEFYNDVDKYSYHIFFTSVA




ETTVEKFISEGKLYLFQLYNKDFSPHSTGKPNLHTIYWRALFSEENLT




SKNIKLNGQAEIFFRPKQIETPFTHKKGSILVNRFDVNGNPIPINVYQEI




KGFKNNVIKWDDLNKTTQEGLENDQYLYFESEFEIIKDRRYTEDQLF




FHVPISFNWDIGSNPKINDLATQYIVNSNDIHIIGIDRGENHLIYYSVID




LQGAIVEQGSLNTITEYTENKFLNNKTNNLRKIPYKDILQQREDERAD




ARIKWHAIDKIKDLKDGYLGQIVHFLAKLIIKYNAIVILEDLNYGFKR




GRFKVERQVYQKFEMALMKKLNVLVFKDYDIDEIGGPLKPWQLTRP




IDSYERMGRQNGILFYVPAAYTSAVDPVTGFANLFYLNNVKNSEKF




HFFSKFESIKYHSDQDMFSFAFDYNNFGTTTRINDLSKSKWQVFTNH




ERSVWNNKEKNYVTQNLTDLIKKLLRTYNIEFKNNQNVLDSILKIEN




NTDKENFARELFRLFRLTIQLRNTTVNENNTEITENELDYIISPVKDKN




GNFFDSRDELKNLPDNGDANGAYNIARKGLLYIEQLQESIKTGKLPT




LSISTLDWFNYIMK





SEQ
Cas12
MTPIFCNFVVYQIMLFNNNININVKTMNKKHLSDFTNLFPVSKTLRFR


ID
Variant
LEPQGKTMENIVKAQTIETDEERSHDYEKTKEYIDDYHRQFIDDTLD


NO:

KFAFKVESTGNNDSLQDYLDAYLSANDNRTKQTEEIQTNLRKAIVSA


36

FKMQPQFNLLFKKEMVKHLLPQFVDTDDKKRIVAKFNDFTTYFTGF




FTNRENMYSDEAKSTSIAYRIVNQNLIKFVENMLTFKSHILPILPQEQL




ATLYDDFKEYLNVASIAEMFELDHFSIVLTQRQIEVYNSVIGGRKDE




NNKQIKPGLNQYINQHNQAVKDKSARLPLLKPLFNQILSEKAGVSFL




PKQFKSASEVVKSLNEAYAELSPVLAAIQDVVTNITDYDCNGIFIKND




LGLTDIAQRFYGNYDAVKRGLRNQYELETPMHNGQKAEKYEEQVA




KHLKSIESVSLAQINQVVTDGGDICDYFKAFGATDDGDIQRENLLASI




NNAHTAISPVLNKENANDNELRKNTMLIKDLLDAIKRLQWFAKPLL




GAGDETNKDQVFYGKFEPLYNQLDETISPLYDKVRSYLTKKPYSLD




KFKINFEKSNLLGGWDPGADRKYQYNAVILRKDNDFYLGIMRDEAT




SKRKCIQVLDCNDEGLDENFEKVEYKQIKPSQNMPRCAFAKKECEE




NADIMELKRKKNAKSYNTNKDDKNALIRHYQRYLDRTYPEFGFVY




KDADEYDTVKAFTDSMDSQDYKLSFLQVSETGLNKLVDEGDLYLFK




ITNKDFSSYAKGRPNLHTIYWRMLFDPKNLANVVYKLEGKAEVFFR




RKSLASTTTHKAKQAIKNKSRYNEAVKPQSTFDYDIIKDRRFTADKF




EFHVPIKMNFKAAGWNSTRLTNEVREFIKSQGVRHIIGIDRGERHLLY




LTMIDMDGNIVKQCSLNAPAQDNARASEVDYHQLLDSKEADRLAA




RRNWGTIENIKELKQGYLSQVVHLLATMMVDNDAILVLENLNAGF




MRGRQKVEKSVYQKFEKMLIDKLNYIVDKGQSPDKPTGALHAVQL




TGLYSDFNKSNMKRANVRQCGFVFYIPAWNTSKIDPVTGFVNLFDT




HLSSMGEIKAFFSKFDSIRYNQDKGWFEFKFDYSRFTTRAEGCRTQW




TVCTYGERIWTHRSKNQNNQFVNDTVNVTQQMLQLLQDCGIDPNG




NLKEAIANIDSKKSLETLLHLFKLTVQMRNSVTGSEVDYMISPVADE




RGHFFDSRESDEHLPANADANGAFNIARKGLMVVRQIMATDDVSKI




KFAVSNKDWLRFAQHIDD





SEQ
Cas12
MNKGGYVIMEKMTEKNRWENQFRITKTIKEEIIPTGYTKVNLQRVN


ID
Variant
MLKREMERNEDLKKMKEICDEYYRNMIDVSLRLEQVRTLGWESLIH


NO:

KYRMLNKDEKEIKALEKEQEDLRKKISKGFGEKKAWTGEQFIKKILP


37

QYLMDHYTGEELEEKLRIVKKFKGCTMFLSTFFKNRENIFTDKPIHT




AVGHRITSENAMLFAANINTYEKMESNVTLEIERLQREFWRRGINISE




IFTDAYYVNVLTQKQIEAYNKICGDINQHMNEYCQKQKLKFSEFRM




RELKKQILAVVGEHFEIPEKIESTKEVYRELNEYYESLKELHGQFEEL




KSVQLKYSQIYVQKKGYDRISRYIGGQWDLIQECMKKDCASGMKGT




KKNHDAKIEEEVAKVKYQSIEHIQKLVCTYEEDRGHKVTDYVDEFIV




SVCDLLGADHIITRDGERIELPLQYEPGTDLLKNDTINQRRLSDIKTIL




DWHMDMLEWLKTFLVNDLVIKDEEFYMAIEELNERMQCVISVYNRI




RNYVTQKGYEPEKIRICFDKGTILTGWTTGDNYQYSGFLLMRNDKY




YLGIINTNEKSVRKILDGNEECKDENDYIRVGYHLINDASKQLPRIFV




MPKAGKKSEILMKDEQCDYIWDGYCHNKHNESKEFMRELIDYYKR




SIMNYDKWEGYCFKFSSTESYDNMQDFYKEVREQSYNISFSYINENV




LEQLDKDGKIYLFQVYNKDFAAGSTGTPNLHTMYLQNLFSSQNLEL




KRLRLGGNAELFYRPGTEKDVTHRKGSILVDRTYVREEKDGIEVRDT




VPEKEYLEIYRYLNGKQKGDLSESAKQWLDKVHYREAPCDIIKDKR




YAQEKYFLHFSVEINPNAEGQTALNDNVRRWLSEEEDIHVIGIDRGE




RNLIYVSLMDGKGRIKDQKSYNIVNSGNKEPVDYLAKLKVREKERD




EARRNWKAIGKIKDIKTGYLSYVVHEIVEMAVREKAIIVMEDLNYGF




KRGRFKVERQVYQKFEEMLINKLNYVVDKQLSVDEPGGLLRGYQL




AFIPKDKKSSMRQNGIVFYVPAGYTSKIDPTTGFVNIFKFPQFGKGDD




DGNGKDYDKIRAFFGKFDEIRYECDEKVTADNTREVKERYRFDFDY




SKFETHLVHMKKTKWTVYAEGERIKRKKVGNYWTSEVISDIALRMS




NTLNIAGIEYKDGHNLVNEICALRGKQAGIILNELLEIVRLTVQLRNS




TTEGDVDERDEIISPVLNEKYGCFYHSTEYKQQNGDVLPKDADANG




AYCIGLKGIYEIRQIKNKWKEDMTKGEGKALNEGMRISHDQWFEFIQ




NMNKGE





SEQ
Cas12
MNELVKNRCKQTKTICQKLIPIGKTRETIEKYNLMEIDRKIAANKEL


ID
Variant
MNKLFSLIAGKHINDTLSKCTDLDFEPLLTSLSSLNNAKENDRDNLRE


NO:

YYDSVFEEKKTLAEEISSRLTAVKFAGKDFFTKNIPDFLETYEGDDKN


38

EMSELVSLVIENTVTAGYVKKLEKIDRSMEYRLVSGTVVKRVLTDN




ADIYEKNIEKAKDFDYGVLNIDEASQFTTLVAKDYANYLTADGIAIY




NVGIGKINLALNEYCQKNKEYSYNKLALLPLQKMLYGEKLSLFEKL




EDFTSDEELINSYNKFAKTVNESGLAEIIKKAVPSYDEIVIKPNKISNY




SNSITGHWSLVNRIMKDYLENNGIKNADKYMEKGLTLSEIGDALEN




KNIKHSDFISNLINDLGHTYTEIKENKESLKKDESVNALIIKKELDMLL




SILQNLKVFDIDNEMFDTGFGIEVSKAIEILGYGVPLYNKIRNYITKKP




DPKKKFMTKFGSATIGTGITTSVEGSKKATFLKDGDAVFLLLYNTAG




CKANNVSVSNLADLINSSLEIENSGKCYQKMIYQTPGDIKKQIPRVFV




YKSEDDDLIKDFKAGLHKTDLSFLNGRLIPYLKEAFATHETYKNYTF




SYRNSYESYDEFCEHMSEQAYILEWKWIDKKLIDDLVEDGSLLMFR




VWNRFMKKKEGKISKHAKIVNELFSDENASNAAIKLLSVFDIFYRDK




QIDNPIVHKAGTTLYNKRTKDGEVIVDYTTMVKNKEKRPNVYTTTK




KYDIIKDRRYTEEQFEIHLHVNIGKEENKEKLETSKVINEKKNTLVVT




RSNEHLLYVVIFDENDNILLKKSLNTVKGMNFKSKLEVVEIQKKENM




QSWKTVGSNQALMEGYLSFAIKEIADLVKEYDAILVLEQNSVGKNIL




NERVYTRFKEMLITNLSLDVDYENKDFYSYTELGGKVASWRDCVTN




GICIQVPSAYKYKDPTTSFSTISMYAKTTAEKSKKLKQIKSFKYNRER




GLFELVIAKGVGLENNIVCDSFGSRSIIENDISKEVSCTLKIEKYLIDAG




IEYNDEKEVLKDLDTAAKTDAVHKAVTLLLKCFNESPDGRYYISPCG




EHFTLCDAPEVLSAINYYIRSRYIREQIVEGVKKMEYKKTILLAK





SEQ
Cas12
MNYKTGLEDFIGKESLSKTLRNALIPTESTKIHMEEMGVIRDDELRAE


ID
Variant
KQQELKEIMDDYYRTFIEEKLGQIQGIQWNSLFQKMEETMEDISVRK


NO:

DLDKIQNEKRKEICCYFTSDKRFKDLFNAKLITDILPNFIKDNKEYTEE


39

EKAEKEQTRVLFQRFATAFTNYFNQRRNNFSEDNISTAISFRIVNENS




EIHLQNMRAFQRIEQQYPEEVCGMEEEYKDMLQEWQMKHIYSVDF




YDRELTQPGIEYYNGICGKINEHMNQFCQKNRINKNDFRMKKLHKQI




LCKKSSYYEIPFRFESDQEVYDALNEFIKTMKKKEIIRRCVHLGQECD




DYDLGKIYISSNKYEQISNALYGSWDTIRKCIKEEYMDALPGKGEKK




EEKAEAAAKKEEYRSIADIDKIISLYGSEMDRTISAKKCITEICDMAG




QISIDPLVCNSDIKLLQNKEKTTEIKTILDSFLHVYQWGQTFIVSDIIEK




DSYFYSELEDVLEDFEGITTLYNHVRSYVTQKPYSTVKFKLHFGSPTL




ANGWSQSKEYDNNAILLMRDQKFYLGIFNVRNKPDKQIIKGHEKEE




KGDYKKMIYNLLPGPSKMLPKVFITSRSGQETYKPSKHILDGYNEKR




HIKSSPKFDLGYCWDLIDYYKECIHKHPDWKNYDFHFSDTKDYEDIS




GFYREVEMQGYQIKWTYISADEIQKLDEKGQIFLFQIYNKDFSVHST




GKDNLHTMYLKNLFSEENLKDIVLKLNGEAELFFRKASIKTPIVHKK




GSVLVNRSYTQTVGNKEIRVSIPEEYYTEIYNYLNHIGKGKLSSEAQR




YLDEGKIKSFTATKDIVKNYRYCCDHYFLHLPITINFKAKSDVAVNE




RTLAYIAKKEDIHIIGIDRGERNLLYISVVDVHGNIREQRSFNIVNGYD




YQQKLKDREKSRDAARKNWEEIEKIKELKEGYLSMVIHYIAQLVVK




YNAVVAMEDLNYGFKTGRFKVERQVYQKFETMLIEKLHYLVFKDR




EVCEEGGVLRGYQLTYIPESLKKVGKQCGFIFYVPAGYTSKIDPTTGF




VNLFSFKNLTNRESRQDFVGKFDEIRYDRDKKMFEFSFDYNNYIKKG




TILASTKWKVYTNGTRLKKIVVNGKYTSQSMEVELTDAMEKMLQR




AGIEYHDGKDLKGQIVEKGIEAEIIDIFRLTVQMRNSRSESEDREYDR




LISPVLNDKGEFFDTATADKTLPQDADANGAYCIALKGLYEVKQIKE




NWKENEQFPRNKLVQDNKTWFDFMQKKRYL





SEQ
Cas12
MEDKQFLERYKEFIGLNSLSKTLRNSLIPVGSTLKHIQEYGILEEDSLR


ID
Variant
AQKREELKGIMDDYYRNYIEMHLRDVHDIDWNELFEALTEVKKNQ


NO:

TDDAKKRLEKIQEKKRKEIYQYLSDDAVFSEMFKEKMISGILPDFIRC


40

NEGYSEEEKEEKLKTVALFHRFTSSFNDFFLNRKNVFTKEAIVTAIGY




RVVHENAEIFLENMVAFQNIQKSAESQISIIERKNEHYFMEWKLSHIF




TADYYMMLMTQKAIEHYNEMCGVVNQQMREYCQKEKKNWNLYR




MKRLHKQILSNASTSFKIPEKYENDAEVYESVNSFLQNVMEKTVME




RIAVLKNSTDNFDLSKIYITAPYYEKISNYLCGSWNTITDCLTHYYEQ




QIAGKGARKDQKVKAAVKADKWKSLSEIEQLLKEYARAEEVKRKP




EEYIAEIENIVSLKEAHLLEYHPEVNLIENEKYATEIKDVLDNYMELF




HWMKWFYIEEAVEKEVNFYGELDDLYEEIKDIVPLYNKVRNYVTQK




PYSDTKIKLNFGTPTLANGWSKSKEYDYNAILLQKDGKYYMGIFNPI




QKPEKEIIEGHSQPLEGNEYKKMVYYYLPSANKMLPKVLLSKKGMEI




YQPSEYIINGYKERRHIKSEEKFDLQFCHDLIDYFKSGIERNSDWKVF




GFDFSDTDTYQDISGFYREVEDQGYKIDWTYIKEADIDRLNEEGKLY




LFQIYNKDFSEKSTGRENLHTMYLKNLFSEENVREQVLKLNGEAEIF




FRKSSVKKPIIHKKGTMLVNRTYMEEVNGNSVRRNIPEKEYQEIYNY




KNHRLKGELSTEAKKYLEKAVCHETKKDIVKDYRYSVDKFFIHLPITI




NYRASGKETLNSVAQRYIAHQNDMHVIGIDRGERNLIYVSVINMQG




EIKEQKSFNIINEFNYKEKLKEREQSRGAARRNWKEIGQIKDLKEGYL




SGVIHEIAKMMIKYHAIIAMEDLNYGFKRGRFKVERQVYQKFENMLI




QKLNYLVFKDRPADEDGGVLRGYQLAYIPDSVKKMGRQCGMIFYV




PAAFTSKIDPTTGFVDIFKHKVYTTEQAKREFILSFDEICYDVERQLFR




FTFDYANFVTQNVTLARNNWTIYTNGTRAQKEFGNGRMRDKEDYN




PKDKMVELLESEGIEFKSGKNLLPALKKVSNAKVFEELQKIVRFTVQ




LRNSKSEENDVDYDHVISPVLNEEGNFFDSSKYKNKEEKKESLLPVD




ADANGAYCIALKGLYIMQAIQKNWSEEKALSPDVLRLNNNDWFDYI




QNKRYR





SEQ
Cas12
MEKSLNDFIGLYSVSKTLRFELKPVSETLENIKKFHFLEEDKKKAND


ID
Variant
YKDVKKIIDNYHKYFIDDVLKNASFNWKKLEEAIREYNKNKSDDSA


NO:

LVAEQKKLGDAILKLFTSDKRYKALTAATPKELFESILPDWFGEQCN


41

QDLNKAALKTFQKFTSYFTGFQENRKNVYSAEAIPTAVPYRIVNDNF




PKFLQNVLIFKTIQEKCPQIIDEVEKELSSYLGKEKLAGIFTLESFNKY




LGQGGKENQRGIDFYNQIIGGVVEKEGGINLRGVNQFLNLYWQQHP




DFTKEDRRIKMVPLYKQILSDRSSLSFKIESIENDEELKNALLECADKL




ELKNDEKKSIFEEVCDLFSSVKNLDLSGIYINRKDINSVSRILTGDWS




WLQSRMNVYAEEKFTTKAEKARWQKSLDDEGENKSKGFYSLTDLN




EVLEYSSENVAETDIRITDYFEHRCRYYVDKETEMFVQGSELVALSL




QEMCDDILKKRKAMNTVLENLSSENKLREKTDDVAVIKEYLDAVQE




LLHRIKPLKVNGVGDSTFYSVYDSIYSALSEVISVYNKTRNYITKKAA




SPEKYKLNFDNPTLADGWDLNKEQANTSVILRKDGMFYLGIMNPKN




KPKFAEKYDCGNESCYEKMIYKQFDATKQIPKCSTQKKEVQKYFLS




GATEPYILNDKKSFKSELIITKDIWFMNNHVWDGEKFVPKRDNETRP




KKFQIGYFKQTGDFDGYKNALSNWISFCKNFLQSYLSATVYDYNFK




NSEEYEGLDEFYNYLNATCYKLNFINIPETEINKMVSEGKLYLFQIYN




KDFASGSTGMPNMHTLYWKNLFSDENLKNVCLKLNGEAELFYRPA




GIKEPVIHKEGSYLVNRTTEDGESIPEKIYFEIYKNANGKLEKLSDEA




QNYISNHEVVIKKAGHEIIKDRHYTEPKFLFHVPLTINFKASGNSYSIN




ENVRKFLKNNPDVNIIGLDRGERHLIYLSLINQKGEIIKQFTFNEVERN




KNGRTIKVNYHEKLDQREKERDAARKSWQAIGKIAELKEGYLSAVI




HQLTKLMVEYNAVVVMEDLNFGFKRGRFHVEKQVYQKFEHILIDKS




NYLVFKDRGLNEPGGVLNGYQIAGQFESFQKLGKQSGMLFYVPAGY




TSKIDPKTGFVSMMNFKDLTNVHKKRDFFSKFDNIHYDEANGSFVFT




FDYKKFDGKAKEEMKLTKWSVYSRDKRIVYFAKTKSYEDVLPTEKL




QKIFESNGIDYKSGNNIQDSVMAIGADLKEGAKPSKEISDFWDGLLS




NFKLILQMRNSNARTGEDYIISPVMADDGTFFDSREEFKKGEDAKLP




LDADANGAYHIALKGLSLINKINLSKDEELKKFDMKISNADWFKFAQ




EKNYAK





SEQ
Cas12
MEEKKMSKIEKFIGKYKISKTLRFRAVPVGKTQDNIEKKGILEKDKK


ID
Variant
RSEDYEKVKAYLDSLHRDFIENTLKKVKLNELNEYACLFFSGTKDD


NO:

GDKKKMEKLEEKMRKTISNEFCNDEMYKKIFSEKILSENNEEDVSDI


42

VSSYKGFFTSLNGYVNNRKNLYVSDAKPTSIAYRCINENLPKFLRNV




ECYKKVVQVIPKEQIEYMSNNLNLSPYRIEDCFNIDFFEFCLSQGGIDL




YNTFIGGYSKKDGTKVQGINEIVNLYNQKNKKDKEKYKLPQFTPLFK




QILSDRDTKSFSIEKLENIYEVVELVKKSYSDEMFDDIETVFSNLNYY




DASGIYVKNGPAITHISMNLTKDWATIRNNWNYEYDEKHSTKKNKN




IEKYEDTRNTMYKKIDSFTLEYISRLVGKDIDELVKYFENEVANFVM




DIKKTYSKLTPLFDRCQKENFDISEDEVNDIKGYLDNVKLLESFMKSF




TINGKENNIDYVFYGKFTDDYDKLHEFDHIYNKVRNYITTSRKPYKL




DKYKLYFDNPQLLGGWDINKEKDYRTVMLTKDGKYYFAIIDKGEHP




FDNIPKDYFDNNGYYKKIIYRQIPNAAKYLSSKQIVPQNPPEEVKRIL




DKKKADSKSLTEEEKNIFIDYIKSDFLKNYKLLFDKNNNPYFNFAFRE




SSTYESLNEFFEDVERQAYSVRYENLPADYIDNLVNEGKIYLFEIYSK




DFSEYSKGTNNLHTMYFKALFDNDNLKNTVFKLSGNAELFIRPASIK




KDELVIHPKNQLLQNKNPLNPKKQSIFDYDLVKDKRFFENQYMLHIS




IEINKNERDAKKIKNINEMVRKELKDSDDNYIIGIDRGERNLLYVCVI




NSAGKIVEQMSLNEIINEYNGIKHTVDYQGLLDKCEKERNAQRQSW




KSIENIKELKDGYISQVVHKLCQLVEKYDAIIAMENLNGGFKRGRTK




FEKQVYQKFENKLINKMEYMADKKRKTTENGGILRGYQLTNGCINN




SYQNGFIFYVPAWLTSKIDPTTGFVDLLKPKYTNVEEAHLWINKFNSI




TYDKKLDMFAFNINYSQFPRADIDYRKIWTFYTNGYRIETFRNSEKN




NEFDWKEVHLTSVIKKLLEEYQINYISGKNIIDDLIQIKDKPFWNSFIK




YIRLTLQMRNSITGRTDVDYIISPVINNEGTFYDSRKDLDEITLPQDAD




ANGAYNIARKALWIIEKLKESPDEELNKVKLAITQREWLEYAQINI





SEQ
Cas12
MIIHNCYIGGSFMKKIDSFTNCYSLSKTLRFKLIPIGATQSNFDLNKML


ID
Variant
DEDKKRAENYSKAKSIIDKYHRFFIDKVLSSVTENKAFDSFLEDVRA


NO:

YAELYYRSNKDDSDKASMKTLESKMRKFIALALQSDEGFKDLFGQN


43

LIKKTLPEFLESDTDKEIIAEFDGFSTYFTGFFNNRKNMYSADDQPTAI




SYRCINDNLPKFLDNVRTFKNSDVASILNDNLKILNEDFDGIYGTSAE




DVFNVDYFPFVLSQKGIEAYNSILGGYTNSDGSKIKGLNEYINLYNQ




KNENIHRIPKMKQLFKQILSERESVSFIPEKFDSDDDVLSSINDYYLER




DGGKVLSIEKTVEKIEKLFSAVTDYSTDGIFVKNAAELTAVCSGAFG




YWGTVQNAWNNEYDALNGYKETEKYIDKRKKAYKSIESFSLADIQK




YADVSESSETNAEVTEWLRNEIKEKCNLAVQGYESSKDLISKPYTES




KKLFNNDNAVELIKNALDSVKELENVLRLLLGTGKEESKDENFYGEF




LPCYERICEVDSLYDKVRNYMTQKLYKTDKIKLNFQNPQFLGGWDR




NKEADYSAVLLRRNSLYYIAIMPSGYKRVFEKIPAPKADETVYEKVI




YKLLPGPNKMLPKVFFSKKGIETFNPPKEILEKYELGTHKTGDGFNL




DDCHALIDYFKSALDVHSDWSNFGFRFSDTSTYKNIADFYNEVKNQ




GYKITFCDVPQSYINELVDEGKLYLFQLYNKDFSEHSKGTPNLHTLY




FKMLFDERNLENVVFKLNGEAEMFYREASISKDDMIVHPKNQPIKN




KNEQNSRKQSTFEYDIVKDRRYTVDQFMLHIPITLNFTANGGTNINN




EVRKALKDCDKNYVIGIDRGERNLLYICVVDSEGRIIEQYSLNEIINEY




NGNTYSTDYHALLDKKEKERLESRKAWKTVENIKELKEGYISQVVH




KICELVEKYDAVIVMEDLNLGFKQGRSGKFEKSVYQKFEKMLIDKL




NYFADKKKSPEEIGSVLNAYQLTNAFESFEKMGKQNGFIFYVPAYLT




SKIDPTTGFADLLHPSSKQSKESMRDFVGRFDSITENKTENYFEFELD




YNKFPRCNTDYRKKWTVCTYGSRIKTFRNPEKNSEWDNKTVELTPA




FMALFEKYSIDVNGDIKAQIMSVDKKDFFVELIGLLRLTLQMRNSET




GKVDRDYLISPVKNSEGVFYNSDDYKGIENASLPKDADANGAYNIA




RKGLWIIEQIKACENDAELNKIRLAISNAEWLEYAQKK





SEQ
Cas12
MKEQFVNQYPISKTLRFSLIPIGKTEENFNKNLLLKEDEKKAEEYQKV


ID
Variant
KGYIDRYHKFFIETALCNINFEGFEEYSLLYYKCSKDDNDLKTMEDIE


NO:

IKLRKQISKTMTSHKLYKDLFGENMIKTILPNFLDSDEEKNSLEMFRG


44

FYTYFSGFNTNRKNMYTEEAKSTSIAYRCINDNLPKFLDNSKSFEKIK




CALNKEELKAKNEEFYEIFQIYATDIFNIDFFNFVLTQPGIDKYNGIIG




GYTCSDGTKVQGLNEIINLYNQQIAKDDKSKRLPLLKMLYKQILSDR




ETVSFIPEKFSSDNEVLESINNYFSKNVSNAIKSLKELFQGFEAYNMN




GIFISSGVAITDLSNAVFGDWNAISTAWEKAYFETNPPKKNKSQEKY




EEELKANYKKIKSFSLDEIQRLGSIAKSPDSIGSVAEYYKITVTEKIDNI




TELYDGSKELLNCNYSESYDKKLIKNDTVIEKVKTLLDAVKSLEKLI




KPLVGTGKEDKDELFYGTFLPLYTSLSAVDRLYDKVRNYATQKPYS




KDKIKLNFNCSSFLSGWATDYSSNGGLIFEKDGLYYLGIVNKKFTTE




EIDYLQQNADENPAQRIVYDFQKPDNKNTPRLFIRSKGTNYSPSVKE




YNLPVEEIVELYDKRYFTTEYRNKNPELYKASLVKLIDYFKLGFTRH




ESYRHYDFKWKKSEEYNDISEFYKDVEISCYSLKQEKINYNTLLNFV




AENRIYLFQIYNKDFSKYSKGTPNLHTRYFKALFDENNLSDVVFKLN




GGSEMFFRKASIKDNEKVVHPANQPIDNKNPDNSKKQSTFDYELIKD




KRFTKHQFSIHIPITMNFKARGRDFINNDIRKAIKSEYKPYVIGIDRGE




RNLIYISVINNNGEIVEQMSLNDIISDNGYKVDYQRLLDRKEKERDNA




RKSWGTIENIKELKEGYISQVIHKICELVIKYDAVIAMEDLNFGFKRG




RFNVEKQVYQKFENMLISKLNYLCDKKSEANSEGGLLKAYQLTNKF




DGVNKGKQNGIIFYVPAWLTSKIDPVTGFVDLLHPKYISVEETHSLFE




KLDDIRYNFEKDMFEFDIDYSKLPKCNADFKQKWTVCTNADRIMTF




RNSEKNNEWDNKRILLSDEFKRLFEEFGIDYCHNLKNKILSISNKDFC




YRFIKLFALTMQMRNSITGSTNPEDDYLISPVRDENGVFYDSRNFIGS




KAGLPIDADANGAYNIARKGLWAINAIKSTADDMLDKVDLSISNAK




WLEYVQK





SEQ
Cas12
MADLSQFTHKYQVPKTLRFELIPQGKTLENLSAYGMVADDKQRSEN


ID
Variant
YKKLKPVIDRIYKYFIEESLKNTNLDWNPLYEAIREYRKEKTTATITN


NO:

LKEQQDICRRAIASRFEGKVPDKGDKSVKDFNKKQSKLFKELFGKEL


45

FTDSVLEQLPGVSLSDEDKALLKSFDKFTTYFVGFYDNRKNVFSSDD




ISTGIPHRLVQENFPKFIDNCDDYKRLVLVAPELKEKLEKAAEATKIF




EDVSLDEIFSIKFYNRLLQQNQIDQFNQLLGGIAGAPGTPKIQGLNETL




NLSMQQDKTLEQKLKSVPHRFSPLYKQILSDRSSLSFIPESFSCDAEVL




LAVQEYLDNLKTEHVIEDLKEVFNRLTTLDLKHIYVNSTKVTAFSQA




LFGDWNLCREQLRVYKMSNGNEKITKKALGELESWLKNSDIAFTEL




QEALADEALPAKVNLKVQEAISGLNEQMAKSLPKELKIPEEKEELKA




LLDAIQEVYHTLEWFIVSDDVETDTDFYVPLKETLQIIQPIIPLYNKVR




NFATQKPYSVEKFKLNFANPTLADGWDENKEQQNCAVLFQKGNNY




YLGILNPKNKPDFDNVDTEKQGNCYQKMVYKQFPDFSKMMPKCTT




QLKEVKQHFEGKDSDYILNNKNFIKPLTITREVYDLNNVLYDGKKKF




QIDYLRKTKDEDGYYTALHTWIDFAKKFVASYKSTSIYDTSTILPPEK




YEKLNEFYGALDNLFYQIKFENIPEEIIDTYVEDGKLFLFQIYNKDFA




AGATGAPNLHTIYWKAVFDPENVKDVVVKLNGQAELFYRPKSNMD




VIRHKVGEKLVNRTLKDGSILTDELHKELYLYANGSLKKGLSEDAKI




ILDKNLAVIYDVHHEIVKDRRFTTDKFFFHVPLTLNYKCDKNPVKFN




AEVQEYLKENPDTYVIGIDRGERNLIYAVVIDPKGRIVEQKSENVING




FDYHGKLDQREKERVKARQAWTAVGKIKELKQGYLSLVVHEISKM




MVRYQAVVVLENLNVGFKRVRSGIAEKAVYQQFEKMLINKLNYLM




FKDAGGTEPGSVLNAYQLTDRFESFAKMGLQTGFLFYIPAAFTSKIDP




ATGFVDPFRWGAIKTLADKREFLSGFESLKFDSTTGNFILHFDVSKNK




NFQKKLEGFVPDWDIIIEANKMKTGKGATYIAGKRIEFVRDNNSQGH




YEDYLPCNALAETLRQCDIPYEEGKDILPLILEKNDSKLLHSVFKVVR




LTLQMRNSNAETGEDYISSPVEDVSGSCFDSRMENEKLPKDADANG




AYHIALKGMLALERLRKDEKMAISNNDWLNYIQEKRA





SEQ
Cas12
MTNFDNFTKKYVNSKTIRLEAIPVGKTLKNIEKMGFIAADRQRDEDY


ID
Variant
QKAKSVIDHIYKAFMDDCLKDLFLDWDPLYEAVVACWRERSPEGR


NO:

QALQIMQADYRKKIADRFRNHELYGSLFTKKIFDGSVAQRLPDLEQS


46

AEEKSLLSNFNKFTSYFRDFFDKRKRLFSDDEKHSAIAYRLINENFLK




FVANCEAFRRMTERVPELREKLQNTGSLQVYNGLALDEVFSADFYN




QLIVQKQIDLYNQLIGGIAGEPGTPNIQGLNATINLALQGDSSLHEKL




AGIPHRFNPLYKQILSDVSTLSFVPSAFQSDGEMLAAVRGFKVQLESG




RVLQNVRRLFNGLETEADLSRVYVNNSKLAAFSSMFFGRWNLCSDA




LFAWKKGKQKKITNKKLTEIKKWLKNSDIAIAEIQEAFGEDFPRGKIN




EKIQAQADALHSQLALPIPENLKALCAKDGLKSMLDTVLGLYRMLQ




WFIVGDDNEKDSDFYFGLGKILGSLDPVLVLYNRVRNYITKKPYSLT




KFRLNFDNSQLLNGWDENNLDTNCASIFIKDGKYYLGISNKNNRPQF




DTVATSGKSGYQRMVYKQFANWGRDLPHSTTQMKKVKKHFSASD




ADYVLDGDKFIRPLIITKEIFDLNNVKFNGKKKLQVDYLRNTGDREG




YTHALHTWINFAKDFCACYKSTSIYDISCLRPTDQYDNLMDFYADLG




NLSHRIVWQTIPEEAIDNYVEQGQLFLFQLYNKDFAPGADGKPNLHT




LYWKAVFNPENLEDVVVKLNGKAELFYRPRSNMDVVRHKVGEKLV




NRKLKNGLTLPSRLHEEIYRYVNGTLNKDLSADARSVLPLAVVRDV




QHEIIKDRRFTADKFFFHASLTFNFKSSDKPVGFNEDVREYLREHPDT




YVVGVDRGERNLIYIVVIDPQGNIVEQRSFNMINGIDYWSLLDQKEK




ERVEAKQAWETVGKIKDLKCGYLSFLIHEITKIIIKYHAVVILENLSLG




FKRVRTGIAEKAVYQQFERMLVTKLGYVVFKDRAGKAPGGVLNAY




QLTDNTRTAENTGIQNGFLFYVPAAFTSRVDPATGFFDFYDWGKIKT




ATDKKNFIAGFNSVRYERSTGDFIVHVGAKNLAVRRVAEDVRTEWD




IVIEANVRKMGIDGNSYISGKRIRYRSGEQGHGQYENHLPCQELIRAL




QQYGIQYETGKDILPAILQQDDAKLTDTVFDVFRLALQMRNTSAETG




EDYFNSVVRDRSGRCFDTRRAEAAMPKEADANDAYHIALKGLFVLE




KLRKGESIGIKNTEWLRYVQQRHS





SEQ
Cas12
MENYGGFTGLYPLQKTLKFELRPQGRTMEHLVSSNFFEEDRDRAEK


ID
Variant
YKIVKKVIDNYHKDFINECLSKRSFDWTPLMKTSEKYYASKEKNGK


NO:

KKQDLDQKIIPTIENLSEKDRKELELEQKRMRKEIVSVFKEDKRFKYL


47

FSEKLFSILLKDEDYSKEKLTEKEILALKSFNKFSGYFIGLHKNRANFY




SEGDESTAIAYRIVNENFPKFLSNLKKYREVCEKYPEIIQDAEQSLAG




LNIKMDDIFPMENFNKVMTQDGIDLYNLAIGGKAQALGEKQKGLNE




FLNEVNQSYKKGNDRIRMTPLFKQILSERTSYSYILDAFDDNSQLITSI




NGFFTEVEKDKEGNTFDRAVGLIASYMKYDLSRVYIRKADLNKVSM




EIFGSWERLGGLLRIFKSELYGDVNAEKTSKKVDKWLNSGEFSLSDV




INAIAGSKSAETFDEYILKMRVARGEIDNALEKIKCINGNFSEDENSK




MIIKAILDSVQRLFHLFSSFQVRADFSQDGDFYAEYNEIYEKLFAIVPL




YNRVRNYLTKNNLSMKKIKLNFKNPALANGWDLNKEYDNTAVIFL




REGKYYLGIMNPSKKKNIKFEEGSGTGPFYKKMAYKLLPDPNKMLP




KVFFAKKNINYYNPSDEIVKGYKAGKYKKGENFDIDFCHKLIDFFKE




SIQKNEDWRAFNYLFSATESYKDISDFYSEVEDQGYRMYFLNVPVA




NIDEYVEKGDLFLFQIYNKDFASGAKGNKDMHTIYWNAAFSDENLR




NVVVKLNGEAELFYRDKSIIEPICHKKGEMLVNRTCFDKTPVPDKIH




KELFDYHNGRAKTLSIEAKGYLDRVGVFQASYEIIKDRRYSENKMYF




HVPLKLNFKADGKKNLNKMVIEKFLSDKDVHIIGIDRGERNLLYYSV




IDRRGNIIDQDSLNIIDGFDYQKKLGQREIERREARQSWNSIGKIKDLK




EGYLSKAVHKVSKMVLEYNAIVVLEDLNFGFKRGRFKVEKQVYQK




FEKMLIDKLNYLVFKEVLDSRDAGGVLNAYQLTTQLESFNKLGKQS




GILFYVPAAYTSKIDPTTGFVSLFNTSRIESDSEKKDFLSGFDSIVYSA




KDGGIFAFKFDYRNRNFQREKTDHKNIWTVYTNGDRIKYKGRMKG




YEITSPTKRIKDVLSSSGIRYDDGQELRDSIIQSGNKVLINEVYNSFIDT




LQMRNSDGEQDYIISPVKNRNGEFFRTDPDRRELPVDADANGAYHIA




LRGELLMQKIAEDFDPKSDKFTMPKMEHKDWFEFMQTRGD





SEQ
Cas12
MLHAFTNQYQLSKTLRFGATLKEDEKKCKSHEELKGFVDISYENMK


ID
Variant
SSATIAESLNENELVKKCERCYSEIVKFHNAWEKIYYRTDQIAVYKD


NO:

FYRQLSRKARFDAGKQNSQLITLASLCGMYQGAKLSRYITNYWKDN


48

ITRQKSFLKDFSQQLHQYTRALEKSDKAHTKPNLINFNKTFMVLANL




VNEIVIPLSNGAISFPNISKLEDGEESHLIEFALNDYSQLSELIGELKDAI




ATNGGYTPFAKVTLNHYTAEQKPHVFKNDIDAKIRELKLIGLVETLK




GKSSEQIEEYFSNLDKFSTYNDRNQSVIVRTQCFKYKPIPFLVKHQLA




KYISEPNGWDEDAVAKVLDAVGAIRSPAHDYANNQEGFDLNHYPIK




VAFDYAWEQLANSLYTTVTFPQEMCEKYLNSIYGCEVSKEPVFKFY




ADLLYIRKNLAVLEHKNNLPSNQEEFICKINNTFENIVLPYKISQFETY




KKDILAWINDGHDHKKYTDAKQQLGFIRGGLKGRIKAEEVSQKDKY




GKIKSYYENPYTKLTNEFKQISSTYGKTFAELRDKFKEKNEITKITHF




GIIIEDKNRDRYLLASELKHEQINHVSTILNKLDKSSEFITYQVKSLTS




KTLIKLIKNHTTKKGAISPYADFHTSKTGFNKNEIEKNWDNYKREQV




LVEYVKDCLTDSTMAKNQNWAEFGWNFEKCNSYEDIEHEIDQKSYL




LQSDTISKQSIASLVEGGCLLLPIINQDITSKERKDKNQFSKDWNHIFE




GSKEFRLHPEFAVSYRTPIEGYPVQKRYGRLQFVCAFNAHIVPQNGE




FINLKKQIENFNDEDVQKRNVTEFNKKVNHALSDKEYVVIGIDRGLK




QLATLCVLDKRGKILGDFEIYKKEFVRAEKRSESHWEHTQAETRHIL




DLSNLRVETTIEGKKVLVDQSLTLVKKNRDTPDEEATEENKQKIKLK




QLSYIRKLQHKMQTNEQDVLDLINNEPSDEEFKKRIEGLISSFGEGQK




YADLPINTMREMISDLQGVIARGNNQTEKNKIIELDAADNLKQGIVA




NMIGIVNYIFAKYSYKAYISLEDLSRAYGGAKSGYDGRYLPSTSQDE




DVDFKEQQNQMLAGLGTYQFFEMQLLKKLQKIQSDNTVLRFVPAFR




SADNYRNILRLEETKYKSKPFGVVHFIDPKFTSKKCPVCSKTNVYRD




KDDILVCKECGFRSDSQLKERENNIHYIHNGDDNGAYHIALKSVENL




IQMK





SEQ
Cas12
MKNGINLFKTKTTKTKGVDMEKYQITKTIRFKLLPDNAHEIVEKVKS


ID
Variant
LKTSNVDELMDEVKNVHLKGLELLFALKKYFYFDGNQCKSFKSTLE


NO:

IKARWLRLYTPDQYYLKKSSKNSYQLKSLSYFKDVFNDWLFNWEES


49

VSELAIIYEKYKICQHQRDSRADIALLIKKLSMKEYFPFISDLIDCVND




KNSNKTFLMKLSEELSVLLEKCNSRALPYQSNGIVVGKASLNYYTVS




KSEKMLQNEYEDVCQSLDKNYDITEMKVILYKEKLDNLNFKDVTIA




NAYNLLKENKALQKRLFSEYVSQGKVLSLIKTELPLFSNINDNDFEK




YKEWSNEIKKLADKKNTFCKKTQQDKIKDIQNKISELKKKRGALFQ




YKFTSFQKHCDNYKKVAVQYGKLKARKKAIEKDEIEANLLRYWSVI




LEQEDKHSLVLIPKNNAKDAKQYIETINTKGGKYIIHHLDSLTLRALN




KLCFNAVDIEKGQMVRENTFYQGIKEEFERNKINCDNQGVLKIQGLY




SFKTEGGQINEKEAVEFFKEVLKSNYAREVLNLPYDLESNIFQKEYT




NLDQFRQDLEKCCYALHSKIGKDDLDEFTRRFEAQVFDITSIDLKSK




KEKTKTTGEMKKHTQLWLEFWKGAIEQNFATRVNPELSIFWRAPKS




SREKKYGKGSDLYDPNKNNRYLYEQYTLALTITENAGSHFKDIAFK




DTSKIKEAIKEFNMSLSQSKYCFGIDRGNAELVSLCLIKNEKDFPFEKF




PVYRLRDLTYQGDFKDKHDQMRYGVAIKNISYFIDQEDLFEKNNLS




AIDMTTAKLIKNKIVLNGDVLTYLKLKEETAKHKLTQFFQGSSINKN




SRVYFDEDENVFKITTNRNHNPEEIIYFYRGEYGAIKNKNDLEDILNE




YLCKMETGESEIVLLNRVNHLRDAISANIVGILSYLIDLFPETIVALEN




LAKGTIDRHVSQSYENITRRFEWALYRKLLNKQLAPPELKENILLRE




GDDKIDQFGIIHFVEEKNTSKDCPNCRKTTQQTNDNKFKEKKFVCKS




CGFDTSKDRKGMDSLNSPDTVAAYNVARKKFES





SEQ
Cas12
MAKETKEFKTFDDFTNLYEVQKTLRFELEAVPETEIVLENRGIWYKR


ID
Variant
DKKRADEKPIVKFYMDILHREFTDEALEKIKESGVLNLSGYFKLFEEL


NO:

RRLQNHGANTKEEKKLKLEEIRAKKREISNELSQIRRVFSVRGFDVV


50

DSDWKKKYTIEGKKIKNDKSKTYLILSENILNFLENRFTSKEVERLRSI




DKKHVEDYGNVVNSGGENIFATFKGFFGYFDSLIKNRENFYETDGK




AGRVATRSVDENLNFFAENLHIFSTDLPKALKDDLSDTQKAIFERSY




YKNCLLQKDIKSYNLIIGDINKEINKHRQQRDTKIKFLNTLFKQILSIE




EKEQYKHIEINNDEDLIRAIRDFISLNESKISEGTKIFNQFIQRCLQKED




LGQIYLPKDSVNTIAHRIFKPWDEIMALFDRKYFVSLEEIKDLTESSV




WKERVLEESKTKSLIFKDTHIHTIISGQEIFSNFILILEKEYKNQFSGFIS




ETRRGKAAFVGYDESLKNLRATIKWFEGKNLKLSETEKVEWIKAIK




DYADAALRIFQMTKYLWLPVVGDEEDKDYLRIKAEIDQLTKDNDFY




NKINAFIDGYKPEPFIYRSSFQEYLTRRPFSTDKFKINFENSRLLDGWD




KDMIDDRMGILLQRDGDYFLGILNKEDRHCLDNLVDVKSEDKNSYA




LMQFKQLTGLYRQLPRMAFPKKKQPVLEANAEIKKIKEDFDFLQKQ




KKEREVNVNVVFDNKKLNLLINHYAEFLKENYKDEKCYDFSLLNKE




KVYESLSDFYADVDKITYSLSFIQVSIDQLIKTGKILLFRLKNKDLLKG




SLGQNKNLHTYYFHALFERENLSQGRIRLGAQAEIFFRPASIEKEKDK




NRSNALKKSPKTRYVKEILKNKRYSEDKVFLHLPIQLNADAYDLPSI




NQNVFEFIKNRQEKVKIIGIDRGEKNLAYYSVISQNSNGKIKIEEPPRD




LNLGYLEPLDELENKRQDERKAWQSISEIKSKRDGYISYAVSKIVEL




MLKYQAIIVLEDLSGKFKRSRMKFEKAPYQQLELALIKKLNYLVKK




NSKSGKPGHYLSAYQLTEPVGSYKEMGKQTGIIFYTQAGYTSRTCPT




CGWRKRVQGLYYKDRTSAQRRFDPKTGVKIFYDSVNDRFVFQYHP




VYEQKELKEWDKEIYSDVTRIRWNNEEKKNNEYRKGDITLKIKRLFR




DRGIDLSRNINEQLVNVGDASFWEELINLLRLITEIRNIDNENNRDFIE




CPHCHFQSENGFHGVAWNGDANGAYNIARKGLLITKAVCDPEKNV




GDITWSDLKVDMKDWDAATDEWAKKNPEK





SEQ
Cas12
MENEKIFSDLTNRYQVVKTLPFELKPVPRTRVLLGLDNPNKGEIFSK


ID
Variant
DRERAENFTIIKKYIDRLHSLFINESLKKADIDFSNFYKQYGKNINTKN


NO:

NKNIDDDNDINDDEKEDSENDNLKKYRQEIANLFNKSKYKSWVNVG


51

KDGDKISGMLFEKGLIDLLRTHFSDNLNEDIEIPELFSNKKIKDTRKLK




EIINSFGKDGKDGQNFTTYFSVSFHNNRKNYYKSDGKMGRVSTRIVD




ENLERFCKNIYLYKEIIGKNEIKEIFSGNWDIYLQKKPNFSNDKTYKK




LDEFKNDKYDWEMIFRDVNSYNKYFLQSDIEFYNYIRGKLNQDINEY




NGKKRDSKEKINSQFENLRNQVHGEKKNYDDDFEIDEDNIIQFINEIF




VRHNQNKMRFSEKLFSDFIDLLMVDNGDKLDKVYFSQKAVENAIAR




YYFVEETTNEGREPLLISLLLQNAGKDRKKLSNKPIKLGDIKFVLDQA




NNKPAEDIFKNRYVLSESNNDGIINANDKNHWANLLRLIKKDFYFHK




DNLIKSQDKLALETKYNKGSDEGERQIETIKNFAESAKAILRMTKYF




DLRKNGVIQNVIGGKDPIHEEVDKYFDGDVLSGEESCRISKYYDALR




NFITKKAWSADKIILNFDCSEFLGGWDRSQEQKKRGIILRHRDGDEE




RYYLAVLGKNGKQYFENRTLFKGCESSDWQKIEYNVIQKPHMSLPK




NLITPFFKKDKITNERFIDRSKKGAKALIEIDINPSDEFLNNYNLGKHT




KENLDKSFLCDYFKYLMDAIAKYYKGEFNFNFPDVSNFDNTQPFYSF




IEKNAYSIKYFGISSKEIEKLIADCYYKEDVYLFQIYCKDFEIDPKIGK




AKYGNEFRTKAEIRKSKGEEAGNENLNTKYFKLLFDEKNLKNQNGI




VYKLNGGAKMFYRPSSIKKDEKIDGKWRYKEDKYSLNITITCNFSSK




KDDLSIDKDINKKIAEVNANSDFRIISIDRGEKNLAYCCVMDENANIL




DIKSLNRITRYDKNGKAIKEKNMFHEVKDGKLCYGEPVYDFYKDYQ




NLLDEREIKRLVNRRSWNVIEDIKNLKKGYVALLINYICKAVVIAINE




GKYPIIVLESLDKGMLHNRVKIEKQIYRGVEEGLVRKLNYFVDKKTD




NVLNAWQLLAKFETVGSSLDRKKQLGIIFYVDPGYTSITCPCCGFRQ




RKYIKAERAEENFKEIKIKFDGKRYSFAYDYRCIDDNGKEKSKEDIIY




SNVKRLLRSGRNGRAVQIEDVTDELTNLFKKHNINIEQDINEQLAGK




DNKFWKQLLWWFNAIEQIRNTQSLRRKFNTEENKLEILENNDCDFIL




CPHCYFDSNKDKFQNKIWNGDANGAFNIGRKGIIDIFEIKKHQRMLS




DFMEQWGIDKLPKANGGNQAVIEIVKNDKKYNLCILNNKKIPYYCL




RIGKEKIDSIADDRKCNQLPDLMVNWKKWDMWLDKWGK





SEQ
Cas12
MPEVKNVFQDFTNLYELSKTLRFELKPVPETEKILELNAAKTKKFPK


ID
Variant
DLYRAENFEIIKKYTDELHRTYIRETLNNVNIDYLKFLEIFRINGKKKN


NO:

EMTDENEESDENNEKDDIQKIKKELRSKIGNLFNKWNNDKDNKFKD


52

WVKIDVGKKEKEVSGDLFGKELITILKNYFKNKLDSKVNVPMLFFNE




QEIKNGEAKKQRKLEAVFENFDKFTTYFTDSFYNNRKNYYKTEGRV




GQVATRIIDENLPRFCSNLIAFNEVVSLYSTLLNNFDLGWKEYLNEK




KINQTWVEKFELSNYDWKALFNDVNYYNQCLLQEGIDKYNYIIKKL




NKDINEYTQNKYKSVEKGNNNNPDINFFQKLHKQIHGERDFKLIEIDI




DENNIFTKILPEFILHSDMKLMTKIDEEVGVEEIVGAERIIKIFIKQELK




DLEKIYLSRRAIETISAKWFHSWETLKDLILGYLNKDLLESKKRKKVP




DFVDFNIIKIVLENNKDDYKDLFKRKYFEADKNEFVDWIDSSGGTKK




LEFGGENWINFLNVFEYEFGTLLTEYKKNKNALLYLIDKKIDYDKNN




EVGQTAAIKNFADSALGIFRMVSYFALRKKGVMVEPKNGKDEIFYA




FVDRYLDGDDNDREEQNKIVQYYNTLRNFVTQKAWSIDKVRLCFDC




GEFLKGWDKDKIHERLGIILRNNNKFYLGILNKNHKQIFIKIKSHDNN




NFYYVIYDYKQLNNVYRQIPRLAFPSRSVKKGDAYMLRAIQERKKK




FFLEDEEFIELQEIKNEYDKIGNDLSKEKLTKLIEYYKKVVISNYSSLY




NVSNLNNKKFNSINEFNQYVENLMYSLIPTRISPDFIKEKISKGELYLF




QIYNKDFELDESIGKEKFGEDFAPVIMDGKNNLHTEYFKLLFNDSNL




KNPNGVVFKLSGGAKMFYRPATENLPIKKDRDGNIIKNKKGENVIVG




QRYKEDKYFLHLPIILNFVNKGKNYSINDMVNKAITNASDDQDKFRII




GLDRGEKHLVYYSVINERQEIIEIGSLNNISRKDNKGEIIEEKNWYHD




KFGNIEKEPTKEYHKDYHNLLDQREIERLKSRQSWEKIENIKELKEG




YISAVINKICNLVIKAIKENKIPIVALENLNSGMKRGRIKIDKQIYQKL




ELKLAKKLNFLVDKKEKNYLSAWQFTPKIETFSGDIEKKNQVGIIFY




VDPAFTSATCPNCGFRKRIKMDPQNAKKKIKDMEITYENGIYKFDYP




IENGENDVVYSDVERLKWDNEKKKVIKTKNVSDDFGKLFEDIKDKN




NLKKELLSIGEENKEFWKEFSRCFNLLLRIRNSKLIKRKLNDDTGKVE




IIADDDLADRDRDFIYCPQCHFHSEGGDVFGEFVKKKYLGKDNFEFN




GDANGAYNIARKTIIAVNKIKDYQLGLNHFIEKYRISELPNNGKDKK




NIFYNNNSYILSFFEVQDEKFRKVKVYGLKKDGDRQIIQKKEMWYR




RYPDIFVNNKEWDKFVQNKS





SEQ
Cas12
MLFFMSTDITNKPREKGVFDNFTNLYEFSKTLTFGLIPLKWDDNKKM


ID
Variant
IVEDEDFSVLRKYGVIEEDKRIAESIKIAKFYLNILHRELIGKVLGSLKF


NO:

EKKNLENYDRLLGEIEKNNKNENISEDKKKEIRKNFKKELSIAQDILL


53

KKVGEVFESNGSGILSSKNCLDELTKRFTRQEVDKLRRENKDIGVEY




PDVAYREKDGKEETKSFFAMDVGYLDDFHKNRKQLYSVKGKKNSL




GRRILDNFEIFCKNKKLYEKYKNLDIDFSEIERNFNLTLEKVFDFDNY




NERLTQEGLDEYAKILGGESNKQERTANIHGLNQIINLYIQKKQSEQK




AEQKETGKKKIKFNKKDYPTFTCLQKQILSQVFRKEIIIESDRDLIREL




KFFVEESKEKVDKARGIIEFLLNHEENDIDLAMVYLPKSKINSFVYKV




FKEPQDFLSVFQDGASNLDFVSFDKIKTHLENNKLTYKIFFKTLIKEN




HDFESFLILLQQEIDLLIDGGETVTLGGKKESITSLDEKKNRLKEKLG




WFEGKVRENEKMKDEEEGEFCSTVLAYSQAVLNITKRAEIFWLNEK




QDAKVGEDNKDMIFYKKFDEFADDGFAPFFYFDKFGNYLKRRSRNT




TKEIKLHFGNDDLLEGWDMNKEPEYWSFILRDRNQYYLGIGKKDGE




IFHKKLGNSVEAVKEAYELENEADFYEKIDYKQLNIDRFEGIAFPKKT




KTEEAFRQVCKKRADEFLGGDTYEFKILLAIKKEYDDFKARRQKEK




DWDSKFSKEKMSKLIEYYITCLGKRDDWKRFNLNFRQPKEYEDRSD




FVRHIQRQAYWIDPRKVSKDYVDKKVAEGEMFLFKVHNKDFYDFE




RKSEDKKNHTANLFTQYLLELFSCENIKNIKSKDLIESIFELDGKAEIR




FRPKTDDVKLKIYQKKGKDVTYADKRDGNKEKEVIQHRRFAKDAL




TLHLKIRLNFGKHVNLFDFNKLVNTELFAKVPVKILGMDRGENNLIY




YCFLDEHGEIENGKCGSLNRVGEQIITLEDDKKVKEPVDYFQLLVDR




EGQRDWEQKNWQKMTRIKDLKKAYLGNVVSWISKEMLSGIKEGVV




TIGVLEDLNSNFKRTRFFRERQVYQGFEKALVNKLGYLVDKKYDNY




RNVYQFAPIVDSVEEMEKNKQIGTLVYVPASYTSKICPHPKCGWRER




LYMKNSASKEKIVGLLKSDGIKISYDQKNDRFYFEYQWEQEHKSDG




KKKKYSGVDKVFSNVSRMRWDVEQKKSIDFVDGTDGSITNKLKSLL




KGKGIELDNINQQIVNQQKELGVEFFQSIIFYFNLIMQIRNYDKEKSGS




EADYIQCPSCLFDSRKPEMNGKLSAITNGDANGAYNIARKGFMQLC




RIRENPQEPMKLITNREWDEAVREWDIYSAAQKIPVLSEEN





SEQ
Cas12
MTIKKHKPFTNFECLTPVQKTLRFRLIPVGRTTEFVKCRNIIEADRKRS


ID
Variant
EMYPLLKELADRFYREFMTDQLSNLLFDWSPLVEALLLARNNTDPR


NO:

ENQRIASLVRDEQKKYRTLLLKRLSGQVDRNGTPLPKNTASVNKKY


54

YDDLFKARFVTETLPAYLEHLKNKPDGRISDELFDAYKDALDSYQKF




TSRLTNFWQARKNIFTDEDIATGFAYRIVHEIVPDYLFNRRVYEQHK




LDFPEPLDLLETELKKKNLIANDESLDALFTIPAINRLLTQKGVDLHN




AVIGGFFTDDHTKVQGFNELANLKNQTLKNVSDNSEIKPVGKMTRL




KKHILSISESTSFLFEQIESDDDLLARIIEFNNTLSEPDIDGLSIADINDQ




LYNIMTGVDPSTILVHARNLNKLSHEASLSWNRLRDGLYQMATESP




YREDERFKRYIDASEEERDLSKLKNDIYFSLQELQFALDQSIDLEEEA




TPTEDIFLPFEFPGMDLKSELTVLFRSIEQLISSETKLIGNPDAIATIKKY




LDAIMARYSIWNLLSCEAVELQDDLFYPEYDRVMGSLSNIILLYNLA




RNYLSRKPSSKEKFRLNFDKPTLADGWSESKVPDNFSVLLRKDDLFY




LGILKDRKAYRVLSYENCDETAKNIKGYYERMIYHFSPDAYRMIPKC




STARKDVKKHFGEQGETTGYTLYPGASNFVKPFTIPYEIYRLQTELV




NDKKRYQADYLKQTEDEEGYRQAVTAWIDFCKSYLESYEGTSTFDY




SHLLKSEDYEDVNQFYADVDRASYSIYFEKVSVDLIHTMVDRGDLY




LFQLYNKDFSPHSTGKPNLHTMYWRALFSNDNLQNNTIKLNGQAEL




FYRPKQVEQPTVHLQGSYLLNRFDKHGDVIPAGLYCEIYNHINERHP




EGYTLSEEATQGLLDGRFVYREAPFELVKDKRYTEDQLFLHVPLEFN




WTASANVPFENLANEYIKKDSDLHIIGIDRGERNLLYYSVINLQGDIV




KQGSLNTLIQQTTLKGETVERQIPYQSMLKQREDERAEARQNWQSID




RIKDLKEGYLSHVIYKLSRLIIKYHAIVVMENLNVGFKRGRFKVERQ




VYQKFEVALINKLNALSFKEYEPNELGGVMRPWQLARRVVSPEDTR




SQNGIVFYVPASYTSIVDPVTGFANLFYLNRIRNKDLNSFYGHFQEIR




YDHEFDRFIFRFNYADFGVFCRIKNVPSRTWNLVSGERKAFNPKRRM




IEKRDTTDEIKKALEAHGIAYQNEQNLLPLLLENENLLARIHRSFRLV




LQLRNSDSDRDDIVSPALDKENNTFDSGQQPYESSLPINADANGAYN




IARKGLLLVDKVKNDKRAVLSNREWFEYLMAEE





SEQ
Cas12
MENKDYSLSRFTKQYQNSKTVRFALTPIGRTEEYIIQNQYIEAARRKN


ID
Variant
QAYKIVKPIIDEKFRSMIDDVLTHCEKQDWVTLDKLILQYQNNKCRE


NO:

NMDALAEQQEEIRKNISEEFTKSDEYKNFFGKEDSKKLFKIFLPEYLN


55

QINASESDKEAVNEFQKFKTYFSNFLIVRADIFKADNKHNTIPYRIVN




ENFMIFAGNKRTFSNIIRLIPNALEEIAKDGMKKEEWSFYNIQNVDSW




FEPDSFQMCMSQKGIQKYNFIIGLVNSYINLYTQQNPQATEVKRSRL




KLRMLHKQILSDRVNPSWLPEQFKEGEEGEKQIYEAILALENDLIKN




CFDKKYDLWIQSIDIQNPRIYIAASEMARVSSALHMGWNGLNDVRK




TILLKSDKKQAKVEKILKQDVSLKDLSDTLNRYADIYKEEQIPSLYQY




IEYGSELLQDCAITRKEYHDLLNGNSNTLSLNQNEKLIEGLKAYLDS




YQAIVHFLNVFIVGDELDKDTDFYAELDGLVESLSEIVPLYNKVRNYI




TRKVYSLDKMRIMFERSDFLGGWGQSFDTKEALLFQKDNLYYIGIIE




KKYTNMDVEYLHEGIKEGNRAIRFIYNFQKADNKNIPRTFIRSKGTN




YAPAVRKYNLPIESIIDIYDVGKFKTNYKKINEKEYYESLEKLIDYFK




DGILKNENYKKFHFNWKPSNEYENINEFYNDTNNACFLLEKEEINYD




HLKEQANQGKIYLFQISSKDFNEGSKGTPNLQTMYWRELFSNQNCK




DGVIKLCGGASIYMRDASIKQPVVHRKNAWLINKWYKVNGQNVVIP




DNTYVKFTKIAQERMNEDELTPQERQLWNSGLIQKKKATHDIMKDR




RFTKKQYMLHAPLTINYKQQDSPRYFNEKVRSFLKDNPDINIIGIDRG




EKNLIYITIIDQKGNILKGMQKSFNQIEEKGKEGRTIDYYSKLESVEAR




HDAARKNWKQIGTIRELKEGYLSQVVHEITQLMIQYNAVIVMENLN




MGFKKGRMKVEKSVYQKFEKMLIDKMNYLAFKRDMQGNAIDPYE




VGGVMNGYQLTDRFTSFADMGSQNGFIFYVPAAYTSVIDPVTGFVN




VFQKTEFKTNDFLHRFDSISWNDKEQSFVFTFDYQNFKCNGTCYQN




KWSLYADVDRIETIIKNNQVDRIEPCNPNQKLIDFFDKKGIIYRDGHNI




VDDLEKYDSKTISEIIHNFKLILQLRNSMRNPDTGEIIDYIASPVMHNE




ERFDSRKRNPELPQDADANGAYHIALKGLMFLQKINEYADSDGNMD




NRKLKITNEEWFKYMQTRKEHTYF





SEQ
Cas12
MSNKTSSITTTNKLSYTGFHNNGKQSKTLMFELKPIGRTTEHLDRKG


ID
Variant
YLADDIDRAESYKTFKEIADNFHKNLIEESLATFTFSDTLKDYFDLWL


NO:

SPVRTNEDTPKLRKMEAKLRKELSSALKQHPSFAATSSGKRLIDEAL


56

YPNASDKERQCLDRFKGRSSYLDSYTEVRSFIYTDLCKHNTIAYRVV




NENLKIYLENILAYEKLMQTAVNGKLETVKEMFHDLYPTFSMDISIFF




TSYGFDYCLSQNAITRYNILLGGWSDDNGIHHKGLNNYINEYNQTVP




RNKRLPKLNKLQKMILSEENSMSFIIDKFENDVDLANAIRYWLKNCQ




FDALNLLIWTLDVHYNLDEIHFKNDNQGKNISDLSQALFKNHHVIRD




AWDYDYDIVNAKAKSRQKPERYAEKRDKAFKKINSFSLSYLANILS




QYDNQYANFVAQFKTRISVHIQNVQQMIADKTLDMRLDPLMLLKSI




SSDTKLVEDIKRVLDSLKDMQRMLTPLLGEGTEPNRDAMFYSDFEPL




MNYVDTLTPLYNKVRNYITKKPYSTKKTSLYFGASNFGSGFDVTKLP




VSHTIIMRDKGCYYLAVIDNNKLIDKLYDHNDNDGYEYMVYKQIPS




PIKYFSLKNILPQDPPDDIRQLLEDRKNGAKWSHDDETRFIDYIVNEF




LPTYPPIHDKNGNPYFSWKFKNPDEYESLNEFFDDVSKQAYQTSFRF




VSRDFVDDAVENGDIFFFQIYNQDFSPASHGKPSPHTLWFRALFSDV




NLETKDIRLKGNATAYFRPASIFYTDEKWRKGHHYEQLKNKFKFPII




KDKRYALDKFFFHITLEINCNATVEKYFNNRVNEEIRKADRYNILAIN




RGERNLLYAVVMDQDGTILEQKSFNIIKSELPNKTVKETDYWKKLH




AREKERDTARKSWKSIECIKDLKKGYLSYVVKTITDMMFEYNAVLV




MENLDIEMKRSRQKIEKNVYAQFQNAIIQKLSMYVNKDIDLHIARTA




PGGTLNPYQLTYIPASRTKTPKQNGFVFFLNPWNITEIDPTTGFVDLF




QTCFRTKNEYKDFFAKFKDIRYNEAQGWFEFDTDYTYFRDKEKAGK




RTRWNICSYGTRLRRFRNPDKNYAEDAMTVYPTQMLKDLFDEYNIP




YAPASAKSTSISIKDDIIQIDKLDFYKKLLYILKLIVQLRNTSPSSTEQE




DDYIISPVINEDTNWFYDSRDYNEESLLPCNTDANGAYSLALKCNMV




IDRIKNTIPGEPVDMYISNADWLDARQ





SEQ
Cas12
MNSKTSIFDFSNIFGRDITLRFKLTPVTINSKGEVKDANGADPYRPYL


ID
Variant
SADEELQEQYELLKTAIDAYHQMYIDKKLKHILCLPLTEKGKDGVEH


NO:

DTAKSKFVKSCLAYIKDYGEKDKKRQTADLRTFISRVFADDNISSLPP


57

YKVKSDFITKTLRQWLEQPDTKVEKKEAILDLIEKNGSKLYANCQGL




LEARQRLYEKDGKSTSVPYRCIDRNLPRFSKDYHLFEKILGDCSDVF




DFEQLDKDFSEELKGIARLSGIRVESVREVFQPLLYLAYLNQEGIQYL




NTIIGTKKEKGTSALGLNEYINQYNQKQGIKKKKDGIPMLNKLNNQI




LFGDEVFIETLAEHKEAIPVIKKVVSSLGKLGAFDGECHENKLYQFLL




SLSSYAGNIYVNTKVVAQISSSLWGDYSILYDAVKHDKNGRLIQKSV




TLGELNEKIERLKLEDNRDAFEYFRRSQVKDVVHGSSNVGVFEQLK




NCYNDFVEKKILKCSFFSEDQVLVIQRLFDSILSLQRIFKVFCPSLYEV




DSDGLFVAKFSDYWNVLRGFDKDYDLLRNLFKRKPYSTDKIRVHFG




LSNLMDGFVDSWTDKKDKGTQYNGYILRQAHSFVDENTSKELQEFQ




RYNYYLVISGNVRLFREKGNALVCEKKKEKLVASDEFSGFERFDYY




QSSINNFNREFKRLTGRDRKSFTDEILQNEGKKELKSTYIENLIKVAK




SMKRLTALQNLVSDEKVRKYSENLDYETLSAEIGQILATGRERKYVP




VSTNEMKNLLKSSKNNKGEEVRTFMFRISNKDLSYAETMQKGERKS




HGAENMHTMYFRALLDTLQNTFDIGTGTVYFRKASDKRKMKYDEK




NPTHRKGDELAFKNPYNKGKKKSVFGYDLIKDRRYTKDSYLFHLSIT




QNYQKKGNAEDLNAMVRDYIRTQEDLRVIGIDRGERNLLYATMIDG




EGHILAQKSFNVIGYQGTTASGESFQVETDYHQLLNEKAEKMRSLQR




EWKEMDKIQDMKDGYLSVVVHELAKMVVENNAIIVMEDLNMGFM




ESRQSQLANVYQKFEEKLRNKLQFYVDKRKRNDEPSGLYHALQLAG




TETKDNQNGFIFYIPAWNTSKIDSVTGFVNLFNLKYTNIKDAKAFFST




FEKIEKNVETGHYDFTFSYSSMARKKMAKRMDGTRDSWTISTHGSRI




VREQKGNYWEYREIESLTSEFDALFEKYSIDTRCRLKEAIDKCGEAEF




FKELIRLMKWTLQLRNYDDRGNDYIVSPVCYRGNEYYCSLDYDNEE




GMCISKIPCQMPKDADANGAFNIARKGLMLCERLKKGEKIGVIKGTE




WLQYVQNMSERYVGMV





SEQ
Cas12
MINTMEQPKKSIWDEFTNLYSLQKTLRFELKPQGKTKELVRTLFINPE


ID
Variant
EHHHKLISDDLELSKNYKKVKKLIDCMHRNIINNVLSKHQFTGEELK


NO:

KLDKNSNAEDNDTETDNADKKDPFAKIRERLTKALNEESKIMFDNK


58

LLNPKKGKNKGECELKKWMDKAEDKYFELGNNEKIDKEAVKADM




ERLEGFFTYFGGFNKNRENVYSSKKIATAIPFRIIHDNFPIFKKNIENY




KKITEKHPELAKLLNEKGANEIFQLEHFNKCLTQDGIDVYNNEKLGII




AKEQGKEQDKGINQLINEYAQKKNKEIKENAKGGEKPKKIKIAVFDK




LKKQILSISKTKSFQFEVFEDTSDIINGINKRYTFLTEAKEGMSIVDEIK




KIIGSVGDEKYSLDEIYLKEKFISTLSKKLFNYSRYIEVALEKWYDDR




YDDKINKSGTDKRKFISAKQFSITSIQDAINYYLEKYEKDEELSKKYT




GKNIIVDYFKNPTITIEHKQKEEVISEEKDLFKELEVRRNVIQHILNGD




YKKDLKEEKQQDGDSEKVKAFLDALLEFNYILNPFIIKDKNLRKEQE




KDEEFYNEIKKLQESIFEAEILDLYNQTRNYITKKPYKLDKFKLTFGS




GYFLSGWSNDMEEREGSILIKYNEDRSKNYYLIIMAKPLTDDDKKQL




FSDNGTHSKICIYEFQKMDMKNFPRMFINSKGSNPAPAIEKYNLPIKTI




WADYQKYKNLNQKGKDKFLEENPDFRHNLIGYFKICAEKHESLAPF




KHQFSSIWKPTKEYENLAQFYKDTLEACYNLKFENVNFDNISQLVSS




GKLHLFKIHNKDFNPGSTGKKNLHTLYWEMLFDEKNLQDVIFKLSG




GAELFYREASILKNKIIHKIGEKVLKKFFKLPDGKLEPVPAESIKNLSA




YFRKELPEHELTEIDRKYIDNYSIIGKKDDKLGIMKDERFTVDKIQFH




CPITINFKSKNKNFINDDVLEYLHKRDDVHIIGLDRGERHLIYLTMIN




KDGKIVDNMQFSLNELQRRYKINGNEEIQKINYQKLLDTREVSRTEA




RRNWQTIENIKNLKEGYLSLIVHQLAKLMIEKNAIVVMENLNYGFKD




SRARVEKQIYQKFESILIKKLQYLVMDKNNLYDSGGVLSAYQLTNQE




VPAYKYISKQNGFLFYVPPDYTSKIDPETGFINLLDTRYYSRKNAVAL




LNKFDKIYYDRDNKYFRFDFDYNSTDSNGNKNFDKLRVDISELTRTK




WSVCSHPAKRSITVQINNKWVRQPINDVTDKLIKLFEDKQIGYESGK




CLKDEILKVEDAKFFEDLLRYLSVLLALRHTYTENGVEYDLIISSVEK




APGSNEFFVSGKDNNLPANADANGAYNIARKGLWLLRKLDEIDNQE




LAIKKFNELKHAKEIKKNGEESKEDKGDRKRKKKWVSQWCPNKEW




LAFAQSMQDVSEK





SEQ
Cas12
MNNGTNNFQNFIGISSLQKTLRNALIPTETTQQFIVKNGIIKEDELRGE


ID
Variant
NRQILKDIMDDYYRGFISETLSSIDDIDWTSLFEKMEIQLKNGDNKDT


NO:

LIKEQTEYRKAIHKKFANDDRFKNMFSAKLISDILPEFVIHNNNYSAS


59

EKEEKTQVIKLFSRFATSFKDYFKNRANCFSADDISSSSCHRIVNDNA




EIFFSNALVYRRIVKSLSNDDINKISGDMKDSLKEMSLEEIYSYEKYG




EFITQEGISFYNDICGKVNSFMNLYCQKNKENKNLYKLQKLHKQILCI




ADTSYEVPYKFESDEEVYQSVNGFLDNISSKHIVERLRKIGDNYNGY




NLDKIYIVSKFYESVSQKTYRDWETINTALEIHYNNILPGNGKSKADK




VKKAVKNDLQKSITEINELVSNYKLCSDDNIKAETYIHEISHILNNFE




AQELKYNPEIHLVESELKASELKNVLDVIMNAFHWCSVFMTEELVD




KDNNFYAELEEIYDEIYPVISLYNLVRNYVTQKPYSTKKIKLNFGIPTL




ADGWSKSKEYSNNAIILMRDNLYYLGIFNAKNKPDKKIIEGNTSENK




GDYKKMIYNLLPGPNKMIPKVFLSSKTGVETYKPSAYILEGYKQNKH




IKSSKDFDITFCHDLIDYFKNCIAIHPEWKNFGFDFSDTSTYEDISGFY




REVELQGYKIDWTYISEKDIDLLQEKGQLYLFQIYNKDFSKKSTGND




NLHTMYLKNLFSEENLKDIVLKLNGEAEIFFRKSSIKNPIIHKKGSILV




NRTYEAEEKDQFGNIQIVRKNIPENIYQELYKYFNDKSDKELSDEAA




KLKNVVGHHEAATNIVKDYRYTYDKYFLHMPITINFKANKTGFIND




RILQYIAKEKDLHVIGIDRGERNLIYVSVIDTCGNIVEQKSFNIVNGYD




YQIKLKQQEGARQIARKEWKEIGKIKEIKEGYLSLVIHEISKMVIKYN




AIIAMEDLSYGFKKGRFKVERQVYQKFETMLINKLNYLVFKDISITEN




GGLLKGYQLTYIPDKLKNVGHQCGCIFYVPAAYTSKIDPTTGFVNIF




KFKDLTVDAKREFIKKFDSIRYDSEKNLFCFTFDYNNFITQNTVMSKS




SWSVYTYGVRIKRRFVNGRFSNESDTIDITKDMEKTLEMTDINWRDG




HDLRQDIIDYEIVQHIFEIFRLTVQMRNSLSELEDRDYDRLISPVLNEN




NIFYDSAKAGDALPKDADANGAYCIALKGLYEIKQITENWKEDGKF




SRDKLKISNKDWFDFIQNKRYL





SEQ
Cas12
MSNLNTFISPEFTGKIKMTKSLKVSMIPIGETEHWIAKHKVFEKDREL


ID
Variant
FDKNLKARPILDEFIKYTVSRALPNLLFDFEAYYLVKKDRTKARAFE


NO:

KELAKTVTDLILKEMDELKSASLIDSADFVKTTLKKFAGTHDIPGLSR


60

IEAIESLEAASKLTALNGKFNTSRIAIINTLIPKRIIENFDIYLSNMEKIR




NVYESGEFGFLFERYPDTLLFMEPANYRTVCSPEAIEDYNRFISGYGD




STESWIKGFNQELSEASNSSKSSNGGVRRYSLIKPLHKQHLFETKKFF




TFASISSDDDVRELINSVKGSTEDACLNALAFFSSSDPKTLFVKGSYL




HTLSAFLYGSANSYILPERIKEGEKARLTAEYDSVAKKTKAVTTRYN




VAMNNISKKINEKIFSLADIDAYCCDISKRRSVREILLGIMQEMYAAV




YGENGKWSNIEAEAVLDSKTKIWKAKNGAVAKAVNDYLTAILEIRK




FIRPFALRMEELEELGLDTSSALDAGEITNTLFEAVRAQKLVHAYLTR




NDADIALSTQVYFGGTQKAAASWWNYETGDIQNRQIALAKKDGMY




YFIGTFDERGSYSIEPASPGEDYYEMLDVKKGQDANKQIKKVLFSNK




AIREHFADSSNDYVITTKVNSPITVRREIFDKYQAGEFKLTSQKIRKG




DLVGEKEMTYYREYMDLLFQMAKGYTEYSRFNMDTLLPIEEYDTE




NDLLDDVNTNTIDYRWVRISAACIDDGVRNGDIFVFRAQTSSMYGK




RENKKGYTGLFLELVSDENLLVTRGMSLNSAMSIYYRAKVHDAITV




HKKGDVLVNKFTNARERIPENSYKAICAFYNSGKSIEELTIEDRDWL




AKATTRICSGEIIKDRRYTKNQYSISISYNINRSVNNRKRVDLATIVDD




TASAGRIISVTRGTKDLVYYTVIDDGGSVIEARSLNVINGINYAKMLA




QISEERHDSNANFDIPKRVETIKEAYCAFAVHEIISAALKHNALIVVEL




ISDAIKDKYSLLDNQVFLKFENVLKNCLMSVKVKGARGMEPGSISNP




LQLCNADDKSFRNGILYQIPSSYINICPVTGYADIIDYYNIVSAGDIRN




FFVRFENIVYNKEKARFEFSFDLKNIPIKLEKCPDRTKWTVLGRGEIT




TYDPLTKSNHYVFDAAQMLAETVSKEGLDPCANIVEHIDELSAATLK




KMFNTFRNIAKGIVSECDEVPVSYYKSPVIDEADIKNKSLDNKSISEIK




CYNLDEKARYYLALAKSSSDGENKNRYVSSTAIEWLNYIQEKRTHE









Alternatively, the Type V CRISPR/Cas enzyme is a programmable Cas14 nuclease. A Cas14 protein of the present disclosure includes 3 partial RuvC domains (RuvC-I, RuvC-II, and RuvC-III, also referred to herein as subdomains) that are not contiguous with respect to the primary amino acid sequence of the Cas14 protein, but form a RuvC domain once the protein is produced and folds. A naturally occurring Cas14 protein functions as an endonuclease that catalyzes cleavage at a specific sequence in a target nucleic acid.


In some instances, the TypeV CRISPR/Cas protein comprises a Cas14 protein. Cas14 proteins may comprise a bilobed structure with distinct amino-terminal and carboxy-terminal domains. The amino- and carboxy-terminal domains may be connected by a flexible linker. The flexible linker may affect the relative conformations of the amino- and carboxyl-terminal domains. The flexible linker may be short, for example less than 10 amino acids, less than 8 amino acids, less than 6 amino acids, less than 5 amino acids, or less than 4 amino acids in length. The flexible linker may be sufficiently long to enable different conformations of the amino- and carboxy-terminal domains among two Cas14 proteins of a Cas14 dimer complex (e.g., the relative orientations of the amino- and carboxy-terminal domains differ between two Cas14 proteins of a Cas14 homodimer complex). The linker domain may comprise a mutation which affects the relative conformations of the amino- and carboxyl-terminal domains. The linker may comprise a mutation which affects Cas14 dimerization. For example, a linker mutation may enhance the stability of a Cas14 dimer.


In some instances, the amino-terminal domain of a Cas14 protein comprises a wedge domain, a recognition domain, a zinc finger domain, or any combination thereof. The wedge domain may comprise a multi-strand β-barrel structure. A multi-strand ß-barrel structure may comprise an oligonucleotide/oligosaccharide-binding fold that is structurally comparable to those of some Cas12 proteins. The recognition domain and the zinc finger domain may each (individually or collectively) be inserted between β-barrel strands of the wedge domain. The recognition domain may comprise a 4-α-helix structure, structurally comparable but shorter than those found in some Cas12 proteins. The recognition domain may comprise a binding affinity for a guide nucleic acid or for a guide nucleic acid-target nucleic acid heteroduplex. In some cases, a REC lobe may comprise a binding affinity for a PAM sequence in the target nucleic acid. The amino-terminal may comprise a wedge domain, a recognition domain, and a zinc finger domain. The carboxy-terminal may comprise a RuvC domain, a zinc finger domain, or any combination thereof. The carboxy-terminal may comprise one RuvC and one zinc finger domain.


Cas14 proteins may comprise a RuvC domain or a partial RuvC domain. The RuvC domain may be defined by a single, contiguous sequence, or a set of partial RuvC domains that are not contiguous with respect to the primary amino acid sequence of the Cas14 protein. In some instances, a partial RuvC domain does not have any substrate binding activity or catalytic activity on its own. A Cas14 protein of the present disclosure may include multiple partial RuvC domains, which may combine to generate a RuvC domain with substrate binding or catalytic activity. For example, a Cas14 may include 3 partial RuvC domains (RuvC-I, RuvC-II, and RuvC-III, also referred to herein as subdomains) that are not contiguous with respect to the primary amino acid sequence of the Cas14 protein, but form a RuvC domain once the protein is produced and folds. A Cas14 protein may comprise a linker loop connecting a carboxy terminal domain of the Cas14 protein with the amino terminal domain of the Cas14 protein, and wherein the carboxy terminal domain comprises one or more RuvC domains and the amino terminal domain comprises a recognition domain.


Cas14 proteins may comprise a zinc finger domain. In some instances, a carboxy terminal domain of a Cas14 protein comprises a zinc finger domain. In some instances, an amino terminal domain of a Cas14 protein comprises a zinc finger domain. In some instances, the amino terminal domain comprises a wedge domain (e.g., a multi-ß-barrel wedge structure), a zinc finger domain, or any combination thereof. In some cases, the carboxy terminal domain comprises the RuvC domains and a zinc finger domain, and the amino terminal domain comprises a recognition domain, a wedge domain, and a zinc finger domain.


Cas14 proteins may be relatively small compared to many other Cas proteins, making them suitable for nucleic acid detection or gene editing. For instance, a Cas14 protein may be less likely to adsorb to a surface or another biological species due to its small size. The smaller nature of these proteins also allows for them to be more easily packaged as a reagent in a system or assay, and delivered with higher efficiency as compared to other larger Cas proteins. In some cases, a Cas14 protein is 400 to 800 amino acid residues long, 400 to 600 amino acid residues long, 440 to 580 amino acid residues long, 460 to 560 amino acid residues long, 460 to 540 amino acid residues long, 460 to 500 amino acid residues long, 400 to 500 amino acid residues long, or 500 to 600 amino acid residues long. In some cases, a Cas14 protein is less than about 550 amino acid residues long. In some cases, a Cas14 protein is less than about 500 amino acid residues long.


In some instances, a Cas14 protein may function as an endonuclease that catalyzes cleavage at a specific position within a target nucleic acid. In some instances, a Cas14 protein is capable of catalyzing non-sequence-specific cleavage of a single stranded nucleic acid. In some cases, a Cas14 protein is activated to perform trans cleavage activity after binding of a guide nucleic acid with a target nucleic acid. This trans cleavage activity is also referred to as “collateral” or “transcollateral” cleavage. Trans cleavage activity may be non-specific cleavage of nearby single-stranded nucleic acid by the activated programmable nuclease, such as trans cleavage of reporters with a detection moiety.


A programmable Cas14 nuclease can be a Cas14a protein, a Cas14b protein, a Cas14c protein, a Cas14d protein, a Cas14e protein, a Cas14f protein, a Cas14g protein, a Cas14h protein, or a Cas14u protein. In some cases, a suitable Cas14 protein comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to any one of SEQ ID NO: 61-SEQ ID NO: 152.









TABLE 2







Cas14 Protein Sequences








SEQ



ID



NO
Sequence





SEQ
MEVQKTVMKTLSLRILRPLYSQEIEKEIKEEKERRKQAGGTGELDGGFYKKLEKKHSE


ID
MFSFDRLNLLLNQLQREIAKVYNHAISELYIATIAQGNKSNKHYISSIVYNRAYGYFYN


NO:
AYIALGICSKVEANFRSNELLTQQSALPTAKSDNFPIVLHKQKGAEGEDGGFRISTEGS


61
DLIFEIPIPFYEYNGENRKEPYKWVKKGGQKPVLKLILSTFRRQRNKGWAKDEGTDAEI



RKVTEGKYQVSQIEINRGKKLGEHQKWFANFSIEQPIYERKPNRSIVGGLDVGIRSPLV



CAINNSFSRYSVDSNDVFKFSKQVFAFRRRLLSKNSLKRKGHGAAHKLEPITEMTEKN



DKFRKKIIERWAKEVTNFFVKNQVGIVQIEDLSTMKDREDHFFNQYLRGFWPYYQMQ



TLIENKLKEYGIEVKRVQAKYTSQLCSNPNCRYWNNYFNFEYRKVNKFPKFKCEKCN



LEISADYNAARNLSTPDIEKFVAKATKGINLPEK





SEQ
MEEAKTVSKTLSLRILRPLYSAEIEKEIKEEKERRKQGGKSGELDSGFYKKLEKKHTQM


ID
FGWDKLNLMLSQLQRQIARVFNQSISELYIETVIQGKKSNKHYTSKIVYNRAYSVFYN


NO:
AYLALGITSKVEANFRSTELLMQKSSLPTAKSDNFPILLHKQKGVEGEEGGFKISADGN


62
DLIFEIPIPFYEYDSANKKEPFKWIKKGGQKPTIKLILSTFRRQRNKGWAKDEGTDAEIR



KVIEGKYQVSHIEINRGKKLGDHQKWFVNFTIEQPIYERKLDKNIIGGIDVGIKSPLVCA



VNNSFARYSVDSNDVLKFSKQAFAFRRRLLSKNSLKRSGHGSKNKLDPITRMTEKNDR



FRKKIIERWAKEVTNFFIKNQVGTVQIEDLSTMKDRQDNFFNQYLRGFWPYYQMQNLI



ENKLKEYGIETKRIKARYTSQLCSNPSCRHWNSYFSFDHRKTNNFPKFKCEKCALEISA



DYNAARNISTPDIEKFVAKATKGINLPDKNENVILE





SEQ
MAKNTITKTLKLRIVRPYNSAEVEKIVADEKNNREKIALEKNKDKVKEACSKHLKVA


ID
AYCTTQVERNACLFCKARKLDDKFYQKLRGQFPDAVFWQEISEIFRQLQKQAAEIYNQ


NO:
SLIELYYEIFIKGKGIANASSVEHYLSDVCYTRAAELFKNAAIASGLRSKIKSNFRLKEL


63
KNMKSGLPTTKSDNFPIPLVKQKGGQYTGFEISNHNSDFIIKIPFGRWQVKKEIDKYRP



WEKFDFEQVQKSPKPISLLLSTQRRKRNKGWSKDEGTEAEIKKVMNGDYQTSYIEVKR



GSKIGEKSAWMLNLSIDVPKIDKGVDPSIIGGIDVGVKSPLVCAINNAFSRYSISDNDLF



HENKKMFARRRILLKKNRHKRAGHGAKNKLKPITILTEKSERFRKKLIERWACEIADFF



IKNKVGTVQMENLESMKRKEDSYFNIRLRGFWPYAEMQNKIEFKLKQYGIEIRKVAPN



NTSKTCSKCGHLNNYFNFEYRKKNKFPHFKCEKCNFKENADYNAALNISNPKLKSTKE



EP





SEQ
MERQKVPQIRKIVRVVPLRILRPKYSDVIENALKKFKEKGDDTNTNDFWRAIRDRDTE


ID
FFRKELNFSEDEINQLERDTLFRVGLDNRVLFSYFDFLQEKLMKDYNKIISKLFINRQSK


NO:
SSFENDLTDEEVEELIEKDVTPFYGAYIGKGIKSVIKSNLGGKFIKSVKIDRETKKVTKL


64
TAINIGLMGLPVAKSDTFPIKIIKTNPDYITFQKSTKENLQKIEDYETGIEYGDLLVQITIP



WFKNENKDFSLIKTKEAIEYYKLNGVGKKDLLNINLVLTTYHIRKKKSWQIDGSSQSL



VREMANGELEEKWKSFFDTFIKKYGDEGKSALVKRRVNKKSRAKGEKGRELNLDERI



KRLYDSIKAKSFPSEINLIPENYKWKLHFSIEIPPMVNDIDSNLYGGIDFGEQNIATLCVK



NIEKDDYDFLTIYGNDLLKHAQASYARRRIMRVQDEYKARGHGKSRKTKAQEDYSER



MQKLRQKITERLVKQISDFFLWRNKFHMAVCSLRYEDLNTLYKGESVKAKRMRQFIN



KQQLFNGIERKLKDYNSEIYVNSRYPHYTSRLCSKCGKLNLYFDFLKFRTKNIIIRKNPD



GSEIKYMPFFICEFCGWKQAGDKNASANIADKDYQDKLNKEKEFCNIRKPKSKKEDIG



EENEEERDYSRRFNRNSFIYNSLKKDNKLNQEKLFDEWKNQLKRKIDGRNKFEPKEYK



DRFSYLFAYYQEIIKNESES





SEQ
MVPTELITKTLQLRVIRPLYFEEIEKELAELKEQKEKEFEETNSLLLESKKIDAKSLKKL


ID
KRKARSSAAVEFWKIAKEKYPDILTKPEMEFIFSEMQKMMARFYNKSMTNIFIEMNND


NO:
EKVNPLSLISKASTEANQVIKCSSISSGLNRKIAGSINKTKFKQVRDGLISLPTARTETFPI


65
SFYKSTANKDEIPISKINLPSEEEADLTITLPFPFFEIKKEKKGQKAYSYFNIIEKSGRSNN



KIDLLLSTHRRQRRKGWKEEGGTSAEIRRLMEGEFDKEWEIYLGEAEKSEKAKNDLIK



NMTRGKLSKDIKEQLEDIQVKYFSDNNVESWNDLSKEQKQELSKLRKKKVEELKDW



KHVKEILKTRAKIGWVELKRGKRQRDRNKWFVNITITRPPFINKELDDTKFGGIDLGV



KVPFVCAVHGSPARLIIKENEILQFNKMVSARNRQITKDSEQRKGRGKKNKFIKKEIFN



ERNELFRKKIIERWANQIVKFFEDQKCATVQIENLESFDRTSYK





SEQ
MKSDTKDKKIIIHQTKTLSLRIVKPQSIPMEEFTDLVRYHQMIIFPVYNNGAIDLYKKLF


ID
KAKIQKGNEARAIKYFMNKIVYAPIANTVKNSYIALGYSTKMQSSFSGKRLWDLRFGE


NO:
ATPPTIKADFPLPFYNQSGFKVSSENGEFIIGIPFGQYTKKTVSDIEKKTSFAWDKFTLED


66
TTKKTLIELLLSTKTRKMNEGWKNNEGTEAEIKRVMDGTYQVTSLEILQRDDSWFVNF



NIAYDSLKKQPDRDKIAGIHMGITRPLTAVIYNNKYRALSIYPNTVMHLTQKQLARIKE



QRTNSKYATGGHGRNAKVTGTDTLSEAYRQRRKKIIEDWIASIVKFAINNEIGTIYLEDI



SNTNSFFAAREQKLIYLEDISNTNSFLSTYKYPISAISDTLQHKLEEKAIQVIRKKAYYV



NQICSLCGHYNKGFTYQFRRKNKFPKMKCQGCLEATSTEFNAAANVANPDYEKLLIK



HGLLQLKK





SEQ
MSTITRQVRLSPTPEQSRLLMAHCQQYISTVNVLVAAFDSEVLTGKVSTKDFRAALPS


ID
AVKNQALRDAQSVFKRSVELGCLPVLKKPHCQWNNQNWRVEGDQLILPICKDGKTQ


NO:
QERFRCAAVALEGKAGILRIKKKRGKWIADLTVTQEDAPESSGSAIMGVDLGIKVPAV


67
AHIGGKGTRFFGNGRSQRSMRRRFYARRKTLQKAKKLRAVRKSKGKEARWMKTINH



QLSRQIVNHAHALGVGTIKIEALQGIRKGTTRKSRGAAARKNNRMTNTWSFSQLTLFIT



YKAQRQGITVEQVDPAYTSQDCPACRARNGAQDRTYVCSECGWRGHRDTVGAINISR



RAGLSGHRRGATGA





SEQ
MIAQKTIKIKLNPTKEQIIKLNSIIEEYIKVSNFTAKKIAEIQESFTDSGLTQGTCSECGKE


ID
KTYRKYHLLKKDNKLFCITCYKRKYSQFTLQKVEFQNKTGLRNVAKLPKTYYTNAIR


NO:
FASDTFSGFDEIIKKKQNRLNSIQNRLNFWKELLYNPSNRNEIKIKVVKYAPKTDTREH


68
PHYYSEAEIKGRIKRLEKQLKKFKMPKYPEFTSETISLQRELYSWKNPDELKISSITDKN



ESMNYYGKEYLKRYIDLINSQTPQILLEKENNSFYLCFPITKNIEMPKIDDTFEPVGIDW



GITRNIAVVSILDSKTKKPKFVKFYSAGYILGKRKHYKSLRKHFGQKKRQDKINKLGT



KEDRFIDSNIHKLAFLIVKEIRNHSNKPIILMENITDNREEAEKSMRQNILLHSVKSRLQN



YIAYKALWNNIPTNLVKPEHTSQICNRCGHQDRENRPKGSKLFKCVKCNYMSNADFN



ASINIARKFYIGEYEPFYKDNEKMKSGVNSISM





SEQ
LKLSEQENITTGVKFKLKLDKETSEGLNDYFDEYGKAINFAIKVIQKELAEDRFAGKVR


ID
LDENKKPLLNEDGKKIWDFPNEFCSCGKQVNRYVNGKSLCQECYKNKFTEYGIRKRM


NO:
YSAKGRKAEQDINIKNSTNKISKTHFNYAIREAFILDKSIKKQRKERFRRLREMKKKLQ


69
EFIEIRDGNKILCPKIEKQRVERYIHPSWINKEKKLEDFRGYSMSNVLGKIKILDRNIKRE



EKSLKEKGQINFKARRLMLDKSVKFLNDNKISFTISKNLPKEYELDLPEKEKRLNWLKE



KIKIIKNQKPKYAYLLRKDDNFYLQYTLETEFNLKEDYSGIVGIDRGVSHIAVYTFVHN



NGKNERPLFLNSSEILRLKNLQKERDRFLRRKHNKKRKKSNMRNIEKKIQLILHNYSKQ



IVDFAKNKNAFIVFEKLEKPKKNRSKMSKKSQYKLSQFTFKKLSDLVDYKAKREGIKV



LYISPEYTSKECSHCGEKVNTQRPFNGNSSLFKCNKCGVELNADYNASINIAKKGLNIL



NSTN





SEQ
MEESIITGVKFKLRIDKETTKKLNEYFDEYGKAINFAVKIIQKELADDRFAGKAKLDQN


ID
KNPILDENGKKIYEFPDEFCSCGKQVNKYVNNKPFCQECYKIRFTENGIRKRMYSAKG


NO:
RKAEHKINILNSTNKISKTHFNYAIREAFILDKSIKKQRKKRNERLRESKKRLQQFIDMR


70
DGKREICPTIKGQKVDRFIHPSWITKDKKLEDFRGYTLSIINSKIKILDRNIKREEKSLKE



KGQIIFKAKRLMLDKSIRFVGDRKVLFTISKTLPKEYELDLPSKEKRLNWLKEKIEIIKN



QKPKYAYLLRKNIESEKKPNYEYYLQYTLEIKPELKDFYDGAIGIDRGINHIAVCTFISN



DGKVTPPKFFSSGEILRLKNLQKERDRFLLRKHNKNRKKGNMRVIENKINLILHRYSKQ



IVDMAKKLNASIVFEELGRIGKSRTKMKKSQRYKLSLFIFKKLSDLVDYKSRREGIRVT



YVPPEYTSKECSHCGEKVNTQRPFNGNYSLFKCNKCGIQLNSDYNASINIAKKGLKIPN



ST





SEQ
LWTIVIGDFIEMPKQDLVTTGIKFKLDVDKETRKKLDDYFDEYGKAINFAVKIIQKNLK


ID
EDRFAGKIALGEDKKPLLDKDGKKIYNYPNESCSCGNQVRRYVNAKPFCVDCYKLKF


NO:
TENGIRKRMYSARGRKADSDINIKNSTNKISKTHFNYAIREGFILDKSLKKQRSKRIKKL


71
LELKRKLQEFIDIRQGQMVLCPKIKNQRVDKFIHPSWLKRDKKLEEFRGYSLSVVEGKI



KIFNRNILREEDSLRQRGHVNFKANRIMLDKSVRFLDGGKVNFNLNKGLPKEYLLDLP



KKENKLSWLNEKISLIKLQKPKYAYLLRREGSFFIQYTIENVPKTFSDYLGAIGIDRGISH



IAVCTFVSKNGVNKAPVFFSSGEILKLKSLQKQRDLFLRGKHNKIRKKSNMRNIDNKIN



LILHKYSRNIVNLAKSEKAFIVFEKLEKIKKSRFKMSKSLQYKLSQFTFKKLSDLVEYK



AKIEGIKVDYVPPEYTSKECSHCGEKVDTQRPFNGNSSLFKCNKCRVQLNADYNASINI



AKKSLNISN





SEQ
MSKTTISVKLKIIDLSSEKKEFLDNYFNEYAKATTFCQLRIRRLLRNTHWLGKKEKSSK


ID
KWIFESGICDLCGENKELVNEDRNSGEPAKICKRCYNGRYGNQMIRKLFVSTKKREVQ


NO:
ENMDIRRVAKLNNTHYHRIPEEAFDMIKAADTAEKRRKKNVEYDKKRQMEFIEMFND


72
EKKRAARPKKPNERETRYVHISKLESPSKGYTLNGIKRKIDGMGKKIERAEKGLSRKKI



FGYQGNRIKLDSNWVRFDLAESEITIPSLFKEMKLRITGPTNVHSKSGQIYFAEWFERIN



KQPNNYCYLIRKTSSNGKYEYYLQYTYEAEVEANKEYAGCLGVDIGCSKLAAAVYYD



SKNKKAQKPIEIFTNPIKKIKMRREKLIKLLSRVKVRHRRRKLMQLSKTEPIIDYTCHKT



ARKIVEMANTAKAFISMENLETGIKQKQQARETKKQKFYRNMFLFRKLSKLIEYKALL



KGIKIVYVKPDYTSQTCSSCGADKEKTERPSQAIFRCLNPTCRYYQRDINADFNAAVNI



AKKALNNTEVVTTLL





SEQ
MARAKNQPYQKLTTTTGIKFKLDLSEEEGKRFDEYFSEYAKAVNFCAKVIYQLRKNL


ID
KFAGKKELAAKEWKFEISNCDFCNKQKEIYYKNIANGQKVCKGCHRTNFSDNAIRKK


NO:
MIPVKGRKVESKFNIHNTTKKISGTHRHWAFEDAADIIESMDKQRKEKQKRLRREKRK


73
LSYFFELFGDPAKRYELPKVGKQRVPRYLHKIIDKDSLTKKRGYSLSYIKNKIKISERNI



ERDEKSLRKASPIAFGARKIKMSKLDPKRAFDLENNVFKIPGKVIKGQYKFFGTNVANE



HGKKFYKDRISKILAGKPKYFYLLRKKVAESDGNPIFEYYVQWSIDTETPAITSYDNIL



GIDAGITNLATTVLIPKNLSAEHCSHCGNNHVKPIFTKFFSGKELKAIKIKSRKQKYFLR



GKHNKLVKIKRIRPIEQKVDGYCHVVSKQIVEMAKERNSCIALEKLEKPKKSKFRQRR



REKYAVSMFVFKKLATFIKYKAAREGIEIIPVEPEGTSYTCSHCKNAQNNQRPYFKPNS



KKSWTSMFKCGKCGIELNSDYNAAFNIAQKALNMTSA





SEQ
MDEKHFFCSYCNKELKISKNLINKISKGSIREDEAVSKAISIHNKKEHSLILGIKFKLFIEN


ID
KLDKKKLNEYFDNYSKAVTFAARIFDKIRSPYKFIGLKDKNTKKWTFPKAKCVFCLEE


NO:
KEVAYANEKDNSKICTECYLKEFGENGIRKKIYSTRGRKVEPKYNIFNSTKELSSTHYN


74
YAIRDAFQLLDALKKQRQKKLKSIFNQKLRLKEFEDIFSDPQKRIELSLKPHQREKRYIH



LSKSGQESINRGYTLRFVRGKIKSLTRNIEREEKSLRKKTPIHFKGNRLMIFPAGIKFDFA



SNKVKISISKNLPNEFNFSGTNVKNEHGKSFFKSRIELIKTQKPKYAYVLRKIKREYSKL



RNYEIEKIRLENPNADLCDFYLQYTIETESRNNEEINGIIGIDRGITNLACLVLLKKGDKK



PSGVKFYKGNKILGMKIAYRKHLYLLKGKRNKLRKQRQIRAIEPKINLILHQISKDIVKI



AKEKNFAIALEQLEKPKKARFAQRKKEKYKLALFTFKNLSTLIEYKSKREGIPVIYVPPE



KTSQMCSHCAINGDEHVDTQRPYKKPNAQKPSYSLFKCNKCGIELNADYNAAFNIAQ



KGLKTLMLNHSH





SEQ
MLQTLLVKLDPSKEQYKMLYETMERFNEACNQIAETVFAIHSANKIEVQKTVYYPIRE


ID
KFGLSAQLTILAIRKVCEAYKRDKSIKPEFRLDGALVYDQRVLSWKGLDKVSLVTLQG


NO:
RQIIPIKFGDYQKARMDRIRGQADLILVKGVFYLCVVVEVSEESPYDPKGVLGVDLGIK


75
NLAVDSDGEVHSGEQTTNTRERLDSLKARLQSKGTKSAKRHLKKLSGRMAKFSKDVN



HCISKKLVAKAKGTLMSIALEDLQGIRDRVTVRKAQRRNLHTWNFGLLRMFVDYKAK



IAGVPLVFVDPRNTSRTCPSCGHVAKANRPTRDEFRCVSCGFAGAADHIAAMNIAFRA



EVSQPIVTRFFVQSQAPSFRVG





SEQ
MDEEPDSAEPNLAPISVKLKLVKLDGEKLAALNDYFNEYAKAVNFCELKMQKIRKNL


ID
VNIRGTYLKEKKAWINQTGECCICKKIDELRCEDKNPDINGKICKKCYNGRYGNQMIR


NO:
KLFVSTNKRAVPKSLDIRKVARLHNTHYHRIPPEAADIIKAIETAERKRRNRILFDERRY


76
NELKDALENEEKRVARPKKPKEREVRYVPISKKDTPSKGYTMNALVRKVSGMAKKIE



RAKRNLNKRKKIEYLGRRILLDKNWVRFDFDKSEISIPTMKEFFGEMRFEITGPSNVMS



PNGREYFTKWFDRIKAQPDNYCYLLRKESEDETDFYLQYTWRPDAHPKKDYTGCLGI



DIGGSKLASAVYFDADKNRAKQPIQIFSNPIGKWKTKRQKVIKVLSKAAVRHKTKKLE



SLRNIEPRIDVHCHRIARKIVGMALAANAFISMENLEGGIREKQKAKETKKQKFSRNMF



VFRKLSKLIEYKALMEGVKVVYIVPDYTSQLCSSCGTNNTKRPKQAIFMCQNTECRYF



GKNINADFNAAINIAKKALNRKDIVRELS





SEQ
MEKNNSEQTSITTGIKFKLKLDKETKEKLNNYFDEYGKAINFAVRIIQMQLNDDRLAG


ID
KYKRDEKGKPILGEDGKKILEIPNDFCSCGNQVNHYVNGVSFCQECYKKRFSENGIRK


NO:
RMYSAKGRKAEQDINIKNSTNKISKTHFNYAIREAFNLDKSIKKQREKRFKKLKDMKR


77
KLQEFLEIRDGKRVICPKIEKQKVERYIHPSWINKEKKLEEFRGYSLSIVNSKIKSFDRNI



QREEKSLKEKGQINFKAQRLMLDKSVKFLKDNKVSFTISKELPKTFELDLPKKEKKLN



WLNEKLEIIKNQKPKYAYLLRKENNIFLQYTLDSIPEIHSEYSGAVGIDRGVSHIAVYTF



LDKDGKNERPFFLSSSGILRLKNLQKERDKFLRKKHNKIRKKGNMRNIEQKINLILHEY



SKQIVNFAKDKNAFIVFELLEKPKKSRERMSKKIQYKLSQFTFKKLSDLVDYKAKREGI



KVIYVEPAYTSKDCSHCGERVNTQRPFNGNFSLFKCNKCGIVLNSDYNASLNIARKGL



NISAN





SEQ
MAEEKFFFCEKCNKDIKIPKNYINKQGAEEKARAKHEHRVHALILGIKFKIYPKKEDIS


ID
KLNDYFDEYAKAVTFTAKIVDKLKAPFLFAGKRDKDTSKKKWVFPVDKCSFCKEKTE


NO:
INYRTKQGKNICNSCYLTEFGEQGLLEKIYATKGRKVSSSFNLFNSTKKLTGTHNNYV


78
VKESLQLLDALKKQRSKRLKKLSNTRRKLKQFEEMFEKEDKRFQLPLKEKQRELRFIH



VSQKDRATEFKGYTMNKIKSKIKVLRRNIEREQRSLNRKSPVFFRGTRIRLSPSVQFDD



KDNKIKLTLSKELPKEYSFSGLNVANEHGRKFFAEKLKLIKENKSKYAYLLRRQVNKN



NKKPIYDYYLQYTVEFLPNIITNYNGILGIDRGINTLACIVLLENKKEKPSFVKFFSGKGI



LNLKNKRRKQLYFLKGVHNKYRKQQKIRPIEPRIDQILHDISKQIIDLAKEKRVAISLEQ



LEKPQKPKFRQSRKAKYKLSQFNFKTLSNYIDYKAKKEGIRVIYIAPEMTSQNCSRCA



MKNDLHVNTQRPYKNTSSLFKCNKCGVELNADYNAAFNIAQKGLKILNS





SEQ
MISLKLKLLPDEEQKKLLDEMFWKWASICTRVGFGRADKEDLKPPKDAEGVWFSLTQ


ID
LNQANTDINDLREAMKHQKHRLEYEKNRLEAQRDDTQDALKNPDRREISTKRKDLFR


NO:
PKASVEKGFLKLKYHQERYWVRRLKEINKLIERKTKTLIKIEKGRIKFKATRITLHQGSF


79
KIRFGDKPAFLIKALSGKNQIDAPFVVVPEQPICGSVVNSKKYLDEITTNFLAYSVNAM



LFGLSRSEEMLLKAKRPEKIKKKEEKLAKKQSAFENKKKELQKLLGRELTQQEEAIIEE



TRNQFFQDFEVKITKQYSELLSKIANELKQKNDFLKVNKYPILLRKPLKKAKSKKINNL



SPSEWKYYLQFGVKPLLKQKSRRKSRNVLGIDRGLKHLLAVTVLEPDKKTFVWNKLY



PNPITGWKWRRRKLLRSLKRLKRRIKSQKHETIHENQTRKKLKSLQGRIDDLLHNISRK



IVETAKEYDAVIVVEDLQSMRQHGRSKGNRLKTLNYALSLFDYANVMQLIKYKAGIE



GIQIYDVKPAGTSQNCAYCLLAQRDSHEYKRSQENSKIGVCLNPNCQNHKKQIDADLN



AARVIASCYALKINDSQPFGTRKRFKKRTTN





SEQ
METLSLKLKLNPSKEQLLVLDKMFWKWASICTRLGLKKAEMSDLEPPKDAEGVWFSK


ID
TQLNQANTDVNDLRKAMQHQGKRIEYELDKVENRRNEIQEMLEKPDRRDISPNRKDL


NO:
FRPKAAVEKGYLKLKYHKLGYWSKELKTANKLIERKRKTLAKIDAGKMKFKPTRISL


80
HTNSFRIKFGEEPKIALSTTSKHEKIELPLITSLQRPLKTSCAKKSKTYLDAAILNFLAYS



TNAALFGLSRSEEMLLKAKKPEKIEKRDRKLATKRESFDKKLKTLEKLLERKLSEKEK



SVFKRKQTEFFDKFCITLDETYVEALHRIAEELVSKNKYLEIKKYPVLLRKPESRLRSKK



LKNLKPEDWTYYIQFGFQPLLDTPKPIKTKTVLGIDRGVRHLLAVSIFDPRTKTFTFNRL



YSNPIVDWKWRRRKLLRSIKRLKRRLKSEKHVHLHENQFKAKLRSLEGRIEDHFHNLS



KEIVDLAKENNSVIVVENLGGMRQHGRGRGKWLKALNYALSHFDYAKVMQLIKYKA



ELAGVFVYDVAPAGTSINCAYCLLNDKDASNYTRGKVINGKKNTKIGECKTCKKEFD



ADLNAARVIALCYEKRLNDPQPFGTRKQFKPKKP





SEQ
MKALKLQLIPTRKQYKILDEMFWKWASLANRVSQKGESKETLAPKKDIQKIQFNATQ


ID
LNQIEKDIKDLRGAMKEQQKQKERLLLQIQERRSTISEMLNDDNNKERDPHRPLNFRP


NO:
KGWRKFHTSKHWVGELSKILRQEDRVKKTIERIVAGKISFKPKRIGIWSSNYKINFFKR


81
KISINPLNSKGFELTLMTEPTQDLIGKNGGKSVLNNKRYLDDSIKSLLMFALHSRFFGL



NNTDTYLLGGKINPSLVKYYKKNQDMGEFGREIVEKFERKLKQEINEQQKKIIMSQIKE



QYSNRDSAFNKDYLGLINEFSEVFNQRKSERAEYLLDSFEDKIKQIKQEIGESLNISDWD



FLIDEAKKAYGYEEGFTEYVYSKRYLEILNKIVKAVLITDIYFDLRKYPILLRKPLDKIK



KISNLKPDEWSYYIQFGYDSINPVQLMSTDKFLGIDRGLTHLLAYSVFDKEKKEFIINQL



EPNPIMGWKWKLRKVKRSLQHLERRIRAQKMVKLPENQMKKKLKSIEPKIEVHYHNI



SRKIVNLAKDYNASIVVESLEGGGLKQHGRKKNARNRSLNYALSLFDYGKIASLIKYK



ADLEGVPMYEVLPAYTSQQCAKCVLEKGSFVDPEIIGYVEDIGIKGSLLDSLFEGTELSS



IQVLKKIKNKIELSARDNHNKEINLILKYNFKGLVIVRGQDKEEIAEHPIKEINGKFAILD



FVYKRGKEKVGKKGNQKVRYTGNKKVGYCSKHGQVDADLNASRVIALCKYLDINDP



ILFGEQRKSFK





SEQ
MVTRAIKLKLDPTKNQYKLLNEMFWKWASLANRFSQKGASKETLAPKDGTQKIQFN


ID
ATQLNQIKKDVDDLRGAMEKQGKQKERLLIQIQERLLTISEILRDDSKKEKDPHRPQNF


NO:
RPFGWRRFHTSAYWSSEASKLTRQVDRVRRTIERIKAGKINFKPKRIGLWSSTYKINFL


82
KKKINISPLKSKSFELDLITEPQQKIIGKEGGKSVANSKKYLDDSIKSLLIFAIKSRLFGLN



NKDKPLFENIITPNLVRYHKKGQEQENFKKEVIKKFENKLKKEISQKQKEIIFSQIERQY



ENRDATFSEDYLRAISEFSEIFNQRKKERAKELLNSFNEKIRQLKKEVNGNISEEDLKIL



EVEAEKAYNYENGFIEWEYSEQFLGVLEKIARAVLISDNYFDLKKYPILIRKPTNKSKKI



TNLKPEEWDYYIQFGYGLINSPMKIETKNFMGIDRGLTHLLAYSIFDRDSEKFTINQLEL



NPIKGWKWKLRKVKRSLQHLERRMRAQKGVKLPENQMKKRLKSIEPKIESYYHNLSR



KIVNLAKANNASIVVESLEGGGLKQHGRKKNSRHRALNYALSLFDYGKIASLIKYKSD



LEGVPMYEVLPAYTSQQCAKCVLKKGSFVEPEIIGYIEEIGFKENLLTLLFEDTGLSSVQ



VLKKSKNKMTLSARDKEGKMVDLVLKYNFKGLVISQEKKKEEIVEFPIKEIDGKFAVL



DSAYKRGKERISKKGNQKLVYTGNKKVGYCSVHGQVDADLNASRVIALCKYLGINEP



IVFGEQRKSFK





SEQ
LDLITEPIQPHKSSSLRSKEFLEYQISDFLNFSLHSLFFGLASNEGPLVDFKIYDKIVIPKPE


ID
ERFPKKESEEGKKLDSFDKRVEEYYSDKLEKKIERKLNTEEKNVIDREKTRIWGEVNK


NO:
LEEIRSIIDEINEIKKQKHISEKSKLLGEKWKKVNNIQETLLSQEYVSLISNLSDELTNKK


83
KELLAKKYSKFDDKIKKIKEDYGLEFDENTIKKEGEKAFLNPDKFSKYQFSSSYLKLIG



EIARSLITYKGFLDLNKYPIIFRKPINKVKKIHNLEPDEWKYYIQFGYEQINNPKLETENI



LGIDRGLTHILAYSVFEPRSSKFILNKLEPNPIEGWKWKLRKLRRSIQNLERRWRAQDN



VKLPENQMKKNLRSIEDKVENLYHNLSRKIVDLAKEKNACIVFEKLEGQGMKQHGRK



KSDRLRGLNYKLSLFDYGKIAKLIKYKAEIEGIPIYRIDSAYTSQNCAKCVLESRRFAQP



EEISCLDDFKEGDNLDKRILEGTGLVEAKIYKKLLKEKKEDFEIEEDIAMFDTKKVIKEN



KEKTVILDYVYTRRKEIIGTNHKKNIKGIAKYTGNTKIGYCMKHGQVDADLNASRTIA



LCKNFDINNPEIWK





SEQ
MSDESLVSSEDKLAIKIKIVPNAEQAKMLDEMFKKWSSICNRISRGKEDIETLRPDEGK


ID
ELQFNSTQLNSATMDVSDLKKAMARQGERLEAEVSKLRGRYETIDASLRDPSRRHTNP


NO:
QKPSSFYPSDWDISGRLTPRFHTARHYSTELRKLKAKEDKMLKTINKIKNGKIVFKPKR


84
ITLWPSSVNMAFKGSRLLLKPFANGFEMELPIVISPQKTADGKSQKASAEYMRNALLG



LAGYSINQLLFGMNRSQKMLANAKKPEKVEKFLEQMKNKDANFDKKIKALEGKWLL



DRKLKESEKSSIAVVRTKFFKSGKVELNEDYLKLLKHMANEILERDGFVNLNKYPILSR



KPMKRYKQKNIDNLKPNMWKYYIQFGYEPIFERKASGKPKNIMGIDRGLTHLLAVAV



FSPDQQKFLFNHLESNPIMHWKWKLRKIRRSIQHMERRIRAEKNKHIHEAQLKKRLGSI



EEKTEQHYHIVSSKIINWAIEYEAAIVLESLSHMKQRGGKKSVRTRALNYALSLFDYEK



VARLITYKARIRGIPVYDVLPGMTSKTCATCLLNGSQGAYVRGLETTKAAGKATKRK



NMKIGKCMVCNSSENSMIDADLNAARVIAICKYKNLNDPQPAGSRKVFKRF





SEQ
MLALKLKIMPTEKQAEILDAMFWKWASICSRIAKMKKKVSVKENKKELSKKIPSNSDI


ID
WFSKTQLCQAEVDVGDHKKALKNFEKRQESLLDELKYKVKAINEVINDESKREIDPN


NO:
NPSKFRIKDSTKKGNLNSPKFFTLKKWQKILQENEKRIKKKESTIEKLKRGNIFFNPTKIS


85
LHEEEYSINFGSSKLLLNCFYKYNKKSGINSDQLENKFNEFQNGLNIICSPLQPIRGSSKR



SFEFIRNSIINFLMYSLYAKLFGIPRSVKALMKSNKDENKLKLEEKLKKKKSSFNKTVK



EFEKMIGRKLSDNESKILNDESKKFFEIIKSNNKYIPSEEYLKLLKDISEEIYNSNIDFKPY



KYSILIRKPLSKFKSKKLYNLKPTDYKYYLQLSYEPFSKQLIATKTILGIDRGLKHLLAV



SVFDPSQNKFVYNKLIKNPVFKWKKRYHDLKRSIRNRERRIRALTGVHIHENQLIKKLK



SMKNKINVLYHNVSKNIVDLAKKYESTIVLERLENLKQHGRSKGKRYKKLNYVLSNF



DYKKIESLISYKAKKEGVPVSNINPKYTSKTCAKCLLEVNQLSELKNEYNRDSKNSKIG



ICNIHGQIDADLNAARVIALCYSKNLNEPHFK





SEQ
VINLFGYKFALYPNKTQEELLNKHLGECGWLYNKAIEQNEYYKADSNIEEAQKKFELL


ID
PDKNSDEAKVLRGNISKDNYVYRTLVKKKKSEINVQIRKAVVLRPAETIRNLAKVKKK


NO:
GLSVGRLKFIPIREWDVLPFKQSDQIRLEENYLILEPYGRLKFKMHRPLLGKPKTFCIKR


86
TATDRWTISFSTEYDDSNMRKNDGGQVGIDVGLKTHLRLSNENPDEDPRYPNPKIWK



RYDRRLTILQRRISKSKKLGKNRTRLRLRLSRLWEKIRNSRADLIQNETYEILSENKLIAI



EDLNVKGMQEKKDKKGRKGRTRAQEKGLHRSISDAAFSEFRRVLEYKAKRFGSEVKP



VSAIDSSKECHNCGNKKGMPLESRIYECPKCGLKIDRDLNSAKVILARATGVRPGSNA



RADTKISATAGASVQTEGTVSEDFRQQMETSDQKPMQGEGSKEPPMNPEHKSSGRGS



KHVNIGCKNKVGLYNEDENSRSTEKQIMDENRSTTEDMVEIGALHSPVLTT





SEQ
MIASIDYEAVSQALIVFEFKAKGKDSQYQAIDEAIRSYRFIRNSCLRYWMDNKKVGKY


ID
DLNKYCKVLAKQYPFANKLNSQARQSAAECSWSAISRFYDNCKRKVSGKKGFPKFKK


NO:
HARSVEYKTSGWKLSENRKAITFTDKNGIGKLKLKGTYDLHFSQLEDMKRVRLVRRA


87
DGYYVQFCISVDVKVETEPTGKAIGLDVGIKYFLADSSGNTIENPQFYRKAEKKLNRA



NRRKSKKYIRGVKPQSKNYHKARCRYARKHLRVSRQRKEYCKRVAYCVIHSNDVVA



YEDLNVKGMVKNRHLAKSISDVAWSTFRHWLEYFAIKYGKLTIPVAPHNTSQNCSNC



DKKVPKSLSTRTHICHHCGYSEDRDVNAAKNILKKALSTVGQTGSLKLGEIEPLLVLEQ



SCTRKFDL





SEQ
LAEENTLHLTLAMSLPLNDLPENRTRSELWRRQWLPQKKLSLLLGVNQSVRKAAADC


ID
LRWFEPYQELLWWEPTDPDGKKLLDKEGRPIKRTAGHMRVLRKLEEIAPFRGYQLGS


NO:
AVKNGLRHKVADLLLSYAKRKLDPQFTDKTSYPSIGDQFPIVWTGAFVCYEQSITGQL


88
YLYLPLFPRGSHQEDITNNYDPDRGPALQVFGEKEIARLSRSTSGLLLPLQFDKWGEAT



FIRGENNPPTWKATHRRSDKKWLSEVLLREKDFQPKRVELLVRNGRIFVNVACEIPTK



PLLEVENFMGVSFGLEHLVTVVVINRDGNVVHQRQEPARRYEKTYFARLERLRRRGG



PFSQELETFHYRQVAQIVEEALRFKSVPAVEQVGNIPKGRYNPRLNLRLSYWPFGKLA



DLTSYKAVKEGLPKPYSVYSATAKMLCSTCGAANKEGDQPISLKGPTVYCGNCGTRH



NTGFNTALNLARRAQELFVKGVVAR





SEQ
MSQSLLKWHDMAGRDKDASRSLQKSAVEGVLLHLTASHRVALEMLEKSVSQTVAVT


ID
MEAAQQRLVIVLEDDPTKATSRKRVISADLQFTREEFGSLPNWAQKLASTCPEIATKY


NO:
ADKHINSIRIAWGVAKESTNGDAVEQKLQWQIRLLDVTMFLQQLVLQLADKALLEQIP


89
SSIRGGIGQEVAQQVTSHIQLLDSGTVLKAELPTISDRNSELARKQWEDAIQTVCTYAL



PFSRERARILDPGKYAAEDPRGDRLINIDPMWARVLKGPTVKSLPLLFVSGSSIRIVKLT



LPRKHAAGHKHTFTATYLVLPVSREWINSLPGTVQEKVQWWKKPDVLATQELLVGK



GALKKSANTLVIPISAGKKRFFNHILPALQRGFPLQWQRIVGRSYRRPATHRKWFAQLT



IGYTNPSSLPEMALGIHFGMKDILWWALADKQGNILKDGSIPGNSILDFSLQEKGKIER



QQKAGKNVAGKKYGKSLLNATYRVVNGVLEFSKGISAEHASQPIGLGLETIRFVDKAS



GSSPVNARHSNWNYGQLSGIFANKAGPAGFSVTEITLKKAQRDLSDAEQARVLAIEAT



KRFASRIKRLATKRKDDTLFV





SEQ
VEPVEKERFYYRTYTFRLDGQPRTQNLTTQSGWGLLTKAVLDNTKHYWEIVHHARIA


ID
NQPIVFENPVIDEQGNPKLNKLGQPRFWKRPISDIVNQLRALFENQNPYQLGSSLIQGT


NO:
YWDVAENLASWYALNKEYLAGTATWGEPSFPEPHPLTEINQWMPLTFSSGKVVRLLK


90
NASGRYFIGLPILGENNPCYRMRTIEKLIPCDGKGRVTSGSLILFPLVGIYAQQHRRMTD



ICESIRTEKGKLAWAQVSIDYVREVDKRRRMRRTRKSQGWIQGPWQEVFILRLVLAHK



APKLYKPRCFAGISLGPKTLASCVILDQDERVVEKQQWSGSELLSLIHQGEERLRSLRE



QSKPTWNAAYRKQLKSLINTQVFTIVTFLRERGAAVRLESIARVRKSTPAPPVNFLLSH



WAYRQITERLKDLAIRNGMPLTHSNGSYGVRFTCSQCGATNQGIKDPTKYKVDIESET



FLCSICSHREIAAVNTATNLAKQLLDE





SEQ
MNDTETSETLTSHRTVCAHLHVVGETGSLPRLVEAALAELITLNGRATQALLSLAKNG


ID
LVLRRDKEENLIAAELTLPCRKNKYADVAAKAGEPILATRINNKGKLVTKKWYGEGN


NO:
SYHIVRFTPETGMFTVRVFDRYAFDEELLHLHSEVVFGSDLPKGIKAKTDSLPANFLQA


91
VFTSFLELPFQGFPDIVVKPAMKQAAEQLLSYVQLEAGENQQAEYPDTNERDPELRLV



EWQKSLHELSVRTEPFEFVRARDIDYYAETDRRGNRFVNITPEWTKFAESPFARRLPLK



IPPEFCILLRRKTEGHAKIPNRIYLGLQIFDGVTPDSTLGVLATAEDGKLFWWHDHLDE



FSNLEGKPEPKLKNKPQLLMVSLEYDREQRFEESVGGDRKICLVTLKETRNFRRGWNG



RILGIHFQHNPVITWALMDHDAEVLEKGFIEGNAFLGKALDKQALNEYLQKGGKWVG



DRSFGNKLKGITHTLASLIVRLAREKDAWIALEEISWVQKQSADSVANHEIVEQPHHSL



TR





SEQ
MNDTETSETLTSHRTVCAHLHVVGETGSLPRLVEAALAELITLNGRATQALLSLAKNG


ID
LVLRRDKEENLIAAELTLPCRKNKYADVAAKAGEPILATRINNKGKLVTKKWYGEGN


NO:
SYHIVRFTPETGMFTVRVFDRYAFDEELLHLHSEVVFGSDLPKGIKAKTDSLPANFLQA


92
VFTSFLELPFQGFPDIVVKPAMKQAAEQLLSYVQLEAGENQQAEYPDTNERDPELRLV



EWQKSLHELSVRTEPFEFVRARDIDYYAETDRRGNRFVNITPEWTKFAESPFARRLPLK



IPPEFCILLRRKTEGHAKIPNRIYLGLQIFDGVTPDSTLGVLATAEDGKLFWWHDHLDE



FSNLEGKPEPKLKNKPQLLMVSLEYDREQRFEESVGGDRKICLVTLKETRNFRRGRHG



HTRTDRLPAGNTLWRADFATSAEVAAPKWNGRILGIHFQHNPVITWALMDHDAEVLE



KGFIEGNAFLGKALDKQALNEYLQKGGKWVGDRSFGNKLKGITHTLASLIVRLAREK



DAWIALEEISWVQKQSADSVANRRFSMWNYSRLATLIEWLGTDIATRDCGTAAPLAH



KVSDYLTHFTCPECGACRKAGQKKEIADTVRAGDILTCRKCGFSGPIPDNFIAEFVAKK



ALERMLKKKPV





SEQ
MAKRNFGEKSEALYRAVRFEVRPSKEELSILLAVSEVLRMLFNSALAERQQVFTEFIAS


ID
LYAELKSASVPEEISEIRKKLREAYKEHSISLFDQINALTARRVEDEAFASVTRNWQEET


NO:
LDALDGAYKSFLSLRRKGDYDAHSPRSRDSGFFQKIPGRSGFKIGEGRIALSCGAGRKL


93
SFPIPDYQQGRLAETTKLKKFELYRDQPNLAKSGRFWISVVYELPKPEATTCQSEQVAF



VALGASSIGVVSQRGEEVIALWRSDKHWVPKIEAVEERMKRRVKGSRGWLRLLNSGK



RRMHMISSRQHVQDEREIVDYLVRNHGSHFVVTELVVRSKEGKLADSSKPERGGSLG



LNWAAQNTGSLSRLVRQLEEKVKEHGGSVRKHKLTLTEAPPARGAENKLWMARKLR



ESFLKEV





SEQ
LAKNDEKELLYQSVKFEIYPDESKIRVLTRVSNILVLVWNSALGERRARFELYIAPLYE


ID
ELKKFPRKSAESNALRQKIREGYKEHIPTFFDQLKKLLTPMRKEDPALLGSVPRAYQEE


NO:
TLNTLNGSFVSFMTLRRNNDMDAKPPKGRAEDRFHEISGRSGFKIDGSEFVLSTKEQKL


94
RFPIPNYQLEKLKEAKQIKKFTLYQSRDRRFWISIAYEIELPDQRPFNPEEVIYIAFGASSI



GVISPEGEKVIDFWRPDKHWKPKIKEVENRMRSCKKGSRAWKKRAAARRKMYAMTQ



RQQKLNHREIVASLLRLGFHFVVTEYTVRSKPGKLADGSNPKRGGAPQGFNWSAQNT



GSFGEFILWLKQKVKEQGGTVQTFRLVLGQSERPEKRGRDNKIEMVRLLREKYLESQT



IVV





SEQ
MAKGKKKEGKPLYRAVRFEIFPTSDQITLFLRVSKNLQQVWNEAWQERQSCYEQFFG


ID
SIYERIGQAKKRAQEAGFSEVWENEAKKGLNKKLRQQEISMQLVSEKESLLQELSIAFQ


NO:
EHGVTLYDQINGLTARRIIGEFALIPRNWQEETLDSLDGSFKSFLALRKNGDPDAKPPR


95
QRVSENSFYKIPGRSGFKVSNGQIYLSFGKIGQTLTSVIPEFQLKRLETAIKLKKFELCRD



ERDMAKPGRFWISVAYEIPKPEKVPVVSKQITYLAIGASRLGVVSPKGEFCLNLPRSDY



HWKPQINALQERLEGVVKGSRKWKKRMAACTRMFAKLGHQQKQHGQYEVVKKLLR



HGVHFVVTELKVRSKPGALADASKSDRKGSPTGPNWSAQNTGNIARLIQKLTDKASE



HGGTVIKRNPPLLSLEERQLPDAQRKIFIAKKLREEFLADQK





SEQ
MAKREKKDDVVLRGTKMRIYPTDRQVTLMDMWRRRCISLWNLLLNLETAAYGAKN


ID
TRSKLGWRSIWARVVEENHAKALIVYQHGKCKKDGSFVLKRDGTVKHPPRERFPGDR


NO:
KILLGLFDALRHTLDKGAKCKCNVNQPYALTRAWLDETGHGARTADIIAWLKDFKGE


96
CDCTAISTAAKYCPAPPTAELLTKIKRAAPADDLPVDQAILLDLFGALRGGLKQKECD



HTHARTVAYFEKHELAGRAEDILAWLIAHGGTCDCKIVEEAANHCPGPRLFIWEHELA



MIMARLKAEPRTEWIGDLPSHAAQTVVKDLVKALQTMLKERAKAAAGDESARKTGF



PKFKKQAYAAGSVYFPNTTMFFDVAAGRVQLPNGCGSMRCEIPRQLVAELLERNLKP



GLVIGAQLGLLGGRIWRQGDRWYLSCQWERPQPTLLPKTGRTAGVKIAASIVFTTYDN



RGQTKEYPMPPADKKLTAVHLVAGKQNSRALEAQKEKEKKLKARKERLRLGKLEKG



HDPNALKPLKRPRVRRSKLFYKSAARLAACEAIERDRRDGFLHRVTNEIVHKFDAVSV



QKMSVAPMMRRQKQKEKQIESKKNEAKKEDNGAAKKPRNLKPVRKLLRHVAMARG



RQFLEYKYNDLRGPGSVLIADRLEPEVQECSRCGTKNPQMKDGRRLLRCIGVLPDGTD



CDAVLPRNRNAARNAEKRLRKHREAHNA





SEQ
MNEVLPIPAVGEDAADTIMRGSKMRIYPSVRQAATMDLWRRRCIQLWNLLLELEQAA


ID
YSGENRRTQIGWRSIWATVVEDSHAEAVRVAREGKKRKDGTFRKAPSGKEIPPLDPA


NO:
MLAKIQRQMNGAVDVDPKTGEVTPAQPRLFMWEHELQKIMARLKQAPRTHWIDDLP


97
SHAAQSVVKDLIKALQAMLRERKKRASGIGGRDTGFPKFKKNRYAAGSVYFANTQLR



FEAKRGKAGDPDAVRGEFARVKLPNGVGWMECRMPRHINAAHAYAQATLMGGRIW



RQGENWYLSCQWKMPKPAPLPRAGRTAAIKIAAAIPITTVDNRGQTREYAMPPIDRERI



AAHAAAGRAQSRALEARKRRAKKREAYAKKRHAKKLERGIAAKPPGRARIKLSPGFY



AAAAKLAKLEAEDANAREAWLHEITTQIVRNFDVIAVPRMEVAKLMKKPEPPEEKEE



QVKAPWQGKRRSLKAARVMMRRTAMALIQTTLKYKAVDLRGPQAYEEIAPLDVTAA



ACSGCGVLKPEWKMARAKGREIMRCQEPLPGGKTCNTVLTYTRNSARVIGRELAVRL



AERQKA





SEQ
MTTQKTYNFCFYDQRFFELSKEAGEVYSRSLEEFWKIYDETGVWLSKFDLQKHMRNK


ID
LERKLLHSDSFLGAMQQVHANLASWKQAKKVVPDACPPRKPKFLQAILFKKSQIKYK


NO:
NGFLRLTLGTEKEFLYLKWDINIPLPIYGSVTYSKTRGWKINLCLETEVEQKNLSENKY


98
LSIDLGVKRVATIFDGENTITLSGKKFMGLMHYRNKLNGKTQSRLSHKKKGSNNYKKI



QRAKRKTTDRLLNIQKEMLHKYSSFIVNYAIRNDIGNIIIGDNSSTHDSPNMRGKTNQKI



SQNPEQKLKNYIKYKFESISGRVDIVPEPYTSRKCPHCKNIKKSSPKGRTYKCKKCGFIF



DRDGVGAINIYNENVSFGQIISPGRIRSLTEPIGMKFHNEIYFKSYVAA





SEQ
MSVRSFQARVECDKQTMEHLWRTHKVFNERLPEIIKILFKMKRGECGQNDKQKSLYK


ID
SISQSILEANAQNADYLLNSVSIKGWKPGTAKKYRNASFTWADDAAKLSSQGIHVYDK


NO:
KQVLGDLPGMMSQMVCRQSVEAISGHIELTKKWEKEHNEWLKEKEKWESEDEHKKY


99
LDLREKFEQFEQSIGGKITKRRGRWHLYLKWLSDNPDFAAWRGNKAVINPLSEKAQIR



INKAKPNKKNSVERDEFFKANPEMKALDNLHGYYERNFVRRRKTKKNPDGFDHKPTF



TLPHPTIHPRWFVFNKPKTNPEGYRKLILPKKAGDLGSLEMRLLTGEKNKGNYPDDWI



SVKFKADPRLSLIRPVKGRRVVRKGKEQGQTKETDSYEFFDKHLKKWRPAKLSGVKLI



FPDKTPKAAYLYFTCDIPDEPLTETAKKIQWLETGDVTKKGKKRKKKVLPHGLVSCAV



DLSMRRGTTGFATLCRYENGKIHILRSRNLWVGYKEGKGCHPYRWTEGPDLGHIAKH



KREIRILRSKRGKPVKGEESHIDLQKHIDYMGEDRFKKAARTIVNFALNTENAASKNGF



YPRADVLLLENLEGLIPDAEKERGINRALAGWNRRHLVERVIEMAKDAGFKRRVFEIP



PYGTSQVCSKCGALGRRYSIIRENNRREIRFGYVEKLFACPNCGYCANADHNASVNLN



RRFLIEDSFKSYYDWKRLSEKKQKEEIETIESKLMDKLCAMHKISRGSISK





SEQ
MHLWRTHCVFNQRLPALLKRLFAMRRGEVGGNEAQRQVYQRVAQFVLARDAKDSV


ID
DLLNAVSLRKRSANSAFKKKATISCNGQAREVTGEEVFAEAVALASKGVFAYDKDDM


NO:
RAGLPDSLFQPLTRDAVACMRSHEELVATWKKEYREWRDRKSEWEAEPEHALYLNL


100
RPKFEEGEAARGGRFRKRAERDHAYLDWLEANPQLAAWRRKAPPAVVPIDEAGKRRI



ARAKAWKQASVRAEEFWKRNPELHALHKIHVQYLREFVRPRRTRRNKRREGFKQRPT



FTMPDPVRHPRWCLFNAPQTSPQGYRLLRLPQSRRTVGSVELRLLTGPSDGAGFPDAW



VNVRFKADPRLAQLRPVKVPRTVTRGKNKGAKVEADGFRYYDDQLLIERDAQVSGV



KLLFRDIRMAPFADKPIEDRLLSATPYLVFAVEIKDEARTERAKAIRFDETSELTKSGKK



RKTLPAGLVSVAVDLDTRGVGFLTRAVIGVPEIQQTHHGVRLLQSRYVAVGQVEARA



SGEAEWSPGPDLAHIARHKREIRRLRQLRGKPVKGERSHVRLQAHIDRMGEDRFKKA



ARKIVNEALRGSNPAAGDPYTRADVLLYESLETLLPDAERERGINRALLRWNRAKLIE



HLKRMCDDAGIRHFPVSPFGTSQVCSKCGALGRRYSLARENGRAVIRFGWVERLFACP



NPECPGRRPDRPDRPFTCNSDHNASVNLHRVFALGDQAVAAFRALAPRDSPARTLAV



KRVEDTLRPQLMRVHKLADAGVDSPF





SEQ
MATLVYRYGVRAHGSARQQDAVVSDPAMLEQLRLGHELRNALVGVQHRYEDGKRA


ID
VWSGFASVAAADHRVTTGETAVAELEKQARAEHSADRTAATRQGTAESLKAARAAV


NO:
KQARADRKAAMAAVAEQAKPKIQALGDDRDAEIKDLYRRFCQDGVLLPRCGRCAGD


101
LRSDGDCTDCGAAHEPRKLYWATYNAIREDHQTAVKLVEAKRKAGQPARLRFRRWT



GDGTLTVQLQRMHGPACRCVTCAEKLTRRARKTDPQAPAVAADPAYPPTDPPRDPAL



LASGQGKWRNVLQLGTWIPPGEWSAMSRAERRRVGRSHIGWQLGGGRQLTLPVQLH



RQMPADADVAMAQLTRVRVGGRHRMSVALTAKLPDPPQVQGLPPVALHLGWRQRP



DGSLRVATWACPQPLDLPPAVADVVVSHGGRWGEVIMPARWLADAEVPPRLLGRRD



KAMEPVLEALADWLEAHTEACTARMTPALVRRWRSQGRLAGLTNRWRGQPPTGSAE



ILTYLEAWRIQDKLLWERESHLRRRLAARRDDAWRRVASWLARHAGVLVVDDADIA



ELRRRDDPADTDPTMPASAAQAARARAALAAPGRLRHLATITATRDGLGVHTVASAG



LTRLHRKCGHQAQPDPRYAASAVVTCPGCGNGYDQDYNAAMLMLDRQQQP





SEQ
MSRVELHRAYKFRLYPTPAQVAELAEWERQLRRLYNLAHSQRLAAMQRHVRPKSPG


ID
VLKSECLSCGAVAVAEIGTDGKAKKTVKHAVGCSVLECRSCGGSPDAEGRTAHTAAC


NO
SFVDYYRQGREMTQLLEEDDQLARVVCSARQETLRDLEKAWQRWHKMPGFGKPHF


102
KKRIDSCRIYFSTPKSWAVDLGYLSFTGVASSVGRIKIRQDRVWPGDAKFSSCHVVRD



VDEWYAVFPLTFTKEIEKPKGGAVGINRGAVHAIADSTGRVVDSPKFYARSLGVIRHR



ARLLDRKVPFGRAVKPSPTKYHGLPKADIDAAAARVNASPGRLVYEARARGSIAAAE



AHLAALVLPAPRQTSQLPSEGRNRERARRFLALAHQRVRRQREWFLHNESAHYAQSY



TKIAIEDWSTKEMTSSEPRDAEEMKRVTRARNRSILDVGWYELGRQIAYKSEATGAEF



AKVDPGLRETETHVPEAIVRERDVDVSGMLRGEAGISGTCSRCGGLLRASASGHADAE



CEVCLHVEVGDVNAAVNVLKRAMFPGAAPPSKEKAKVTIGIKGRKKKRAA





SEQ
MSRVELHRAYKFRLYPTPVQVAELSEWERQLRRLYNLGHEQRLLTLTRHLRPKSPGV


ID
LKGECLSCDSTQVQEVGADGRPKTTVRHAEQCPTLACRSCGALRDAEGRTAHTVACA


NO:
FVDYYRQGREMTELLAADDQLARVVCSARQEVLRDLDKAWQRWRKMPGFGKPRFK


103
RRTDSCRIYFSTPKAWKLEGGHLSFTGAATTVGAIKMRQDRNWPASVQFSSCHVVRD



VDEWYAVFPLTFVAEVARPKGGAVGINRGAVHAIADSTGRVVDSPRYYARALGVIRH



RARLFDRKVPSGHAVKPSPTKYRGLSAIEVDRVARATGFTPGRVVTEALNRGGVAYA



ECALAAIAVLGHGPERPLTSDGRNREKARKFLALAHQRVRRQREWFLHNESAHYART



YSKIAIEDWSTKEMTASEPQGEETRRVTRSRNRSILDVGWYELGRQLAYKTEATGAEF



AQVDPGLKETETNVPKAIADARDVDVSGMLRGEAGISGTCSKCGGLLRAPASGHADA



ECEICLNVEVGDVNAAVNVLKRAMFPGDAPPASGEKPKVSIGIKGRQKKKKAA





SEQ
MEAIATGMSPERRVELGILPGSVELKRAYKFRLYPMKVQQAELSEWERQLRRLYNLA


ID
HEQRLAALLRYRDWDFQKGACPSCRVAVPGVHTAACDHVDYFRQAREMTQLLEVD


NO:
AQLSRVICCARQEVLRDLDKAWQRWRKKLGGRPRFKRRTDSCRIYLSTPKHWEIAGR


104
YLRLSGLASSVGEIRIEQDRAFPEGALLSSCSIVRDVDEWYACLPLTFTQPIERAPHRSV



GLNRGVVHALADSDGRVVDSPKFFERALATVQKRSRDLARKVSGSRNAHKARIKLAK



AHQRVRRQRAAFLHQESAYYSKGFDLVALEDMSVRKMTATAGEAPEMGRGAQRDL



NRGILDVGWYELARQIDYKRLAHGGELLRVDPGQTTPLACVTEEQPARGISSACAVCG



IPLARPASGNARMRCTACGSSQVGDVNAAENVLTRALSSAPSGPKSPKASIKIKGRQK



RLGTPANRAGEASGGDPPVRGPVEGGTLAYVVEPVSESQSDT





SEQ
MTVRTYKYRAYPTPEQAEALTSWLRFASQLYNAALEHRKNAWGRHDAHGRGFRFW


ID
DGDAAPRKKSDPPGRWVYRGGGGAHISKNDQGKLLTEFRREHAELLPPGMPALVQHE


NO:
VLARLERSMAAFFQRATKGQKAGYPRWRSEHRYDSLTFGLTSPSKERFDPETGESLGR


105
GKTVGAGTYHNGDLRLTGLGELRILEHRRIPMGAIPKSVIVRRSGKRWFVSIAMEMPS



VEPAASGRPAVGLDMGVVTWGTAFTADTSAAAALVADLRRMATDPSDCRRLEELER



EAAQLSEVLAHCRARGLDPARPRRCPKELTKLYRRSLHRLGELDRACARIRRRLQAAH



DIAEPVPDEAGSAVLIEGSNAGMRHARRVARTQRRVARRTRAGHAHSNRRKKAVQA



YARAKERERSARGDHRHKVSRALVRQFEEISVEALDIKQLTVAPEHNPDPQPDLPAHV



QRRRNRGELDAAWGAFFAALDYKAADAGGRVARKPAPHTTQECARCGTLVPKPISLR



VHRCPACGYTAPRTVNSARNVLQRPLEEPGRAGPSGANGRGVPHAVA





SEQ
MNCRYRYRIYPTPGQRQSLARLFGCVRVVWNDALFLCRQSEKLPKNSELQKLCITQA


ID
KKTEARGWLGQVSAIPLQQSVADLGVAFKNFFQSRSGKRKGKKVNPPRVKRRNNRQ


NO:
GARFTRGGFKVKTSKVYLARIGDIKIKWSRPLPSEPSSVTVIKDCAGQYFLSFVVEVKP


106
EIKPPKNPSIGIDLGLKTFASCSNGEKIDSPDYSRLYRKLKRCQRRLAKRQRGSKRRER



MRVKVAKLNAQIRDKRKDFLHKLSTKVVNENQVIALEDLNVGGMLKNRKLSRAISQA



GWYEFRSLCEGKAEKHNRDFRVISRWEPTSQVCSECGYRWGKIDLSVRSIVCINCGVE



HDRDDNASVNIEQAGLKVGVGHTHDSKRTGSACKTSNGAVCVEPSTHREYVQLTLFD



W





SEQ
MKSRWTFRCYPTPEQEQHLARTFGCVRFVWNWALRARTDAFRAGERIGYPATDKAL


ID
TLLKQQPETVWLNEVSSVCLQQALRDLQVAFSNFFDKRAAHPSFKRKEARQSANYTE


NO:
RGFSFDHERRILKLAKIGAIKVKWSRKAIPHPSSIRLIRTASGKYFVSLVVETQPAPMPE


107
TGESVGVDFGVARLATLSNGERISNPKHGAKWQRRLAFYQKRLARATKGSKRRMRIK



RHVARIHEKIGNSRSDTLHKLSTDLVTRFDLICVEDLNLRGMVKNHSLARSLHDASIGS



AIRMIEEKAERYGKNVVKIDRWFPSSKTCSDCGHIVEQLPLNVREWTCPECGTTHDRD



ANAAANILAVGQTVSAHGGTVRRSRAKASERKSQRSANRQGVNRA





SEQ
KEPLNIGKTAKAVFKEIDPTSLNRAANYDASIELNCKECKFKPFKNVKRYEFNFYNNW


ID
YRCNPNSCLQSTYKAQVRKVEIGYEKLKNEILTQMQYYPWFGRLYQNFFHDERDKMT


NO:
SLDEIQVIGVQNKVFFNTVEKAWREIIKKRFKDNKETMETIPELKHAAGHGKRKLSNK


108
SLLRRRFAFVQKSFKFVDNSDVSYRSFSNNIACVLPSRIGVDLGGVISRNPKREYIPQEIS



FNAFWKQHEGLKKGRNIEIQSVQYKGETVKRIEADTGEDKAWGKNRQRRFTSLILKL



VPKQGGKKVWKYPEKRNEGNYEYFPIPIEFILDSGETSIRFGGDEGEAGKQKHLVIPFN



DSKATPLASQQTLLENSRFNAEVKSCIGLAIYANYFYGYARNYVISSIYHKNSKNGQAI



TAIYLESIAHNYVKAIERQLQNLLLNLRDFSFMESHKKELKKYFGGDLEGTGGAQKRR



EKEEKIEKEIEQSYLPRLIRLSLTKMVTKQVEM





SEQ
ELIVNENKDPLNIGKTAKAVFKEIDPTSINRAANYDASIELACKECKFKPFNNTKRHDFS


ID
FYSNWHRCSPNSCLQSTYRAKIRKTEIGYEKLKNEILNQMQYYPWFGRLYQNFFNDQR


NO:
DKMTSLDEIQVTGVQNKIFFNTVEKAWREIIKKRFRDNKETMRTIPDLKNKSGHGSRK


109
LSNKSLLRRRFAFAQKSFKLVDNSDVSYRAFSNNVACVLPSKIGVDIGGIINKDLKREYI



PQEITFNVFWKQHDGLKKGRNIEIHSVQYKGEIVKRIEADTGEDKAWGKNRQRRFTSL



ILKITPKQGGKKIWKFPEKKNASDYEYFPIPIEFILDNGDASIKFGGEEGEVGKQKHLLIP



FNDSKATPLSSKQMLLETSRFNAEVKSTIGLALYANYFVSYARNYVIKSTYHKNSKKG



QIVTEIYLESISQNFVRAIQRQLQSLMLNLKDWGFMQTHKKELKKYFGSDLEGSKGGQ



KRREKEEKIEKEIEASYLPRLIRLSLTKSVTKAEEM





SEQ
PEEKTSKLKPNSINLAANYDANEKFNCKECKFHPFKNKKRYEFNFYNNLHGCKSCTKS


ID
TNNPAVKRIEIGYQKLKFEIKNQMEAYPWFGRLRINFYSDEKRKMSELNEMQVTGVK


NO:
NKIFFDAIECAWREILKKRFRESKETLITIPKLKNKAGHGARKHRNKKLLIRRRAFMKK


110
NFHFLDNDSISYRSFANNIACVLPSKVGVDIGGIISPDVGKDIKPVDISLNLMWASKEGI



KSGRKVEIYSTQYDGNMVKKIEAETGEDKSWGKNRKRRQTSLLLSIPKPSKQVQEFDF



KEWPRYKDIEKKVQWRGFPIKIIFDSNHNSIEFGTYQGGKQKVLPIPFNDSKTTPLGSK



MNKLEKLRFNSKIKSRLGSAIAANKFLEAARTYCVDSLYHEVSSANAIGKGKIFIEYYL



EILSQNYIEAAQKQLQRFIESIEQWFVADPFQGRLKQYFKDDLKRAKCFLCANREVQTT



CYAAVKLHKSCAEKVKDKNKELAIKERNNKEDAVIKEVEASNYPRVIRLKLTKTITNK



AM





SEQ
SESENKIIEQYYAFLYSFRDKYEKPEFKNRGDIKRKLQNKWEDFLKEQNLKNDKKLSN


ID
YIFSNRNFRRSYDREEENEEGIDEKKSKPKRINCFEKEKNLKDQYDKDAINASANKDG


NO:
AQKWGCFECIFFPMYKIESGDPNKRIIINKTRFKLFDFYLNLKGCKSCLRSTYHPYRSNV


111
YIESNYDKLKREIGNFLQQKNIFQRMRKAKVSEGKYLTNLDEYRLSCVAMHFKNRWL



FFDSIQKVLRETIKQRLKQMRESYDEQAKTKRSKGHGRAKYEDQVRMIRRRAYSAQA



HKLLDNGYITLFDYDDKEINKVCLTAINQEGFDIGGYLNSDIDNVMPPIEISFHLKWKY



NEPILNIESPFSKAKISDYLRKIREDLNLERGKEGKARSKKNVRRKVLASKGEDGYKKI



FTDFFSKWKEELEGNAMERVLSQSSGDIQWSKKKRIHYTTLVLNINLLDKKGVGNLK



YYEIAEKTKILSFDKNENKFWPITIQVLLDGYEIGTEYDEIKQLNEKTSKQFTIYDPNTKI



IKIPFTDSKAVPLGMLGINIATLKTVKKTERDIKVSKIFKGGLNSKIVSKIGKGIYAGYFP



TVDKEILEEVEEDTLDNEFSSKSQRNIFLKSIIKNYDKMLKEQLFDFYSFLVRNDLGVRF



LTDRELQNIEDESFNLEKRFFETDRDRIARWFDNTNTDDGKEKFKKLANEIVDSYKPRL



IRLPVVRVIKRIQPVKQREM





SEQ
KYSTRDFSELNEIQVTACKQDEFFKVIQNAWREIIKKRFLENRENFIEKKIFKNKKGRG


ID
KRQESDKTIQRNRASVMKNFQLIENEKIILRAPSGHVACVFPVKVGLDIGGFKTDDLEK


NO:
NIFPPRTITINVFWKNRDRQRKGRKLEVWGIKARTKLIEKVHKWDKLEEVKKKRLKSL


112
EQKQEKSLDNWSEVNNDSFYKVQIDELQEKIDKSLKGRTMNKILDNKAKESKEAEGL



YIEWEKDFEGEMLRRIEASTGGEEKWGKRRQRRHTSLLLDIKNNSRGSKEIINFYSYAK



QGKKEKKIEFFPFPLTITLDAEEESPLNIKSIPIEDKNATSKYFSIPFTETRATPLSILGDRV



QKFKTKNISGAIKRNLGSSISSCKIVQNAETSAKSILSLPNVKEDNNMEIFINTMSKNYF



RAMMKQMESFIFEMEPKTLIDPYKEKAIKWFEVAASSRAKRKLKKLSKADIKKSELLL



SNTEEFEKEKQEKLEALEKEIEEFYLPRIVRLQLTKTILETPVM





SEQ
KKLQLLGHKILLKEYDPNAVNAAANFETSTAELCGQCKMKPFKNKRRFQYTFGKNYH


ID
GCLSCIQNVYYAKKRIVQIAKEELKHQLTDSIASIPYKYTSLFSNTNSIDELYILKQERA


NO:
AFFSNTNSIDELYITGIENNIAFKVISAIWDEIIKKRRQRYAESLTDTGTVKANRGHGGT


113
AYKSNTRQEKIRALQKQTLHMVTNPYISLARYKNNYIVATLPRTIGMHIGAIKDRDPQ



KKLSDYAINFNVFWSDDRQLIELSTVQYTGDMVRKIEAETGENNKWGENMKRTKTSL



LLEILTKKTTDELTFKDWAFSTKKEIDSVTKKTYQGFPIGIIFEGNESSVKFGSQNYFPLP



FDAKITPPTAEGFRLDWLRKGSFSSQMKTSYGLAIYSNKVTNAIPAYVIKNMFYKIARA



ENGKQIKAKFLKKYLDIAGNNYVPFIIMQHYRVLDTFEEMPISQPKVIRLSLTKTQHIIIK



KDKTDSKM





SEQ
NTSNLINLGKKAINISANYDANLEVGCKNCKFLSSNGNFPRQTNVKEGCHSCEKSTYEP


ID
SIYLVKIGERKAKYDVLDSLKKFTFQSLKYQSKKSMKSRNKKPKELKEFVIFANKNKA


NO:
FDVIQKSYNHLILQIKKEINRMNSKKRKKNHKRRLFRDREKQLNKLRLIESSNLFLPRE


114
NKGNNHVFTYVAIHSVGRDIGVIGSYDEKLNFETELTYQLYFNDDKRLLYAYKPKQN



KIIKIKEKLWNLRKEKEPLDLEYEKPLNKSITFSIKNDNLFKVSKDLMLRRAKFNIQGKE



KLSKEERKINRDLIKIKGLVNSMSYGRFDELKKEKNIWSPHIYREVRQKEIKPCLIKNGD



RIEIFEQLKKKMERLRRFREKRQKKISKDLIFAERIAYNFHTKSIKNTSNKINIDQEAKRG



KASYMRKRIGYETFKNKYCEQCLSKGNVYRNVQKGCSCFENPFDWIKKGDENLLPKK



NEDLRVKGAFRDEALEKQIVKIAFNIAKGYEDFYDNLGESTEKDLKLKFKVGTTINEQ



ESLKL





SEQ
TSNPIKLGKKAINISANYDSNLQIGCKNCKFLSYNGNFPRQTNVKEGCHSCEKSTYEPP


ID
VYTVRIGERRSKYDVLDSLKKFIFLSLKYRQSKKMKTRSKGIRGLEEFVISANLKKAM


NO:
DVIQKSYRHLILNIKNEIVRMNGKKRNKNHKRLLFRDREKQLNKLRLIEGSSFFKPPTV


115
KGDNSIFTCVAIHNIGRDIGIAGDYFDKLEPKIELTYQLYYEYNPKKESEINKRLLYAYK



PKQNKIIEIKEKLWNLRKEKSPLDLEYEKPLTKSITFLVKRDGVFRISKDLMLRKAKFII



QGKEKLSKEERKINRDLIKIKSNIISLTYGRFDELKKDKTIWSPHIFRDVKQGKITPCIER



KGDRMDIFQQLRKKSERLRENRKKRQKKISKDLIFAERIAYNFHTKSIKNTSNLINIKHE



AKRGKASYMRKRIGNETFRIKYCEQCFPKNNVYKNVQKGCSCFEDPFEYIKKGNEDLI



PNKNQDLKAKGAFRDDALEKQIIKVAFNIAKGYEDFYENLKKTTEKDIRLKFKVGTIIS



EEM





SEQ
NNSINLSKKAINISANYDANLQVRCKNCKFLSSNGNFPRQTDVKEGCHSCEKSTYEPPV


ID
YDVKIGEIKAKYEVLDSLKKFTFQSLKYQLSKSMKFRSKKIKELKEFVIFAKESKALNV


NO:
INRSYKHLILNIKNDINRMNSKKRIKNHKGRLFLDRQKQLSKLKLIEGSSFFVPAKNVG


116
NKSVFTCVAIHSIGRDIGIAGLYDSFTKPVNEITYQIFFSGERRLLYAYKPKQLKILSIKE



NLWSLKNEKKPLDLLYEKPLGKNLNFNVKGGDLFRVSKDLMIRNAKFNVHGRQRLSD



EERLINRNFIKIKGEVVSLSYGRFEELKKDRKLWSPHIFKDVRQNKIKPCLVMQGQRIDI



FEQLKRKLELLKKIRKSRQKKLSKDLIFGERIAYNFHTKSIKNTSNKINIDSDAKRGRAS



YMRKRIGNETFKLKYCDVCFPKANVYRRVQNGCSCSENPYNYIKKGDKDLLPKKDEG



LAIKGAFRDEKLNKQIIKVAFNIAKGYEDFYDDLKKRTEKDVDLKFKIGTTVLDQKPM



EIFDGIVITWL





SEQ
LLTTVVETNNLAKKAINVAANFDANIDRQYYRCTPNLCRFIAQSPRETKEKDAGCSSC


ID
TQSTYDPKVYVIKIGKLLAKYEILKSLKRFLFMNRYFKQKKTERAQQKQKIGTELNEM


NO:
SIFAKATNAMEVIKRATKHCTYDIIPETKSLQMLKRRRHRVKVRSLLKILKERRMKIKK


117
IPNTFIEIPKQAKKNKSDYYVAAALKSCGIDVGLCGAYEKNAEVEAEYTYQLYYEYKG



NSSTKRILYCYNNPQKNIREFWEAFYIQGSKSHVNTPGTIRLKMEKFLSPITIESEALDFR



VWNSDLKIRNGQYGFIKKRSLGKEAREIKKGMGDIKRKIGNLTYGKSPSELKSIHVYRT



ERENPKKPRAARKKEDNFMEIFEMQRKKDYEVNKKRRKEATDAAKIMDFAEEPIRHY



HTNNLKAVRRIDMNEQVERKKTSVFLKRIMQNGYRGNYCRKCIKAPEGSNRDENVLE



KNEGCLDCIGSEFIWKKSSKEKKGLWHTNRLLRRIRLQCFTTAKAYENFYNDLFEKKE



SSLDIIKLKVSITTKSM





SEQ
ASTMNLAKQAINFAANYDSNLEIGCKGCKFMSTWSKKSNPKFYPRQNNQANKCHSCT


ID
YSTGEPEVPIIEIGERAAKYKIFTALKKFVFMSVAYKERRRQRFKSKKPKELKELAICSN


NO:
REKAMEVIQKSVVHCYGDVKQEIPRIRKIKVLKNHKGRLFYKQKRSKIKIAKLEKGSFF


118
KTFIPKVHNNGCHSCHEASLNKPILVTTALNTIGADIGLINDYSTIAPTETDISWQVYYE



FIPNGDSEAVKKRLLYFYKPKGALIKSIRDKYFKKGHENAVNTGFFKYQGKIVKGPIKF



VNNELDFARKPDLKSMKIKRAGFAIPSAKRLSKEDREINRESIKIKNKIYSLSYGRKKTL



SDKDIIKHLYRPVRQKGVKPLEYRKAPDGFLEFFYSLKRKERRLRKQKEKRQKDMSEII



DAADEFAWHRHTGSIKKTTNHINFKSEVKRGKVPIMKKRIANDSFNTRHCGKCVKQG



NAINKYYIEKQKNCFDCNSIEFKWEKAALEKKGAFKLNKRLQYIVKACFNVAKAYESF



YEDFRKGEEESLDLKFKIGTTTTLKQYPQNKARAM





SEQ
HSHNLMLTKLGKQAINFAANYDANLEIGCKNCKFLSYSPKQANPKKYPRQTDVHEDG


ID
NIACHSCMQSTKEPPVYIVPIGERKSKYEILTSLNKFTFLALKYKEKKRQAFRAKKPKE


NO:
LQELAIAFNKEKAIKVIDKSIQHLILNIKPEIARIQRQKRLKNRKGKLLYLHKRYAIKMG


119
LIKNGKYFKVGSPKKDGKKLLVLCALNTIGRDIGIIGNIEENNRSETEITYQLYFDCLDA



NPNELRIKEIEYNRLKSYERKIKRLVYAYKPKQTKILEIRSKFFSKGHENKVNTGSFNFE



NPLNKSISIKVKNSAFDFKIGAPFIMLRNGKFHIPTKKRLSKEEREINRTLSKIKGRVFRL



TYGRNISEQGSKSLHIYRKERQHPKLSLEIRKQPDSFIDEFEKLRLKQNFISKLKKQRQK



KLADLLQFADRIAYNYHTSSLEKTSNFINYKPEVKRGRTSYIKKRIGNEGFEKLYCETCI



KSNDKENAYAVEKEELCFVCKAKPFTWKKTNKDKLGIFKYPSRIKDFIRAAFTVAKSY



NDFYENLKKKDLKNEIFLKFKIGLILSHEKKNHISIAKSVAEDERISGKSIKNILNKSIKLE



KNCYSCFFHKEDM





SEQ
SLERVIDKRNLAKKAINIAANFDANINKGFYRCETNQCMFIAQKPRKTNNTGCSSCLQS


ID
TYDPVIYVVKVGEMLAKYEILKSLKRFVFMNRSFKQKKTEKAKQKERIGGELNEMSIF


NO:
ANAALAMGVIKRAIRHCHVDIRPEINRLSELKKTKHRVAAKSLVKIVKQRKTKWKGIP


120
NSFIQIPQKARNKDADFYVASALKSGGIDIGLCGTYDKKPHADPRWTYQLYFDTEDES



EKRLLYCYNDPQAKIRDFWKTFYERGNPSMVNSPGTIEFRMEGFFEKMTPISIESKDFD



FRVWNKDLLIRRGLYEIKKRKNLNRKAREIKKAMGSVKRVLANMTYGKSPTDKKSIP



VYRVEREKPKKPRAVRKEENELADKLENYRREDFLIRNRRKREATEIAKIIDAAEPPIR



HYHTNHLRAVKRIDLSKPVARKNTSVFLKRIMQNGYRGNYCKKCIKGNIDPNKDECR



LEDIKKCICCEGTQNIWAKKEKLYTGRINVLNKRIKQMKLECFNVAKAYENFYDNLA



ALKEGDLKVLKLKVSIPALNPEASDPEEDM





SEQ
NASINLGKRAINLSANYDSNLVIGCKNCKFLSFNGNFPRQTNVREGCHSCDKSTYAPE


ID
VYIVKIGERKAKYDVLDSLKKFTFQSLKYQIKKSMRERSKKPKELLEFVIFANKDKAFN


NO:
VIQKSYEHLILNIKQEINRMNGKKRIKNHKKRLFKDREKQLNKLRLIGSSSLFFPRENKG


121
DKDLFTYVAIHSVGRDIGVAGSYESHIEPISDLTYQLFINNEKRLLYAYKPKQNKIIELK



ENLWNLKKEKKPLDLEFTKPLEKSITFSVKNDKLFKVSKDLMLRQAKFNIQGKEKLSK



EERQINRDFSKIKSNVISLSYGRFEELKKEKNIWSPHIYREVKQKEIKPCIVRKGDRIELF



EQLKRKMDKLKKFRKERQKKISKDLNFAERIAYNFHTKSIKNTSNKINIDQEAKRGKA



SYMRKRIGNESFRKKYCEQCFSVGNVYHNVQNGCSCFDNPIELIKKGDEGLIPKGKED



RKYKGALRDDNLQMQIIRVAFNIAKGYEDFYNNLKEKTEKDLKLKFKIGTTISTQESN



NKEM





SEQ
SNLIKLGKQAINFAANYDANLEVGCKNCKFLSSTNKYPRQTNVHLDNKMACRSCNQS


ID
TMEPAIYIVRIGEKKAKYDIYNSLTKFNFQSLKYKAKRSQRFKPKQPKELQELSIAVRK


NO:
EKALDIIQKSIDHLIQDIRPEIPRIKQQKRYKNHVGKLFYLQKRRKNKLNLIGKGSFFKV


122
FSPKEKKNELLVICALTNIGRDIGLIGNYNTIINPLFEVTYQLYYDYIPKKNNKNVQRRL



LYAYKSKNEKILKLKEAFFKRGHENAVNLGSFSYEKPLEKSLTLKIKNDKDDFQVSPSL



RIRTGRFFVPSKRNLSRQEREINRRLVKIKSKIKNMTYGKFETARDKQSVHIFRLERQKE



KLPLQFRKDEKEFMEEFQKLKRRTNSLKKLRKSRQKKLADLLQLSEKVVYNNHTGTL



KKTSNFLNFSSSVKRGKTAYIKELLGQEGFETLYCSNCINKGQKTRYNIETKEKCFSCK



DVPFVWKKKSTDKDRKGAFLFPAKLKDVIKATFTVAKAYEDFYDNLKSIDEKKPYIKF



KIGLILAHVRHEHKARAKEEAGQKNIYNKPIKIDKNCKECFFFKEEAM





SEQ
NTTRKKFRKRTGFPQSDNIKLAYCSAIVRAANLDADIQKKHNQCNPNLCVGIKSNEQS


ID
RKYEHSDRQALLCYACNQSTGAPKVDYIQIGEIGAKYKILQMVNAYDFLSLAYNLTKL


NO:
RNGKSRGHQRMSQLDEVVIVADYEKATEVIKRSINHLLDDIRGQLSKLKKRTQNEHIT


123
EHKQSKIRRKLRKLSRLLKRRRWKWGTIPNPYLKNWVFTKKDPELVTVALLHKLGRD



IGLVNRSKRRSKQKLLPKVGFQLYYKWESPSLNNIKKSKAKKLPKRLLIPYKNVKLFD



NKQKLENAIKSLLESYQKTIKVEFDQFFQNRTEEIIAEEQQTLERGLLKQLEKKKNEFAS



QKKALKEEKKKIKEPRKAKLLMEESRSLGFLMANVSYALFNTTIEDLYKKSNVVSGCI



PQEPVVVFPADIQNKGSLAKILFAPKDGFRIKFSGQHLTIRTAKFKIRGKEIKILTKTKRE



ILKNIEKLRRVWYREQHYKLKLFGKEVSAKPRFLDKRKTSIERRDPNKLADQTDDRQA



ELRNKEYELRHKQHKMAERLDNIDTNAQNLQTLSFWVGEADKPPKLDEKDARGFGV



RTCISAWKWFMEDLLKKQEEDPLLKLKLSIM





SEQ
PKKPKFQKRTGFPQPDNLRKEYCLAIVRAANLDADFEKKCTKCEGIKTNKKGNIVKGR


ID
TYNSADKDNLLCYACNISTGAPAVDYVFVGALEAKYKILQMVKAYDFHSLAYNLAK


NO:
LWKGRGRGHQRMGGLNEVVIVSNNEKALDVIEKSLNHFHDEIRGELSRLKAKFQNEH


124
LHVHKESKLRRKLRKISRLLKRRRWKWDVIPNSYLRNFTFTKTRPDFISVALLHRVGR



DIGLVTKTKIPKPTDLLPQFGFQIYYTWDEPKLNKLKKSRLRSEPKRLLVPYKKIELYK



NKSVLEEAIRHLAEVYTEDLTICFKDFFETQKRKFVSKEKESLKRELLKELTKLKKDFS



ERKTALKRDRKEIKEPKKAKLLMEESRSLGFLAANTSYALFNLIAADLYTKSKKACST



KLPRQLSTILPLEIKEHKSTTSLAIKPEEGFKIRFSNTHLSIRTPKFKMKGADIKALTKRK



REILKNATKLEKSWYGLKHYKLKLYGKEVAAKPRFLDKRNPSIDRRDPKELMEQIENR



RNEVKDLEYEIRKGQHQMAKRLDNVDTNAQNLQTKSFWVGEADKPPELDSMEAKKL



GLRTCISAWKWFMKDLVLLQEKSPNLKLKLSLTEM





SEQ
KFSKRQEGFLIPDNIDLYKCLAIVRSANLDADVQGHKSCYGVKKNGTYRVKQNGKKG


ID
VKEKGRKYVFDLIAFKGNIEKIPHEAIEEKDQGRVIVLGKFNYKLILNIEKNHNDRASLE


NO:
IKNKIKKLVQISSLETGEFLSDLLSGKIGIDEVYGIIEPDVFSGKELVCKACQQSTYAPLV


125
EYMPVGELDAKYKILSAIKGYDFLSLAYNLSRNRANKKRGHQKLGGGELSEVVISAN



YDKALNVIKRSINHYHVEIKPEISKLKKKMQNEPLKVMKQARIRRELHQLSRKVKRLK



WKWGMIPNPELQNIIFEKKEKDFVSYALLHTLGRDIGLFKDTSMLQVPNISDYGFQIYY



SWEDPKLNSIKKIKDLPKRLLIPYKRLDFYIDTILVAKVIKNLIELYRKSYVYETFGEEY



GYAKKAEDILFDWDSINLSEGIEQKIQKIKDEFSDLLYEARESKRQNFVESFENILGLYD



KNFASDRNSYQEKIQSMIIKKQQENIEQKLKREFKEVIERGFEGMDQNKKYYKVLSPNI



KGGLLYTDTNNLGFFRSHLAFMLLSKISDDLYRKNNLVSKGGNKGILDQTPETMLTLE



FGKSNLPNISIKRKFFNIKYNSSWIGIRKPKFSIKGAVIREITKKVRDEQRLIKSLEGVWH



KSTHFKRWGKPRFNLPRHPDREKNNDDNLMESITSRREQIQLLLREKQKQQEKMAGR



LDKIDKEIQNLQTANFQIKQIDKKPALTEKSEGKQSVRNALSAWKWFMEDLIKYQKRT



PILQLKLAKM





SEQ
KFSKRQEGFVIPENIGLYKCLAIVRSANLDADVQGHVSCYGVKKNGTYVLKQNGKKSI


ID
REKGRKYASDLVAFKGDIEKIPFEVIEEKKKEQSIVLGKFNYKLVLDVMKGEKDRASL


NO:
TMKNKSKKLVQVSSLGTDEFLLTLLNEKFGIEEIYGIIEPEVFSGKKLVCKACQQSTYAP


126
LVEYMPVGELDSKYKILSAIKGYDFLSLAYNLARHRSNKKRGHQKLGGGELSEVVISA



NNAKALNVIKRSLNHYYSEIKPEISKLRKKMQNEPLKVGKQARMRRELHQLSRKVKR



LKWKWGKIPNLELQNITFKESDRDFISYALLHTLGRDIGMFNKTEIKMPSNILGYGFQI



YYDWEEPKLNTIKKSKNTPKRILIPYKKLDFYNDSILVARAIKELVGLFQESYEWEIFGN



EYNYAKEAEVELIKLDEESINGNVEKKLQRIKENFSNLLEKAREKKRQNFIESFESIARL



YDESFTADRNEYQREIQSFIIEKQKQSIEKKLKNEFKKIVEKKFNEQEQGKKHYRVLNP



TIINEFLPKDKNNLGFLRSKIAFILLSKISDDLYKKSNAVSKGGEKGIIKQQPETILDLEFS



KSKLPSINIKKKLFNIKYTSSWLGIRKPKFNIKGAKIREITRRVRDVQRTLKSAESSWYA



STHFRRWGFPRFNQPRHPDKEKKSDDRLIESITLLREQIQILLREKQKGQKEMAGRLDD



VDKKIQNLQTANFQIKQTGDKPALTEKSAGKQSFRNALSAWKWFMENLLKYQNKTP



DLKLKIARTVM





SEQ
KWIEPNNIDFNKCLAITRSANLDADVQGHKMCYGIKTNGTYKAIGKINKKHNTGIIEKR


ID
RTYVYDLIVTKEKNEKIVKKTDFMAIDEEIEFDEKKEKLLKKYIKAEVLGTGELIRKDL


NO:
NDGEKFDDLCSIEEPQAFRRSELVCKACNQSTYASDIRYIPIGEIEAKYKILKAIKGYDFL


127
SLKYNLGRLRDSKKRGHQKMGQGELKEFVICANKEKALDVIKRSLNHYLNEVKDEIS



RLNKKMQNEPLKVNDQARWRRELNQISRRLKRLKWKWGEIPNPELKNLIFKSSRPEFV



SYALIHTLGRDIGLINETELKPNNIQEYGFQIYYKWEDPELNHIKKVKNIPKRFIIPYKNL



DLFGKYTILSRAIEGILKLYSSSFQYKSFKDPNLFAKEGEKKITNEDFELGYDEKIKKIKD



DFKSYKKALLEKKKNTLEDSLNSILSVYEQSLLTEQINNVKKWKEGLLKSKESIHKQK



KIENIEDIISRIEELKNVEGWIRTKERDIVNKEETNLKREIKKELKDSYYEEVRKDFSDLK



KGEESEKKPFREEPKPIVIKDYIKFDVLPGENSALGFFLSHLSFNLFDSIQYELFEKSRLSS



SKHPQIPETILDL





SEQ
FRKFVKRSGAPQPDNLNKYKCIAIVRAANLDADIMSNESSNCVMCKGIKMNKRKTAK


ID
GAAKTTELGRVYAGQSGNLLCTACTKSTMGPLVDYVPIGRIRAKYTILRAVKEYDFLS


NO:
LAYNLARTRVSKKGGRQKMHSLSELVIAAEYEIAWNIIKSSVIHYHQETKEEISGLRKK


128
LQAEHIHKNKEARIRREMHQISRRIKRLKWKWHMIPNSELHNFLFKQQDPSFVAVALL



HTLGRDIGMINKPKGSAKREFIPEYGFQIYYKWMNPKLNDINKQKYRKMPKRSLIPYK



NLNVFGDRELIENAMHKLLKLYDENLEVKGSKFFKTRVVAISSKESEKLKRDLLWKGE



LAKIKKDFNADKNKMQELFKEVKEPKKANALMKQSRNMGFLLQNISYGALGLLANR



MYEASAKQSKGDATKQPSIVIPLEMEFGNAFPKLLLRSGKFAMNVSSPWLTIRKPKFVI



KGNKIKNITKLMKDEKAKLKRLETSYHRATHFRPTLRGSIDWDSPYFSSPKQPNTHRRS



PDRLSADITEYRGRLKSVEAELREGQRAMAKKLDSVDMTASNLQTSNFQLEKGEDPR



LTEIDEKGRSIRNCISSWKKFMEDLMKAQEANPVIKIKIALKDESSVLSEDSM





SEQ
KFHPENLNKSYCLAIVRAANLDADIQGHINCIGIKSNKSDRNYENKLESLQNVELLCKA


ID
CTKSTYKPNINSVPVGEKKAKYSILSEIKKYDFNSLVYNLKKYRKGKSRGHQKLNELR


NO:
ELVITSEYKKALDVINKSVNHYLVNIKNKMSKLKKILQNEHIHVGTLARIRRERNRISR


129
KLDHYRKKWKFVPNKILKNYVFKNQSPDFVSVALLHKLGRDIGLITKTAILQKSFPEYS



LQLYYKYDTPKLNYLKKSKFKSLPKRILISYKYPKFDINSNYIEESIDKLLKLYEESPIYK



NNSKIIEFFKKSEDNLIKSENDSLKRGIMKEFEKVTKNFSSKKKKLKEELKLKNEDKNS



KMLAKVSRPIGFLKAYLSYMLFNIISNRIFEFSRKSSGRIPQLPSCIINLGNQFENFKNEL



QDSNIGSKKNYKYFCNLLLKSSGFNISYEEEHLSIKTPNFFINGRKLKEITSEKKKIRKEN



EQLIKQWKKLTFFKPSNLNGKKTSDKIRFKSPNNPDIERKSEDNIVENIAKVKYKLEDL



LSEQRKEFNKLAKKHDGVDVEAQCLQTKSFWIDSNSPIKKSLEKKNEKVSVKKKMKA



IRSCISAWKWFMADLIEAQKETPMIKLKLALM





SEQ
TTLVPSHLAGIEVMDETTSRNEDMIQKETSRSNEDENYLGVKNKCGINVHKSGRGSSK


ID
HEPNMPPEKSGEGQMPKQDSTEMQQRFDESVTGETQVSAGATASIKTDARANSGPRV


NO:
GTARALIVKASNLDRDIKLGCKPCEYIRSELPMGKKNGCNHCEKSSDIASVPKVESGFR


130
KAKYELVRRFESFAADSISRHLGKEQARTRGKRGKKDKKEQMGKVNLDEIAILKNESL



IEYTENQILDARSNRIKEWLRSLRLRLRTRNKGLKKSKSIRRQLITLRRDYRKWIKPNPY



RPDEDPNENSLRLHTKLGVDIGVQGGDNKRMNSDDYETSFSITWRDTATRKICFTKPK



GLLPRHMKFKLRGYPELILYNEELRIQDSQKFPLVDWERIPIFKLRGVSLGKKKVKALN



RITEAPRLVVAKRIQVNIESKKKKVLTRYVYNDKSINGRLVKAEDSNKDPLLEFKKQA



EEINSDAKYYENQEIAKNYLWGCEGLHKNLLEEQTKNPYLAFKYGFLNIV





SEQ
LDFKRTCSQELVLLPEIEGLKLSGTQGVTSLAKKLINKAANVDRDESYGCHHCIHTRTS


ID
LSKPVKKDCNSCNQSTNHPAVPITLKGYKIAFYELWHRFTSWAVDSISKALHRNKVM


NO:
GKVNLDEYAVVDNSHIVCYAVRKCYEKRQRSVRLHKRAYRCRAKHYNKSQPKVGRI


131
YKKSKRRNARNLKKEAKRYFQPNEITNGSSDALFYKIGVDLGIAKGTPETEVKVDVSI



CFQVYYGDARRVLRVRKMDELQSFHLDYTGKLKLKGIGNKDTFTIAKRNESLKWGST



KYEVSRAHKKFKPFGKKGSVKRKCNDYFRSIASWSCEAASQRAQSNLKNAFPYQKAL



VKCYKNLDYKGVKKNDMWYRLCSNRIFRYSRIAEDIAQYQSDKGKAKFEFVILAQSV



AEYDISAIM





SEQ
VFLTDDKRKTALRKIRSAFRKTAEIALVRAQEADSLDRQAKKLTIETVSFGAPGAKNA


ID
FIGSLQGYNWNSHRANVPSSGSAKDVFRITELGLGIPQSAHEASIGKSFELVGNVVRYT


NO:
ANLLSKGYKKGAVNKGAKQQREIKGKEQLSFDLISNGPISGDKLINGQKDALAWWLI


132
DKMGFHIGLAMEPLSSPNTYGITLQAFWKRHTAPRRYSRGVIRQWQLPFGRQLAPLIH



NFFRKKGASIPIVLTNASKKLAGKGVLLEQTALVDPKKWWQVKEQVTGPLSNIWERS



VPLVLYTATFTHKHGAAHKRPLTLKVIRISSGSVFLLPLSKVTPGKLVRAWMPDINILR



DGRPDEAAYKGPDLIRARERSFPLAYTCVTQIADEWQKRALESNRDSITPLEAKLVTGS



DLLQIHSTVQQAVEQGIGGRISSPIQELLAKDALQLVLQQLFMTVDLLRIQWQLKQEV



ADGNTSEKAVGWAIRISNIHKDAYKTAIEPCTSALKQAWNPLSGFEERTFQLDASIVRK



RSTAKTPDDELVIVLRQQAAEMTVAVTQSVSKELMELAVRHSATLHLLVGEVASKQL



SRSADKDRGAMDHWKLLSQSM





SEQ
EDLLQKALNTATNVAAIERHSCISCLFTESEIDVKYKTPDKIGQNTAGCQSCTFRVGYS


ID
GNSHTLPMGNRIALDKLRETIQRYAWHSLLFNVPPAPTSKRVRAISELRVAAGRERLFT


NO:
VITFVQTNILSKLQKRYAANWTPKSQERLSRLREEGQHILSLLESGSWQQKEVVREDQ


133
DLIVCSALTKPGLSIGAFCRPKYLKPAKHALVLRLIFVEQWPGQIWGQSKRTRRMRRR



KDVERVYDISVQAWALKGKETRISECIDTMRRHQQAYIGVLPFLILSGSTVRGKGDCPI



LKEITRMRYCPNNEGLIPLGIFYRGSANKLLRVVKGSSFTLPMWQNIETLPHPEPFSPEG



WTATGALYEKNLAYWSALNEAVDWYTGQILSSGLQYPNQNEFLARLQNVIDSIPRKW



FRPQGLKNLKPNGQEDIVPNEFVIPQNAIRAHHVIEWYHKTNDLVAKTLLGWGSQTTL



NQTRPQGDLRFTYTRYYFREKEVPEV





SEQ
VPKKKLMRELAKKAVFEAIFNDPIPGSFGCKRCTLIDGARVTDAIEKKQGAKRCAGCE


ID
PCTFHTLYDSVKHALPAATGCDRTAIDTGLWEILTALRSYNWMSFRRNAVSDASQKQ


NO:
VWSIEELAIWADKERALRVILSALTHTIGKLKNGFSRDGVWKGGKQLYENLAQKDLA


134
KGLFANGEIFGKELVEADHDMLAWTIVPNHQFHIGLIRGNWKPAAVEASTAFDARWL



TNGAPLRDTRTHGHRGRRFNRTEKLTVLCIKRDGGVSEEFRQERDYELSVMLLQPKN



KLKPEPKGELNSFEDLHDHWWFLKGDEATALVGLTSDPTVGDFIQLGLYIRNPIKAHG



ETKRRLLICFEPPIKLPLRRAFPSEAFKTWEPTINVFRNGRRDTEAYYDIDRARVFEFPET



RVSLEHLSKQWEVLRLEPDRENTDPYEAQQNEGAELQVYSLLQEAAQKMAPKVVIDP



FGQFPLELFSTFVAQLFNAPLSDTKAKIGKPLDSGFVVESHLHLLEEDFAYRDFVRVTF



MGTEPTFRVIHYSNGEGYWKKTVLKGKNNIRTALIPEGAKAAVDAYKNKRCPLTLEA



AILNEEKDRRLVLGNKALSLLAQTARGNLTILEALAAEVLRPLSGTEGVVHLHACVTR



HSTLTESTETDNM





SEQ
VEKLFSERLKRAMWLKNEAGRAPPAETLTLKHKRVSGGHEKVKEELQRVLRSLSGTN


ID
QAAWNLGLSGGREPKSSDALKGEKSRVVLETVVFHSGHNRVLYDVIEREDQVHQRSS


NO:
IMHMRRKGSNLLRLWGRSGKVRRKMREEVAEIKPVWHKDSRWLAIVEEGRQSVVGIS


135
SAGLAVFAVQESQCTTAEPKPLEYVVSIWFRGSKALNPQDRYLEFKKLKTTEALRGQQ



YDPIPFSLKRGAGCSLAIRGEGIKFGSRGPIKQFFGSDRSRPSHADYDGKRRLSLFSKYA



GDLADLTEEQWNRTVSAFAEDEVRRATLANIQDFLSISHEKYAERLKKRIESIEEPVSA



SKLEAYLSAIFETFVQQREALASNFLMRLVESVALLISLEEKSPRVEFRVARYLAESKE



GFNRKAM





SEQ
VVITQSELYKERLLRVMEIKNDRGRKEPRESQGLVLRFTQVTGGQEKVKQKLWLIFEG


ID
FSGTNQASWNFGQPAGGRKPNSGDALKGPKSRVTYETVVFHFGLRLLSAVIERHNLK


NO:
QQRQTMAYMKRRAAARKKWARSGKKCSRMRNEVEKIKPKWHKDPRWFDIVKEGEP


136
SIVGISSAGFAIYIVEEPNFPRQDPLEIEYAISIWFRRDRSQYLTFKKIQKAEKLKELQYNP



IPFRLKQEKTSLVFESGDIKFGSRGSIEHFRDEARGKPPKADMDNNRRLTMFSVFSGNL



TNLTEEQYARPVSGLLAPDEKRMPTLLKKLQDFFTPIHEKYGERIKQRLANSEASKRPF



KKLEEYLPAIYLEFRARREGLASNWVLVLINSVRTLVRIKSEDPYIEFKVSQYLLEKED



NKAL





SEQ
KQDALFEERLKKAIFIKRQADPLQREELSLLPPNRKIVTGGHESAKDTLKQILRAINGTN


ID
QASWNPGTPSGKRDSKSADALAGPKSRVKLETVVFHVGHRLLKKVVEYQGHQKQQH


NO:
GLKAFMRTCAAMRKKWKRSGKVVGELREQLANIQPKWHYDSRPLNLCFEGKPSVVG


137
LRSAGIALYTIQKSVVPVKEPKPIEYAVSIWFRGPKAMDREDRCLEFKKLKIATELRKL



QFEPIVSTLTQGIKGFSLYIQGNSVKFGSRGPIKYFSNESVRQRPPKADPDGNKRLALFS



KFSGDLSDLTEEQWNRPILAFEGIIRRATLGNIQDYLTVGHEQFAISLEQLLSEKESVLQ



MSIEQQRLKKNLGKKAENEWVESFGAEQARKKAQGIREYISGFFQEYCSQREQWAEN



WVQQLNKSVRLFLTIQDSTPFIEFRVARYLPKGEKKKGKAM





SEQ
ANHAERHKRLRKEANRAANRNRPLVADCDTGDPLVGICRLLRRGDKMQPNKTGCRS


ID
CEQVEPELRDAILVSGPGRLDNYKYELFQRGRAMAVHRLLKRVPKLNRPKKAAGNDE


NO:
KKAENKKSEIQKEKQKQRRMMPAVSMKQVSVADFKHVIENTVRHLFGDRRDREIAEC


138
AALRAASKYFLKSRRVRPRKLPKLANPDHGKELKGLRLREKRAKLKKEKEKQAELAR



SNQKGAVLHVATLKKDAPPMPYEKTQGRNDYTTFVISAAIKVGATRGTKPLLTPQPRE



WQCSLYWRDGQRWIRGGLLGLQAGIVLGPKLNRELLEAVLQRPIECRMSGCGNPLQV



RGAAVDFFMTTNPFYVSGAAYAQKKFKPFGTKRASEDGAAAKAREKLMTQLAKVLD



KVVTQAAHSPLDGIWETRPEAKLRAMIMALEHEWIFLRPGPCHNAAEEVIKCDCTGG



HAILWALIDEARGALEHKEFYAVTRAHTHDCEKQKLGGRLAGFLDLLIAQDVPLDDA



PAARKIKTLLEATPPAPCYKAATSIATCDCEGKFDKLWAIIDATRAGHGTEDLWARTL



AYPQNVNCKCKAGKDLTHRLADFLGLLIKRDGPFRERPPHKVTGDRKLVFSGDKKCK



GHQYVILAKAHNEEVVRAWISRWGLKSRTNKAGYAATELNLLLNWLSICRRRWMDM



LTVQRDTPYIRMKTGRLVVDDKKERKAM





SEQ
AKQREALRVALERGIVRASNRTYTLVTNCTKGGPLPEQCRMIERGKARAMKWEPKLV


ID
GCGSCAAATVDLPAIEEYAQPGRLDVAKYKLTTQILAMATRRMMVRAAKLSRRKGQ


NO:
WPAKVQEEKEEPPEPKKMLKAVEMRPVAIVDFNRVIQTTIEHLWAERANADEAELKA


139
LKAAAAYFGPSLKIRARGPPKAAIGRELKKAHRKKAYAERKKARRKRAELARSQARG



AAAHAAIRERDIPPMAYERTQGRNDVTTIPIAAAIKIAATRGARPLPAPKPMKWQCSLY



WNEGQRWIRGGMLTAQAYAHAANIHRPMRCEMWGVGNPLKVRAFEGRVADPDGA



KGRKAEFRLQTNAFYVSGAAYRNKKFKPFGTDRGGIGSARKKRERLMAQLAKILDKV



VSQAAHSPLDDIWHTRPAQKLRAMIKQLEHEWMFLRPQAPTVEGTKPDVDVAGNMQ



RQIKALMAPDLPPIEKGSPAKRFTGDKRKKGERAVRVAEAHSDEVVTAWISRWGIQTR



RNEGSYAAQELELLLNWLQICRRRWLDMTAAQRVSPYIRMKSGRMITDAADEGVAPI



PLVENM





SEQ
KSISGRSIKHMACLKDMLKSEITEIEEKQKKESLRKWDYYSKFSDEILFRRNLNVSANH


ID
DANACYGCNPCAFLKEVYGFRIERRNNERIISYRRGLAGCKSCVQSTGYPPIEFVRRKF


NO:
GADKAMEIVREVLHRRNWGALARNIGREKEADPILGELNELLLVDARPYFGNKSAAN


140
ETNLAFNVITRAAKKFRDEGMYDIHKQLDIHSEEGKVPKGRKSRLIRIERKHKAIHGLD



PGETWRYPHCGKGEKYGVWLNRSRLIHIKGNEYRCLTAFGTTGRRMSLDVACSVLGH



PLVKKKRKKGKKTVDGTELWQIKKATETLPEDPIDCTFYLYAAKPTKDPFILKVGSLK



APRWKKLHKDFFEYSDTEKTQGQEKGKRVVRRGKVPRILSLRPDAKFKVSIWDDPYN



GKNKEGTLLRMELSGLDGAKKPLILKRYGEPNTKPKNFVFWRPHITPHPLTFTPKHDF



GDPNKKTKRRRVFNREYYGHLNDLAKMEPNAKFFEDREVSNKKNPKAKNIRIQAKES



LPNIVAKNGRWAAFDPNDSLWKLYLHWRGRRKTIKGGISQEFQEFKERLDLYKKHED



ESEWKEKEKLWENHEKEWKKTLEIHGSIAEVSQRCVMQSMMGPLDGLVQKKDYVHI



GQSSLKAADDAWTFSANRYKKATGPKWGKISVSNLLYDANQANAELISQSISKYLSK



QKDNQGCEGRKMKFLIKIIEPLRENFVKHTRWLHEMTQKDCEVRAQFSRVSM





SEQ
FPSDVGADALKHVRMLQPRLTDEVRKVALTRAPSDRPALARFAAVAQDGLAFVRHL


ID
NVSANHDSNCTFPRDPRDPRRGPCEPNPCAFLREVWGFRIVARGNERALSYRRGLAGC


NO:
KSCVQSTGFPSVPFHRIGADDCMRKLHEILKARNWRLLARNIGREREADPLLTELSEYL


141
LVDARTYPDGAAPNSGRLAENVIKRAAKKFRDEGMRDIHAQLRVHSREGKVPKGRLQ



RLRRIERKHRAIHALDPGPSWEAEGSARAEVQGVAVYRSQLLRVGHHTQQIEPVGIVA



RTLFGVGRTDLDVAVSVLGAPLTKRKKGSKTLESTEDFRIAKARETRAEDKIEVAFVL



YPTASLLRDEIPKDAFPAMRIDRFLLKVGSVQADREILLQDDYYRFGDAEVKAGKNKG



RTVTRPVKVPRLQALRPDAKFRVNVWADPFGAGDSPGTLLRLEVSGVTRRSQPLRLLR



YGQPSTQPANFLCWRPHRVPDPMTFTPRQKFGERRKNRRTRRPRVFERLYQVHIKHLA



HLEPNRKWFEEARVSAQKWAKARAIRRKGAEDIPVVAPPAKRRWAALQPNAELWDL



YAHDREARKRFRGGRAAEGEEFKPRLNLYLAHEPEAEWESKRDRWERYEKKWTAVL



EEHSRMCAVADRTLPQFLSDPLGARMDDKDYAFVGKSALAVAEAFVEEGTVERAQG



NCSITAKKKFASNASRKRLSVANLLDVSDKADRALVFQAVRQYVQRQAENGGVEGR



RMAFLRKLLAPLRQNFVCHTRWLHM





SEQ
AARKKKRGKIGITVKAKEKSPPAAGPFMARKLVNVAANVDGVEVHLCVECEADAHG


ID
SASARLLGGCRSCTGSIGAEGRLMGSVDVDRERVIAEPVHTETERLGPDVKAFEAGTA


NO:
ESKYAIQRGLEYWGVDLISRNRARTVRKMEEADRPESSTMEKTSWDEIAIKTYSQAYH


142
ASENHLFWERQRRVRQHALALFRRARERNRGESPLQSTQRPAPLVLAALHAEAAAISG



RARAEYVLRGPSANVRAAAADIDAKPLGHYKTPSPKVARGFPVKRDLLRARHRIVGL



SRAYFKPSDVVRGTSDAIAHVAGRNIGVAGGKPKEIEKTFTLPFVAYWEDVDRVVHCS



SFKADGPWVRDQRIKIRGVSSAVGTFSLYGLDVAWSKPTSFYIRCSDIRKKFHPKGFGP



MKHWRQWAKELDRLTEQRASCVVRALQDDEELLQTMERGQRYYDVFSCAATHATR



GEADPSGGCSRCELVSCGVAHKVTKKAKGDTGIEAVAVAGCSLCESKLVGPSKPRVH



RQMAALRQSHALNYLRRLQREWEALEAVQAPTPYLRFKYARHLEVRSM





SEQ
AAKKKKQRGKIGISVKPKEGSAPPADGPFMARKLVNVAANVDGVEVNLCIECEADAH


ID
GSAPARLLGGCKSCTGSIGAEGRLMGSVDVDRADAIAKPVNTETEKLGPDVQAFEAG


NO:
TAETKYALQRGLEYWGVDLISRNRSRTVRRTEEGQPESATMEKTSWDEIAIKSYTRAY


143
HASENHLFWERQRRVRQHALALFKRAKERNRGDSTLPREPGHGLVAIAALACEAYAV



GGRNLAETVVRGPTFGTARAVRDVEIASLGRYKTPSPKVAHGSPVKRDFLRARHRIVG



LARAYYRPSDVVRGTSDAIAHVAGRNIGVAGGKPRAVEAVFTLPFVAYWEDVDRVV



HCSSFQVSAPWNRDQRMKIAGVTTAAGTFSLHGGELKWAKPTSFYIRCSDTRRKFRPK



GFGPMKRWRQWAKDLDRLVEQRASCVVRALQDDAALLETMERGQRYYDVFACAVT



HATRGEADRLAGCSRCALTPCQEAHRVTTKPRGDAGVEQVQTSDCSLCEGKLVGPSK



PRLHRTLTLLRQEHGLNYLRRLQREWESLEAVQVPTPYLRFKYARHLEVRSM





SEQ
TDSQSESVPEVVYALTGGEVPGRVPPDGGSAEGARNAPTGLRKQRGKIKISAKPSKPGS


ID
PASSLARTLVNEAANVDGVQSSGCATCRMRANGSAPRALPIGCVACASSIGRAPQEET


NO:
VCALPTTQGPDVRLLEGGHALRKYDIQRALEYWGVDLIGRNLDRQAGRGMEPAEGA


144
TATMKRVSMDELAVLDFGKSYYASEQHLFAARQRRVRQHAKALKIRAKHANRSGSV



KRALDRSRKQVTALAREFFKPSDVVRGDSDALAHVVGRNLGVSRHPAREIPQTFTLPL



CAYWEDVDRVISCSSLLAGEPFARDQEIRIEGVSSALGSLRLYRGAIEWHKPTSLYIRCS



DTRRKFRPRGGLKKRWRQWAKDLDRLVEQRACCIVRSLQADVELLQTMERAQRFYD



VHDCAATHVGPVAVRCSPCAGKQFDWDRYRLLAALRQEHALNYLRRLQREWESLEA



QQVKMPYLRFKYARKLEVSGPLIGLEVRREPSMGTAIAEM





SEQ
AGTAGRRHGSLGARRSINIAGVTDRHGRWGCESCVYTRDQAGNRARCAPCDQSTYAP


ID
DVQEVTIGQRQAKYTIFLTLQSFSWTNTMRNNKRAAAGRSKRTTGKRIGQLAEIKITG


NO:
VGLAHAHNVIQRSLQHNITKMWRAEKGKSKRVARLKKAKQLTKRRAYFRRRMSRQS


145
RGNGFFRTGKGGIHAVAPVKIGLDVGMIASGSSEPADEQTVTLDAIWKGRKKKIRLIG



AKGELAVAACRFREQQTKGDKCIPLILQDGEVRWNQNNWQCHPKKLVPLCGLEVSR



KFVSQADRLAQNKVASPLAARFDKTSVKGTLVESDFAAVLVNVTSIYQQCHAMLLRS



QEPTPSLRVQRTITSM





SEQ
GVRFSPAQSQVFFRTVIPQSVEARFAINMAAIHDAAGAFGCSVCRFEDRTPRNAKAVH


ID
GCSPCTRSTNRPDVFVLPVGAIKAKYDVFMRLLGFNWTHLNRRQAKRVTVRDRIGQL


NO:
DELAISMLTGKAKAVLKKSICHNVDKSFKAMRGSLKKLHRKASKTGKSQLRAKLSDL


146
RERTNTTQEGSHVEGDSDVALNKIGLDVGLVGKPDYPSEESVEVVVCLYFVGKVLILD



AQGRIRDMRAKQYDGFKIPIIQRGQLTVLSVKDLGKWSLVRQDYVLAGDLRFEPKISK



DRKYAECVKRIALITLQASLGFKERIPYYVTKQVEIKNASHIAFVTEAIQNCAENFREM



TEYLMKYQEKSPDLKVLLTQLM





SEQ
RAVVGKVFLEQARRALNLATNFGTNHRTGCNGCYVTPGKLSIPQDGEKNAAGCTSCL


ID
MKATASYVSYPKPLGEKVAKYSTLDALKGFPWYSLRLNLRPNYRGKPINGVQEVAPV


NO:
SKFRLAEEVIQAVQRYHFTELEQSFPGGRRRLRELRAFYTKEYRRAPEQRQHVVNGDR


147
NIVVVTVLHELGFSVGMFNEVELLPKTPIECAVNVFIRGNRVLLEVRKPQFDKERLLVE



SLWKKDSRRHTAKWTPPNNEGRIFTAEGWKDFQLPLLLGSTSRSLRAIEKEGFVQLAP



GRDPDYNNTIDEQHSGRPFLPLYLYLQGTISQEYCVFAGTWVIPFQDGISPYSTKDTFQP



DLKRKAYSLLLDAVKHRLGNKVASGLQYGRFPAIEELKRLVRMHGATRKIPRGEKDL



LKKGDPDTPEWWLLEQYPEFWRLCDAAAKRVSQNVGLLLSLKKQPLWQRRWLESRT



RNEPLDNLPLSMALTLHLTNEEAL





SEQ
AAVYSKFYIENHFKMGIPETLSRIRGPSIIQGFSVNENYINIAGVGDRDFIFGCKKCKYTR


ID
GKPSSKKINKCHPCKRSTYPEPVIDVRGSISEFKYKIYNKLKQEPNQSIKQNTKGRMNPS


NO:
DHTSSNDGIIINGIDNRIAYNVIFSSYKHLMEKQINLLRDTTKRKARQIKKYNNSGKKK


148
HSLRSQTKGNLKNRYHMLGMFKKGSLTITNEGDFITAVRKVGLDISLYKNESLNKQEV



ETELCLNIKWGRTKSYTVSGYIPLPINIDWKLYLFEKETGLTLRLFGNKYKIQSKKFLIA



QLFKPKRPPCADPVVKKAQKWSALNAHVQQMAGLFSDSHLLKRELKNRMHKQLDFK



SLWVGTEDYIKWFEELSRSYVEGAEKSLEFFRQDYFCFNYTKQTTM





SEQ
PQQQRDLMLMAANYDQDYGNGCGPCTVVASAAYRPDPQAQHGCKRHLRTLGASAV


ID
THVGLGDRTATITALHRLRGPAALAARARAAQAASAPMTPDTDAPDDRRRLEAIDAD


NO:
DVVLVGAHRALWSAVRRWADDRRAALRRRLHSEREWLLKDQIRWAELYTLIEASGT


149
PPQGRWRNTLGALRGQSRWRRVLAPTMRATCAETHAELWDALAELVPEMAKDRRG



LLRPPVEADALWRAPMIVEGWRGGHSVVVDAVAPPLDLPQPCAWTAVRLSGDPRQR



WGLHLAVPPLGQVQPPDPLKATLAVSMRHRGGVRVRTLQAMAVDADAPMQRHLQV



PLTLQRGGGLQWGIHSRGVRRREARSMASWEGPPIWTGLQLVNRWKGQGSALLAPD



RPPDTPPYAPDAAVAPAQPDTKRARRTLKEACTVCRCAPGHMRQLQVTLTGDGTWR



RFRLRAPQGAKRKAEVLKVATQHDERIANYTAWYLKRPEHAAGCDTCDGDSRLDGA



CRGCRPLLVGDQCFRRYLDKIEADRDDGLAQIKPKAQEAVAAMAAKRDARAQKVAA



RAAKLSEATGQRTAATRDASHEARAQKELEAVATEGTTVRHDAAAVSAFGSWVARK



GDEYRHQVGVLANRLEHGLRLQELMAPDSVVADQQRASGHARVGYRYVLTAM





SEQ
AVAHPVGRGNAGSPGARGPEELPRQLVNRASNVTRPATYGCAPCRHVRLSIPKPVLTG


ID
CRACEQTTHPAPKRAVRGGADAAKYDLAAFFAGWAADLEGRNRRRQVHAPLDPQPD


NO:
PNHEPAVTLQKIDLAEVSIEEFQRVLARSVKHRHDGRASREREKARAYAQVAKKRRN


150
SHAHGARTRRAVRRQTRAVRRAHRMGANSGEILVASGAEDPVPEAIDHAAQLRRRIR



ACARDLEGLRHLSRRYLKTLEKPCRRPRAPDLGRARCHALVESLQAAERELEELRRCD



SPDTAMRRLDAVLAAAASTDATFATGWTVVGMDLGVAPRGSAAPEVSPMEMAISVF



WRKGSRRVIVSKPIAGMPIRRHELIRLEGLGTLRLDGNHYTGAGVTKGRGLSEGTEPDF



REKSPSTLGFTLSDYRHESRWRPYGAKQGKTARQFFAAMSRELRALVEHQVLAPMGP



PLLEAHERRFETLLKGQDNKSIHAGGGGRYVWRGPPDSKKRPAADGDWFRFGRGHA



DHRGWANKRHELAANYLQSAFRLWSTLAEAQEPTPYARYKYTRVTM





SEQ
WDFLTLQVYERHTSPEVCVAGNSTKCASGTRKSDHTHGVGVKLGAQEINVSANDDR


ID
DHEVGCNICVISRVSLDIKGWRYGCESCVQSTPEWRSIVRFDRNHKEAKGECLSRFEY


NO:
WGAQSIARSLKRNKLMGGVNLDELAIVQNENVVKTSLKHLFDKRKDRIQANLKAVK


151
VRMRERRKSGRQRKALRRQCRKLKRYLRSYDPSDIKEGNSCSAFTKLGLDIGISPNKPP



KIEPKVEVVFSLFYQGACDKIVTVSSPESPLPRSWKIKIDGIRALYVKSTKVKFGGRTFR



AGQRNNRRKVRPPNVKKGKRKGSRSQFFNKFAVGLDAVSQQLPIASVQGLWGRAET



KKAQTICLKQLESNKPLKESQRCLFLADNWVVRVCGFLRALSQRQGPTPYIRYRYRCN



M





SEQ
ARNVGQRNASRQSKRESAKARSRRVTGGHASVTQGVALINAAANADRDHTTGCEPC


ID
TWERVNLPLQEVIHGCDSCTKSSPFWRDIKVVNKGYREAKEEIMRIASGISADHLSRAL


NO:
SHNKVMGRLNLDEVCILDFRTVLDTSLKHLTDSRSNGIKEHIRAVHRKIRMRRKSGKT


152
ARALRKQYFALRRQWKAGHKPNSIREGNSLTALRAVGFDVGVSEGTEPMPAPQTEVV



LSVFYKGSATRILRISSPHPIAKRSWKVKIAGIKALKLIRREHDFSFGRETYNASQRAEK



RKFSPHAARKDFFNSFAVQLDRLAQQLCVSSVENLWVTEPQQKLLTLAKDTAPYGIRE



GARFADTRARLAWNWVFRVCGFTRALHQEQEPTPYCRFTWRSKM









In some embodiments, the Type V CRISPR/Cas enzyme is a CasΦ nuclease. A CasΦ polypeptide can function as an endonuclease that catalyzes cleavage at a specific sequence in a target nucleic acid. A programmable CasΦ nuclease of the present disclosure may have a single active site in a RuvC domain that is capable of catalyzing pre-crRNA processing and nicking or cleaving of nucleic acids. This compact catalytic site may render the programmable CasΦ nuclease especially advantageous for genome engineering and new functionalities for genome manipulation.


TABLE 3 provides amino acid sequences of illustrative CasΦ polypeptides that can be used in compositions and methods of the disclosure.









TABLE 3







CasΦ Amino Acid Sequences










SEQ ID



Name
NO
Amino Acid Sequence





CasΦ.1
SEQ ID
MADTPTLFTQFLRHHLPGQRFRKDILKQAGRILANKGEDATIA



NO: 221
FLRGKSEESPPDFQPPVKCPIIACSRPLTEWPIYQASVAIQGYV




YGQSLAEFEASDPGCSKDGLLGWFDKTGVCTDYFSVQGLNLI




FQNARKRYIGVQTKVTNRNEKRHKKLKRINAKRIAEGLPELT




SDEPESALDETGHLIDPPGLNTNIYCYQQVSPKPLALSEVNQLP




TAYAGYSTSGDDPIQPMVTKDRLSISKGQPGYIPEHQRALLSQ




KKHRRMRGYGLKARALLVIVRIQDDWAVIDLRSLLRNAYWR




RIVQTKEPSTITKLLKLVTGDPVLDATRMVATFTYKPGIVQVR




SAKCLKNKQGSKLFSERYLNETVSVTSIDLGSNNLVAVATYR




LVNGNTPELLQRFTLPSHLVKDFERYKQAHDTLEDSIQKTAV




ASLPQGQQTEIRMWSMYGFREAQERVCQELGLADGSIPWNV




MTATSTILTDLFLARGGDPKKCMFTSEPKKKKNSKQVLYKIR




DRAWAKMYRTLLSKETREAWNKALWGLKRGSPDYARLSKR




KEELARRCVNYTISTAEKRAQCGRTIVALEDLNIGFFHGRGKQ




EPGWVGLFTRKKENRWLMQALHKAFLELAHHRGYHVIEVNP




AYTSQTCPVCRHCDPDNRDQHNREAFHCIGCGFRGNADLDV




ATHNIAMVAITGESLKRARGSVASKTPQPLAAE





CasΦ.2
SEQ ID
MPKPAVESEFSKVLKKHFPGERFRSSYMKRGGKILAAQGEEA



NO: 222
VVAYLQGKSEEEPPNFQPPAKCHVVTKSRDFAEWPIMKASEA




IQRYIYALSTTERAACKPGKSSESHAAWFAATGVSNHGYSHV




QGLNLIFDHTLGRYDGVLKKVQLRNEKARARLESINASRADE




GLPEIKAEEEEVATNETGHLLQPPGINPSFYVYQTISPQAYRPR




DEIVLPPEYAGYVRDPNAPIPLGVVRNRCDIQKGCPGYIPEWQ




REAGTAISPKTGKAVTVPGLSPKKNKRMRRYWRSEKEKAQD




ALLVTVRIGTDWVVIDVRGLLRNARWRTIAPKDISLNALLDLF




TGDPVIDVRRNIVTFTYTLDACGTYARKWTLKGKQTKATLD




KLTATQTVALVAIDLGQTNPISAGISRVTQENGALQCEPLDRF




TLPDDLLKDISAYRIAWDRNEEELRARSVEALPEAQQAEVRA




LDGVSKETARTQLCADFGLDPKRLPWDKMSSNTTFISEALLS




NSVSRDQVFFTPAPKKGAKKKAPVEVMRKDRTWARAYKPRL




SVEAQKLKNEALWALKRTSPEYLKLSRRKEELCRRSINYVIEK




TRRRTQCQIVIPVIEDLNVRFFHGSGKRLPGWDNFFTAKKENR




WFIQGLHKAFSDLRTHRSFYVFEVRPERTSITCPKCGHCEVGN




RDGEAFQCLSCGKTCNADLDVATHNLTQVALTGKTMPKREE




PRDAQGTAPARKTKKASKSKAPPAEREDQTPAQEPSQTS





CasΦ.3
SEQ ID
MYILEMADLKSEPSLLAKLLRDRFPGKYWLPKYWKLAEKKR



NO: 223
LTGGEEAACEYMADKQLDSPPPNFRPPARCVILAKSRPFEDW




PVHRVASKAQSFVIGLSEQGFAALRAAPPSTADARRDWLRSH




GASEDDLMALEAQLLETIMGNAISLHGGVLKKIDNANVKAA




KRLSGRNEARLNKGLQELPPEQEGSAYGADGLLVNPPGLNLN




IYCRKSCCPKPVKNTARFVGHYPGYLRDSDSILISGTMDRLTII




EGMPGHIPAWQREQGLVKPGGRRRRLSGSESNMRQKVDPST




GPRRSTRSGTVNRSNQRTGRNGDPLLVEIRMKEDWVLLDAR




GLLRNLRWRESKRGLSCDHEDLSLSGLLALFSGDPVIDPVRNE




VVFLYGEGIIPVRSTKPVGTRQSKKLLERQASMGPLTLISCDL




GQTNLIAGRASAISLTHGSLGVRSSVRIELDPEIIKSFERLRKDA




DRLETEILTAAKETLSDEQRGEVNSHEKDSPQTAKASLCRELG




LHPPSLPWGQMGPSTTFIADMLISHGRDDDAFLSHGEFPTLEK




RKKFDKRFCLESRPLLSSETRKALNESLWEVKRTSSEYARLSQ




RKKEMARRAVNFVVEISRRKTGLSNVIVNIEDLNVRIFHGGG




KQAPGWDGFFRPKSENRWFIQAIHKAFSDLAAHHGIPVIESDP




QRTSMTCPECGHCDSKNRNGVRFLCKGCGASMDADFDAACR




NLERVALTGKPMPKPSTSCERLLSATTGKVCSDHSLSHDAIEK




AS





CasΦ.4
SEQ ID
MEKEITELTKIRREFPNKKFSSTDMKKAGKLLKAEGPDAVRD



NO: 224
FLNSCQEIIGDFKPPVKTNIVSISRPFEEWPVSMVGRAIQEYYFS




LTKEELESVHPGTSSEDHKSFFNITGLSNYNYTSVQGLNLIFKN




AKAIYDGTLVKANNKNKKLEKKFNEINHKRSLEGLPIITPDFE




EPFDENGHLNNPPGINRNIYGYQGCAAKVFVPSKHKMVSLPK




EYEGYNRDPNLSLAGFRNRLEIPEGEPGHVPWFQRMDIPEGQI




GHVNKIQRFNFVHGKNSGKVKFSDKTGRVKRYHHSKYKDAT




KPYKFLEESKKVSALDSILAIITIGDDWVVFDIRGLYRNVFYRE




LAQKGLTAVQLLDLFTGDPVIDPKKGVVTFSYKEGVVPVFSQ




KIVPRFKSRDTLEKLTSQGPVALLSVDLGQNEPVAARVCSLK




NINDKITLDNSCRISFLDDYKKQIKDYRDSLDELEIKIRLEAINS




LETNQQVEIRDLDVFSADRAKANTVDMFDIDPNLISWDSMSD




ARVSTQISDLYLKNGGDESRVYFEINNKRIKRSDYNISQLVRP




KLSDSTRKNLNDSIWKLKRTSEEYLKLSKRKLELSRAVVNYTI




RQSKLLSGINDIVIILEDLDVKKKFNGRGIRDIGWDNFFSSRKE




NRWFIPAFHKAFSELSSNRGLCVIEVNPAWTSATCPDCGFCSK




ENRDGINFTCRKCGVSYHADIDVATLNIARVAVLGKPMSGPA




DRERLGDTKKPRVARSRKTMKRKDISNSTVEAMVTA





CasΦ.5
SEQ ID
MDMLDTETNYATETPAQQQDYSPKPPKKAQRAPKGFSKKAR



NO: 225
PEKKPPKPITLFTQKHFSGVRFLKRVIRDASKILKLSESRTITFL




EQAIERDGSAPPDVTPPVHNTIMAVTRPFEEWPEVILSKALQK




HCYALTKKIKIKTWPKKGPGKKCLAAWSARTKIPLIPGQVQA




TNGLFDRIGSIYDGVEKKVTNRNANKKLEYDEAIKEGRNPAV




PEYETAYNIDGTLINKPGYNPNLYITQSRTPRLITEADRPLVEKI




LWQMVEKKTQSRNQARRARLEKAAHLQGLPVPKFVPEKVD




RSQKIEIRIIDPLDKIEPYMPQDRMAIKASQDGHVPYWQRPFLS




KRRNRRVRAGWGKQVSSIQAWLTGALLVIVRLGNEAFLADIR




GALRNAQWRKLLKPDATYQSLFNLFTGDPVVNTRTNHLTMA




YREGVVNIVKSRSFKGRQTREHLLTLLGQGKTVAGVSFDLGQ




KHAAGLLAAHFGLGEDGNPVFTPIQACFLPQRYLDSLTNYRN




RYDALTLDMRRQSLLALTPAQQQEFADAQRDPGGQAKRACC




LKLNLNPDEIRWDLVSGISTMISDLYIERGGDPRDVHQQVETK




PKGKRKSEIRILKIRDGKWAYDFRPKIADETRKAQREQLWKL




QKASSEFERLSRYKINIARAIANWALQWGRELSGCDIVIPVLE




DLNVGSKFFDGKGKWLLGWDNRFTPKKENRWFIKVLHKAV




AELAPHRGVPVYEVMPHRTSMTCPACHYCHPTNREGDRFEC




QSCHVVKNTDRDVAPYNILRVAVEGKTLDRWQAEKKPQAEP




DRPMILIDNQES





CasΦ.6
SEQ ID
MDMLDTETNYATETPAQQQDYSPKPPKKAQRAPKGFSKKAR



NO: 226
PEKKPPKPITLFTQKHFSGVRFLKRVIRDASKILKLSESRTITFL




EQAIERDGSAPPDVTPPVHNTIMAVTRPFEEWPEVILSKALQK




HCYALTKKIKIKTWPKKGPGKKCLAAWSARTKIPLIPGQVQA




TNGLFDRIGSIYDGVEKKVTNRNANKKLEYDEAIKEGRNPAV




PEYETAYNIDGTLINKPGYNPNLYITQSRTPRLITEADRPLVEKI




LWQMVEKKTQSRNQARRARLEKAAHLQGLPVPKFVPEKVD




RSQKIEIRIIDPLDKIEPYMPQDRMAIKASQDGHVPYWQRPFLS




KRRNRRVRAGWGKQVSSIQAWLTGALLVIVRLGNEAFLADIR




GALRNAQWRKLLKPDATYQSLFNLFTGDPVVNTRTNHLTMA




YREGVVDIVKSRSFKGRQTREHLLTLLGQGKTVAGVSFDLGQ




KHAAGLLAAHFGLGEDGNPVFTPIQACFLPQRYLDSLTNYRN




RYDALTLDMRRQSLLALTPAQQQEFADAQRDPGGQAKRACC




LKLNLNPDEIRWDLVSGISTMISDLYIERGGDPRDVHQQVETK




PKGKRKSEIRILKIRDGKWAYDFRPKIADETRKAQREQLWKL




QKASSEFERLSRYKINIARAIANWALQWGRELSGCDIVIPVLE




DLNVGSKFFDGKGKWLLGWDNRFTPKKENRWFIKVLHKAV




AELAPHKGVPVYEVMPHRTSMTCPACHYCHPTNREGDRFEC




QSCHVVKNTDRDVAPYNILRVAVEGKTLDRWQAEKKPQAEP




DRPMILIDNQES





CasΦ.7
SEQ ID
MSSLPTPLELLKQKHADLFKGLQFSSKDNKMAGKVLKKDGE



NO: 227
EAALAFLSERGVSRGELPNFRPPAKTLVVAQSRPFEEFPIYRVS




EAIQLYVYSLSVKELETVPSGSSTKKEHQRFFQDSSVPDFGYT




SVQGLNKIFGLARGIYLGVITRGENQLQKAKSKHEALNKKRR




ASGEAETEFDPTPYEYMTPERKLAKPPGVNHSIMCYVDISVDE




FDFRNPDGIVLPSEYAGYCREINTAIEKGTVDRLGHLKGGPGY




IPGHQRKESTTEGPKINFRKGRIRRSYTALYAKRDSRRVRQGK




LALPSYRHHMMRLNSNAESAILAVIFFGKDWVVFDLRGLLRN




VRWRNLFVDGSTPSTLLGMFGDPVIDPKRGVVAFCYKEQIVP




VVSKSITKMVKAPELLNKLYLKSEDPLVLVAIDLGQTNPVGV




GVYRVMNASLDYEVVTRFALESELLREIESYRQRTNAFEAQIR




AETFDAMTSEEQEEITRVRAFSASKAKENVCHRFGMPVDAVD




WATMGSNTIHIAKWVMRHGDPSLVEVLEYRKDNEIKLDKNG




VPKKVKLTDKRIANLTSIRLRFSQETSKHYNDTMWELRRKHP




VYQKLSKSKADFSRRVVNSIIRRVNHLVPRARIVFIIEDLKNLG




KVFHGSGKRELGWDSYFEPKSENRWFIQVLHKAFSETGKHK




GYYIIECWPNWTSCTCPKCSCCDSENRHGEVFRCLACGYTCN




TDFGTAPDNLVKIATTGKGLPGPKKRCKGSSKGKNPKIARSSE




TGVSVTESGAPKVKKSSPTQTSQSSSQSAP





CasΦ.8
SEQ ID
MNKIEKEKTPLAKLMNENFAGLRFPFAIIKQAGKKLLKEGEL



NO: 228
KTIEYMTGKGSIEPLPNFKPPVKCLIVAKRRDLKYFPICKASCE




IQSYVYSLNYKDFMDYFSTPMTSQKQHEEFFKKSGLNIEYQN




VAGLNLIFNNVKNTYNGVILKVKNRNEKLKKKAIKNNYEFEE




IKTFNDDGCLINKPGINNVIYCFQSISPKILKNITHLPKEYNDYD




CSVDRNIIQKYVSRLDIPESQPGHVPEWQRKLPEFNNTNNPRR




RRKWYSNGRNISKGYSVDQVNQAKIEDSLLAQIKIGEDWIILD




IRGLLRDLNRRELISYKNKLTIKDVLGFFSDYPIIDIKKNLVTFC




YKEGVIQVVSQKSIGNKKSKQLLEKLIENKPIALVSIDLGQTNP




VSVKISKLNKINNKISIESFTYRFLNEEILKEIEKYRKDYDKLEL




KLINEA





CasΦ.9
SEQ ID
MDMLDTETNYATETPSQQQDYSPKPPKKDRRAPKGFSKKAR



NO: 229
PEKKPPKPITLFTQKHFSGVRFLKRVIRDASKILKLSESRTITFL




EQAIERDGSAPPDVTPPVHNTIMAVTRPFEEWPEVILSKALQK




HCYALTKKIKIKTWPKKGPGKKCLAAWSARTKIPLIPGQVQA




TNGLFDRIGSIYDGVEKKVTNRNANKKLEYDEAIKEGRNPAV




PEYETAYNIDGTLINKPGYNPNLYITQSRTPRLITEADRPLVEKI




LWQMVEKKTQSRNQARRARLEKAAHLQGLPVPKFVPEKVD




RSQKIEIRIIDPLDKIEPYMPQDRMAIKASQDGHVPYWQRPFLS




KRRNRRVRAGWGKQVSSIQAWLTGALLVIVRLGNEAFLADIR




GALRNAQWRKLLKPDATYQSLFNLFTGDPVVNTRTNHLTMA




YREGVVDIVKSRSFKGRQTREHLLTLLGQGKTVAGVSFDLGQ




KHAAGLLAAHFGLGEDGNPVFTPIQACFLPQRYLDSLTNYRN




RYDALTLDMRRQSLLALTPAQQQEFADAQRDPGGQAKRACC




LKLNLNPDEIRWDLVSGISTMISDLYIERGGDPRDVHQQVETK




PKGKRKSEIRILKIRDGKWAYDFRPKIADETRKAQREQLWKL




QKASSEFERLSRYKINIARAIANWALQWGRELSGCDIVIPVLE




DLNVGSKFFDGKGKWLLGWDNRFTPKKENRWFIKVLHKAV




AELAPHRGVPVYEVMPHRTSMTCPACHYCHPTNREGDRFEC




QSCHVVKNTDRDVAPYNILRVAVEGKTLDRWQAEKKPQAEP




DRPMILIDNQES





CasΦ.10
SEQ ID
MDMLDTETNYATETPSQQQDYSPKPPKKDRRAPKGFSKKAR



NO: 230
PEKKPPKPITLFTQKHFSGVRFLKRVIRDASKILKLSESRTITFL




EQAIERDGSAPPDVTPPVHNTIMAVTRPFEEWPEVILSKALQK




HCYALTKKIKIKTWPKKGPGKKCLAAWSARTKIPLIPGQVQA




TNGLFDRIGSIYDGVEKKVTNRNANKKLEYDEAIKEGRNPAV




PEYETAYNIDGTLINKPGYNPNLYITQSRTPRLITEADRPLVEKI




LWQMVEKKTQSRNQARRARLEKAAHLQGLPVPKFVPEKVD




RSQKIEIRIIDPLDKIEPYMPQDRMAIKASQDGHVPYWQRPFLS




KRRNRRVRAGWGKQVSSIQAWLTGALLVIVRLGNEAFLADIR




GALRNAQWRKLLKPDATYQSLFNLFTGDPVVNTRTNHLTMA




YREGVVNIVKSRSFKGRQTREHLLTLLGQGKTVAGVSFDLGQ




KHAAGLLAAHFGLGEDGNPVFTPIQACFLPQRYLDSLTNYRN




RYDALTLDMRRQSLLALTPAQQQEFADAQRDPGGQAKRACC




LKLNLNPDEIRWDLVSGISTMISDLYIERGGDPRDVHQQVETK




PKGKRKSEIRILKIRDGKWAYDFRPKIADETRKAQREQLWKL




QKASSEFERLSRYKINIARAIANWALQWGRELSGCDIVIPVLE




DLNVGSKFFDGKGKWLLGWDNRFTPKKENRWFIKVLHKAV




AELAPHRGVPVYEVMPHRTSMTCPACHYCHPTNREGDRFEC




QSCHVVKNTDRDVAPYNILRVAVEGKTLDRWQAEKKPQAEP




DRPMILIDNQES





CasΦ.11
SEQ ID
MSNKTTPPSPLSLLLRAHFPGLKFESQDYKIAGKKLRDGGPEA



NO: 231
VISYLTGKGQAKLKDVKPPAKAFVIAQSRPFIEWDLVRVSRQI




QEKIFGIPATKGRPKQDGLSETAFNEAVASLEVDGKSKLNEET




RAAFYEVLGLDAPSLHAQAQNALIKSAISIREGVLKKVENRNE




KNLSKTKRRKEAGEEATFVEEKAHDERGYLIHPPGVNQTIPG




YQAVVIKSCPSDFIGLPSGCLAKESAEALTDYLPHDRMTIPKG




QPGYVPEWQHPLLNRRKNRRRRDWYSASLNKPKATCSKRSG




TPNRKNSRTDQIQSGRFKGAIPVLMRFQDEWVIIDIRGLLRNA




RYRKLLKEKSTIPDLLSLFTGDPSIDMRQGVCTFIYKAGQACS




AKMVKTKNAPEILSELTKSGPVVLVSIDLGQTNPIAAKVSRVT




QLSDGQLSHETLLRELLSNDSSDGKEIARYRVASDRLRDKLA




NLAVERLSPEHKSEILRAKNDTPALCKARVCAALGLNPEMIA




WDKMTPYTEFLATAYLEKGGDRKVATLKPKNRPEMLRRDIK




FKGTEGVRIEVSPEAAEAYREAQWDLQRTSPEYLRLSTWKQE




LTKRILNQLRHKAAKSSQCEVVVMAFEDLNIKMMHGNGKW




ADGGWDAFFIKKRENRWFMQAFHKSLTELGAHKGVPTIEVT




PHRTSITCTKCGHCDKANRDGERFACQKCGFVAHADLEIATD




NIERVALTGKPMPKPESERSGDAKKSVGARKAAFKPEEDAEA




AE





CasΦ.12
SEQ ID
MIKPTVSQFLTPGFKLIRNHSRTAGLKLKNEGEEACKKFVREN



NO: 232
EIPKDECPNFQGGPAIANIIAKSREFTEWEIYQSSLAIQEVIFTLP




KDKLPEPILKEEWRAQWLSEHGLDTVPYKEAAGLNLIIKNAV




NTYKGVQVKVDNKNKNNLAKINRKNEIAKLNGEQEISFEEIK




AFDDKGYLLQKPSPNKSIYCYQSVSPKPFITSKYHNVNLPEEYI




GYYRKSNEPIVSPYQFDRLRIPIGEPGYVPKWQYTFLSKKENK




RRKLSKRIKNVSPILGIICIKKDWCVFDMRGLLRTNHWKKYH




KPTDSINDLFDYFTGDPVIDTKANVVRFRYKMENGIVNYKPV




REKKGKELLENICDQNGSCKLATVDVGQNNPVAIGLFELKKV




NGELTKTLISRHPTPIDFCNKITAYRERYDKLESSIKLDAIKQLT




SEQKIEVDNYNNNFTPQNTKQIVCSKLNINPNDLPWDKMISGT




HFISEKAQVSNKSEIYFTSTDKGKTKDVMKSDYKWFQDYKPK




LSKEVRDALSDIEWRLRRESLEFNKLSKSREQDARQLANWISS




MCDVIGIENLVKKNNFFGGSGKREPGWDNFYKPKKENRWWI




NAIHKALTELSQNKGKRVILLPAMRTSITCPKCKYCDSKNRN




GEKFNCLKCGIELNADIDVATENLATVAITAQSMPKPTCERSG




DAKKPVRARKAKAPEFHDKLAPSYTVVLREAV





CasΦ.13
SEQ ID
MRQPAEKTAFQVFRQEVIGTQKLSGGDAKTAGRLYKQGKME



NO: 233
AAREWLLKGARDDVPPNFQPPAKCLVVAVSHPFEEWDISKTN




HDVQAYIYAQPLQAEGHLNGLSEKWEDTSADQHKLWFEKTG




VPDRGLPVQAINKIAKAAVNRAFGVVRKVENRNEKRRSRDN




RIAEHNRENGLTEVVREAPEVATNADGFLLHPPGIDPSILSYAS




VSPVPYNSSKHSFVRLPEEYQAYNVEPDAPIPQFVVEDRFAIPP




GQPGYVPEWQRLKCSTNKHRRMRQWSNQDYKPKAGRRAKP




LEFQAHLTRERAKGALLVVMRIKEDWVVFDVRGLLRNVEWR




KVLSEEAREKLTLKGLLDLFTGDPVIDTKRGIVTFLYKAEITKI




LSKRTVKTKNARDLLLRLTEPGEDGLRREVGLVAVDLGQTHP




IAAAIYRIGRTSAGALESTVLHRQGLREDQKEKLKEYRKRHT




ALDSRLRKEAFETLSVEQQKEIVTVSGSGAQITKDKVCNYLG




VDPSTLPWEKMGSYTHFISDDFLRRGGDPNIVHFDRQPKKGK




VSKKSQRIKRSDSQWVGRMRPRLSQETAKARMEADWAAQN




ENEEYKRLARSKQELARWCVNTLLQNTRCITQCDEIVVVIED




LNVKSLHGKGAREPGWDNFFTPKTENRWFIQILHKTFSELPK




HRGEHVIEGCPLRTSITCPACSYCDKNSRNGEKFVCVACGATF




HADFEVATYNLVRLATTGMPMPKSLERQGGGEKAGGARKA




RKKAKQVEKIVVQANANVTMNGASLHSP





CasΦ.14
SEQ ID
MSSLPTPLELLKQKHADLFKGLQFSSKDNKMAGKVLKKDGE



NO: 234
EAALAFLSERGVSRGELPNFRPPAKTLVVAQSRPFEEFPIYRVS




EAIQLYVYSLSVKELETVPSGSSTKKEHQRFFQDSSVPDFGYT




SVQGLNKIFGLARGIYLGVITRGENQLQKAKSKHEALNKKRR




ASGEAETEFDPTPYEYMTPERKLAKPPGVNHSIMCYVDISVDE




FDFRNPDGIVLPSEYAGYCREINTAIEKGTVDRLGHLKGGPGY




IPGHQRKESTTEGPKINFRKGRIRRSYTALYAKRDSRRVRQGK




LALPSYRHHMMRLNSNAESAILAVIFFGKDWVVFDLRGLLRN




VRWRNLFVDGSTPSTLLGMFGDPVIDPKRGVVAFCYKEQIVP




VVSKSITKMVKAPELLNKLYLKSEDPLVLVAIDLGQTNPVGV




GVYRVMNASLDYEVVTRFALESELLREIESYRQRTNAFEAQIR




AETFDAMTSEEQEEITRVRAFSASKAKENVCHRFGMPVDAVD




WATMGSNTIHIAKWVMRHGDPSLVEVLEYRKDNEIKLDKNG




VPKKVKLTDKRIANLTSIRLRFSQETSKHYNDTMWELRRKHP




VYQKLSKSKADFSRRVVNSIIRRVNHLVPRARIVFIIEDLKNLG




KVFHGSGKRELGWDSYFEPKSENRWFIQVLHKAFSETGKHK




GYYIIECWPNWTSCTCPKCSCCDSENRHGEVFRCLACGYTCN




TDFGTAPDNLVKIATTGKGLPGPKKRCKGSSKGKNPKIARSSE




TGVSVTESGAPKVKKSSPTQTSQSSSQSAP





CasΦ.15
SEQ ID
MIKPTVSQFLTPGFKLIRNHSRTAGLKLKNEGEEACKKFVREN



NO: 235
EIPKDECPNFQGGPAIANIIAKSREFTEWEIYQSSLAIQEVIFTLP




KDKLPEPILKEEWRAQWLSEHGLDTVPYKEAAGLNLIIKNAV




NTYKGVQVKVDNKNKNNLAKINRKNEIAKLNGEQEISFEEIK




AFDDKGYLLQKPSPNKSIYCYQSVSPKPFITSKYHNVNLPEEYI




GYYRKSNEPIVSPYQFDRLRIPIGEPGYVPKWQYTFLSKKENK




RRKLSKRIKNVSPILGIICIKKDWCVFDMRGLLRTNHWKKYH




KPTDSINDLFDYFTGDPVIDTKANVVRFRYKMENGIVNYKPV




REKKGKELLENICDQNGSCKLATVDVGQNNPVAIGLFELKKV




NGELTKTLISRHPTPIDFCNKITAYRERYDKLESSIKLDAIKQLT




SEQKIEVDNYNNNFTPQNTKQIVCSKLNINPNDLPWDKMISGT




HFISEKAQVSNKSEIYFTSTDKGKTKDVMKSDYKWFQDYKPK




LSKEVRDALSDIEWRLRRESLEFNKLSKSREQDARQLANWISS




MCDVIGIENLVKKNNFFGGSGKREPGWDNFYKPKKENRWWI




NAIHKALTELSQNKGKRVILLPAMRTSITCPKCKYCDSKNRN




GEKFNCLKCGIELNADIDVATENLATVAITAQSMPKPTCERSG




DAKKPVRARKAKAPEFHDKLAPSYTVVLREAV





CasΦ.16
SEQ ID
MSNKTTPPSPLSLLLRAHFPGLKFESQDYKIAGKKLRDGGPEA



NO: 236
VISYLTGKGQAKLKDVKPPAKAFVIAQSRPFIEWDLVRVSRQI




QEKIFGIPATKGRPKQDGLSETAFNEAVASLEVDGKSKLNEET




RAAFYEVLGLDAPSLHAQAQNALIKSAISIREGVLKKVENRNE




KNLSKTKRRKEAGEEATFVEEKAHDERGYLIHPPGVNQTIPG




YQAVVIKSCPSDFIGLPSGCLAKESAEALTDYLPHDRMTIPKG




QPGYVPEWQHPLLNRRKNRRRRDWYSASLNKPKATCSKRSG




TPNRKNSRTDQIQSGRFKGAIPVLMRFQDEWVIIDIRGLLRNA




RYRKLLKEKSTIPDLLSLFTGDPSIDMRQGVCTFIYKAGQACS




AKMVKTKNAPEILSELTKSGPVVLVSIDLGQTNPIAAKVSRVT




QLSDGQLSHETLLRELLSNDSSDGKEIARYRVASDRLRDKLA




NLAVERLSPEHKSEILRAKNDTPALCKARVCAALGLNPEMIA




WDKMTPYTEFLATAYLEKGGDRKVATLKPKNRPEMLRRDIK




FKGTEGVRIEVSPEAAEAYREAQWDLQRTSPEYLRLSTWKQE




LTKRILNQLRHKAAKSSQCEVVVMAFEDLNIKMMHGNGKW




ADGGWDAFFIKKRENRWFMQAFHKSLTELGAHKGVPTIEVT




PHRTSITCTKCGHCDKANRDGERFACQKCGFVAHADLEIATD




NIERVALTGKPMPKPESERSGDAKKSVGARKAAFKPEEDAEA




AE


CasΦ.17
SEQ ID
MYSLEMADLKSEPSLLAKLLRDRFPGKYWLPKYWKLAEKKR



NO: 237
LTGGEEAACEYMADKQLDSPPPNFRPPARCVILAKSRPFEDW




PVHRVASKAQSFVIGLSEQGFAALRAAPPSTADARRDWLRSH




GASEDDLMALEAQLLETIMGNAISLHGGVLKKIDNANVKAA




KRLSGRNEARLNKGLQELPPEQEGSAYGADGLLVNPPGLNLN




IYCRKSCCPKPVKNTARFVGHYPGYLRDSDSILISGTMDRLTII




EGMPGHIPAWQREQGLVKPGGRRRRLSGSESNMRQKVDPST




GPRRSTRSGTVNRSNQRTGRNGDPLLVEIRMKEDWVLLDAR




GLLRNLRWRESKRGLSCDHEDLSLSGLLALFSGDPVIDPVRNE




VVFLYGEGIIPVRSTKPVGTRQSKKLLERQASMGPLTLISCDL




GQTNLIAGRASAISLTHGSLGVRSSVRIELDPEIIKSFERLRKDA




DRLETEILTAAKETLSDEQRGEVNSHEKDSPQTAKASLCRELG




LHPPSLPWGQMGPSTTFIADMLISHGRDDDAFLSHGEFPTLEK




RKKFDKRFCLESRPLLSSETRKALNESLWEVKRTSSEYARLSQ




RKKEMARRAVNFVVEISRRKTGLSNVIVNIEDLNVRIFHGGG




KQAPGWDGFFRPKSENRWFIQAIHKAFSDLAAHHGIPVIESDP




QRTSMTCPECGHCDSKNRNGVRFLCKGCGASMDADFDAACR




NLERVALTGKPMPKPSTSCERLLSATTGKVCSDHSLSHDAIEK




AS





CasΦ.18
SEQ ID
MEKEITELTKIRREFPNKKFSSTDMKKAGKLLKAEGPDAVRD



NO: 238
FLNSCQEIIGDFKPPVKTNIVSISRPFEEWPVSMVGRAIQEYYFS




LTKEELESVHPGTSSEDHKSFFNITGLSNYNYTSVQGLNLIFKN




AKAIYDGTLVKANNKNKKLEKKFNEINHKRSLEGLPIITPDFE




EPFDENGHLNNPPGINRNIYGYQGCAAKVFVPSKHKMVSLPK




EYEGYNRDPNLSLAGFRNRLEIPEGEPGHVPWFQRMDIPEGQI




GHVNKIQRFNFVHGKNSGKVKFSDKTGRVKRYHHSKYKDAT




KPYKFLEESKKVSALDSILAHITIGDDWVVFDIRGLYRNVFYRE




LAQKGLTAVQLLDLFTGDPVIDPKKGVVTFSYKEGVVPVFSQ




KIVPRFKSRDTLEKLTSQGPVALLSVDLGQNEPVAARVCSLK




NINDKITLDNSCRISFLDDYKKQIKDYRDSLDELEIKIRLEAINS




LETNQQVEIRDLDVFSADRAKANTVDMFDIDPNLISWDSMSD




ARVSTQISDLYLKNGGDESRVYFEINNKRIKRSDYNISQLVRP




KLSDSTRKNLNDSIWKLKRTSEEYLKLSKRKLELSRAVVNYTI




RQSKLLSGINDIVIILEDLDVKKKFNGRGIRDIGWDNFFSSRKE




NRWFIPAFHKTFSELSSNRGLCVIEVNPAWTSATCPDCGFCSK




ENRDGINFTCRKCGVSYHADIDVATLNIARVAVLGKPMSGPA




DRERLGDTKKPRVARSRKTMKRKDISNSTVEAMVTA





CasΦ.19
SEQ ID
MLVRTSTLVQDNKNSRSASRAFLKKPKMPKNKHIKEPTELAK



NO: 239
LIRELFPGQRFTRAINTQAGKILKHKGRDEVVEFLKNKGIDKE




QFMDFRPPTKARIVATSGAIEEFSYLRVSMAIQECCFGKYKFP




KEKVNGKLVLETVGLTKEELDDFLPKKYYENKKSRDRFFLKT




GICDYGYTYAQGLNEIFRNTRAIYEGVFTKVNNRNEKRREKK




DKYNEERRSKGLSEEPYDEDESATDESGHLINPPGVNLNIWTC




EGFCKGPYVTKLSGTPGYEVILPKVFDGYNRDPNEIISCGITDR




FAIPEGEPGHIPWHQRLEIPEGQPGYVPGHQRFADTGQNNSGK




ANPNKKGRMRKYYGHGTKYTQPGEYQEVFRKGHREGNKRR




YWEEDFRSEAHDCILYVIHIGDDWVVCDLRGPLRDAYRRGLV




PKEGITTQELCNLFSGDPVIDPKHGVVTFCYKNGLVRAQKTIS




AGKKSRELLGALTSQGPIALIGVDLGQTEPVGARAFIVNQARG




SLSLPTLKGSFLLTAENSSSWNVFKGEIKAYREAIDDLAIRLKK




EAVATLSVEQQTEIESYEAFSAEDAKQLACEKFGVDSSFILWE




DMTPYHTGPATYYFAKQFLKKNGGNKSLIEYIPYQKKKSKKT




PKAVLRSDYNIACCVRPKLLPETRKALNEAIRIVQKNSDEYQR




LSKRKLEFCRRVVNYLVRKAKKLTGLERVIIAIEDLKSLEKFF




TGSGKRDNGWSNFFRPKKENRWFIPAFHKAFSELAPNRGFYV




IECNPARTSITDPDCGYCDGDNRDGIKFECKKCGAKHHTDLD




VAPLNIAIVAVTGRPMPKTVSNKSKRERSGGEKSVGASRKRN




HRKSKANQEMLDATSSAAE





CasΦ.20
SEQ ID
MPKIKKPTEISLLRKEVFPDLHFAKDRMRAASLVLKNEGREA



NO: 240
AIEYLRVNHEDKPPNFMPPAKTPYVALSRPLEQWPIAQASIAI




QKYIFGLTKDEFSATKKLLYGDKSTPNTESRKRWFEVTGVPN




FGYMSAQGLNAIFSGALARYEGVVQKVENRNKKRFEKLSEK




NQLLIEEGQPVKDYVPDTAYHTPETLQKLAENNHVRVEDLGD




MIDRLVHPPGIHRSIYGYQQVPPFAYDPDNPKGIILPKAYAGY




TRKPHDIIEAMPNRLNIPEGQAGYIPEHQRDKLKKGGRVKRLR




TTRVRVDATETVRAKAEALNAEKARLRGKEAILAVFQIEEDW




ALIDMRGLLRNVYMRKLIAAGELTPTTLLGYFTETLTLDPRRT




EATFCYHLRSEGALHAEYVRHGKNTRELLLDLTKDNEKIALV




TIDLGQRNPLAAAIFRVGRDASGDLTENSLEPVSRMLLPQAYL




DQIKAYRDAYDSFRQNIWDTALASLTPEQQRQILAYEAYTPD




DSKENVLRLLLGGNVMPDDLPWEDMTKNTHYISDRYLADGG




DPSKVWFVPGPRKRKKNAPPLKKPPKPRELVKRSDHNISHLSE




FRPQLLKETRDAFEKAKIDTERGHVGYQKLSTRKDQLCKEIL




NWLEAEAVRLTRCKTMVLGLEDLNGPFFNQGKGKVRGWVS




FFRQKQENRWIVNGFRKNALARAHDKGKYILELWPSWTSQT




CPKCKHVHADNRHGDDFVCLQCGARLHADAEVATWNLAVV




AIQGHSLPGPVREKSNDRKKSGSARKSKKANESGKVVGAWA




AQATPKRATSKKETGTARNPVYNPLETQASCPAP





CasΦ.21
SEQ ID
MTPSPQIARLVETPLAAALKAHHPGKKFRSDYLKKAGKILKD



NO: 241
QGVEAAMAHLDGKDQAEPPNFKPPAKCRIVARSREFSEWPIV




KASVEIQKYIYGLTLEERKACDPGKSSASHKAWFAKTGVNTF




GYSSVQGFNLIFGHTLGRYDGVLVKTENLNKKRAEKNERFRA




KALAEGRAEPVCPPLVTATNDTGQDVTLEDGRVVRPGQLLQP




PGINPNIYAYQQVSPKAYVPGIIELPEEFQGYSRDPNAVILPLV




PRDRLSIPKGQPGYVPEPHREGLTGRKDRRMRRYYETERGTK




LKRPPLTAKGRADKANEALLVVVRIDSDWVVMDVRGLLRNA




RWRRLVSKEGITLNGLLDLFTGDPVLNPKDCSVSRDTGDPVN




DPRHGVVTFCYKLGVVDVCSKDRPIKGFRTKEVLERLTSSGT




VGMVSIDLGQTNPVAAAVSRVTKGLQAETLETFTLPDDLLGK




VRAYRAKTDRMEEGFRRNALRKLTAEQQAEITRYNDATEQQ




AKALVCSTYGIGPEEVPWERMTSNTTYISDHILDHGGDPDTVF




FMATKRGQNKPTLHKRKDKAWGQKFRPAISVETRLARQAAE




WELRRASLEFQKLSVWKTELCRQAVNYVMERTKKRTQCDVI




IPVIEDLPVPLFHGSGKRDPGWANFFVHKRENRWFIDGLHKAF




SELGKHRGIYVFEVCPQRTSITCPKCGHCDPDNRDGEKFVCLS




CQATLNADLDVATTNLVRVALTGKVMPRSERSGDAQTPGPA




RKARTGKIKGSKPTSAPQGATQTDAKAHLSQTGV





CasΦ.22
SEQ ID
MTPSPQIARLVETPLAAALKAHHPGKKFRSDYLKKAGKILKD



NO: 242
QGVEAAMAHLDGKDQAEPPNFKPPAKCRIVARSREFSEWPIV




KASVEIQKYIYGLTLEERKACDPGKSSASHKAWFAKTGVNTF




GYSSVQGFNLIFGHTLGRYDGVLVKTENLNKKRAEKNERFRA




KALAEGRAEPVCPPLVTATNDTGQDVTLEDGRVVRPGQLLQP




PGINPNIYAYQQVSPKAYVPGIIELPEEFQGYSRDPNAVILPLV




PRDRLSIPKGQPGYVPEPHREGLTGRKDRRMRRYYETERGTK




LKRPPLTAKGRADKANEALLVVVRIDSDWVVMDVRGLLRNA




RWRRLVSKEGITLNGLLDLFTGDPVLNPKDCSVSRDTGDPVN




DPRHGVVTFCYKLGVVDVCSKDRPIKGFRTKEVLERLTSSGT




VGMVSIDLGQTNPVAAAVSRVTKGLQAETLETFTLPDDLLGK




VRAYRAKTDRMEEGFRRNALRKLTAEQQAEITRYNDATEQQ




AKALVCSTYGIGPEEVPWERMTSNTTYISDHILDHGGDPDTVF




FMATKRGQNKPTLHKRKDKAWGQKFRPAISVETRLARQAAE




WELRRASLEFQKLSVWKTELCRQAVNYVMERTKKRTQCDVI




IPVIEDLPVPLFHGSGKRDPGWANFFVHKRENRWFIDGLHKAF




SELGKHRGIYVFEVCPQRTSITCPKCGHCDPDNRDGEKFVCLS




CQATLHADLDVATTNLVRVALTGKVMPRSERSGDAQTPGPA




RKARTGKIKGSKPTSAPQGATQTDAKAHLSQTGV





CasΦ.23
SEQ ID
MKTEKPKTALTLLREEVFPGKKYRLDVLKEAGKKLSTKGRE



NO: 243
ATIEFLTGKDEERPQNFQPPAKTSIVAQSRPFDQWPIVQVSLA




VQKYIYGLTQSEFEANKKALYGETGKAISTESRRAWFEATGV




DNFGFTAAQGINPIFSQAVARYEGVIKKVENRNEKKLKKLTK




KNLLRLESGEEIEDFEPEATFNEEGRLLQPPGANPNIYCYQQIS




PRIYDPSDPKGVILPQIYAGYDRKPEDIISAGVPNRLAIPEGQPG




YIPEHQRAGLKTQGRIRCRASVEAKARAAILAVVHLGEDWVV




LDLRGLLRNVYWRKLASPGTLTLKGLLDFFTGGPVLDARRGI




ATFSYTLKSAAAVHAENTYKGKGTREVLLKLTENNSVALVT




VDLGQRNPLAAMIARVSRTSQGDLTYPESVEPLTRLFLPDPFL




EEVRKYRSSYDALRLSIREAAIASLTPEQQAEIRYIEKFSAGDA




KKNVAEVFGIDPTQLPWDAMTPRTTYISDLFLRMGGDRSRVF




FEVPPKKAKKAPKKPPKKPAGPRIVKRTDGMIARLREIRPRLS




AETNKAFQEARWEGERSNVAFQKLSVRRKQFARTVVNHLVQ




TAQKMSRCDTVVLGIEDLNVPFFHGRGKYQPGWEGFFRQKK




ENRWLINDMHKALSERGPHRGGYVLELTPFWTSLRCPKCGH




TDSANRDGDDFVCVKCGAKLHSDLEVATANLALVAITGQSIP




RPPREQSSGKKSTGTARMKKTSGETQGKGSKACVSEALNKIE




QGTARDPVYNPLNSQVSCPAP





CasΦ.24
SEQ ID
VYNPDMKKPNNIRRIREEHFEGLCFGKDVLTKAGKIYEKDGE



NO: 244
EAAIDFLMGKDEEDPPNFKPPAKTTIVAQSRPFDQWPIYQVSQ




AVQERVFAYTEEEFNASKEALFSGDISSKSRDFWFKTNNISDQ




GIGAQGLNTILSHAFSRYSGVIKKVENRNKKRLKKLSKKNQL




KIEEGLEILEFKPDSAFNENGLLAQPPGINPNIYGYQAVTPFVF




DPDNPGDVILPKQYEGYSRKPDDIIEKGPSRLDIPKGQPGYVPE




HQRKNLKKKGRVRLYRRTPPKTKALASILAVLQIGKDWVLFD




MRGLLRSVYMREAATPGQISAKDLLDTFTGCPVLNTRTGEFT




FCYKLRSEGALHARKIYTKGETRTLLTSLTSENNTIALVTVDL




GQRNPAAIMISRLSRKEELSEKDIQPVSRRLLPDRYLNELKRY




RDAYDAFRQEVRDEAFTSLCPEHQEQVQQYEALTPEKAKNL




VLKHFFGTHDPDLPWDDMTSNTHYIANLYLERGGDPSKVFFT




RPLKKDSKSKKPRKPTKRTDASISRLPEIRPKMPEDARKAFEK




AKWEIYTGHEKFPKLAKRVNQLCREIANWIEKEAKRLTLCDT




VVVGIEDLSLPPKRGKGKFQETWQGFFRQKFENRWVIDTLKK




AIQNRAHDKGKYVLGLAPYWTSQRCPACGFIHKSNRNGDHF




KCLKCEALFHADSEVATWNLALVAVLGKGITNPDSKKPSGQ




KKTGTTRKKQIKGKNKGKETVNVPPTTQEVEDIIAFFEKDDET




VRNPVYKPTGT





CasΦ.25
SEQ ID
MKKPNNIRRIREEHFEGLCFGKDVLTKAGKIYEKDGEEAAIDF



NO: 245
LMGKDEEDPPNFKPPAKTTIVAQSRPFDQWPIYQVSQAVQER




VFAYTEEEFNASKEALFSGDISSKSRDFWFKTNNISDQGIGAQ




GLNTILSHAFSRYSGVIKKVENRNKKRLKKLSKKNQLKIEEGL




EILEFKPDSAFNENGLLAQPPGINPNIYGYQAVTPFVFDPDNPG




DVILPKQYEGYSRKPDDIIEKGPSRLDIPKGQPGYVPEHQRKN




LKKKGRVRLYRRTPPKTKALASILAVLQIGKDWVLFDMRGLL




RSVYMREAATPGQISAKDLLDTFTGCPVLNTRTGEFTFCYKL




RSEGALHARKIYTKGETRTLLTSLTSENNTIALVTVDLGQRNP




AAIMISRLSRKEELSEKDIQPVSRRLLPDRYLNELKRYRDAYD




AFRQEVRDEAFTSLCPEHQEQVQQYEALTPEKAKNLVLKHFF




GTHDPDLPWDDMTSNTHYIANLYLERGGDPSKVFFTRPLKKD




SKSKKPRKPTKRTDASISRLPEIRPKMPEDARKAFEKAKWEIY




TGHEKFPKLAKRVNQLCREIANWIEKEAKRLTLCDTVVVGIE




DLSLPPKRGKGKFQETWQGFFRQKFENRWVIDTLKKAIQNRA




HDKGKYVLGLAPYWTSQRCPACGFIHKSNRNGDHFKCLKCE




ALFHADSEVATWNLALVAVLGKGITNPDSKKPSGQKKTGTT




RKKQIKGKNKGKETVNVPPTTQEVEDIIAFFEKDDETVRNPVY




KPTGT





CasΦ.26
SEQ ID
VIKTHFPAGRFRKDHQKTAGKKLKHEGEEACVEYLRNKVSD



NO: 246
YPPNFKPPAKGTIVAQSRPFSEWPIVRASEAIQKYVYGLTVAE




LDVFSPGTSKPSHAEWFAKTGVENYGYRQVQGLNTIFQNTVN




RFKGVLKKVENRNKKSLKRQEGANRRRVEEGLPEVPVTVES




ATDDEGRLLQPPGVNPSIYGYQGVAPRVCTDLQGFSGMSVDF




AGYRRDPDAVLVESLPEGRLSIPKGERGYVPEWQRDPERNKF




PLREGSRRQRKWYSNACHKPKPGRTSKYDPEALKKASAKDA




LLVSISIGEDWAIIDVRGLLRDARRRGFTPEEGLSLNSLLGLFT




EYPVFDVQRGLITFTYKLGQVDVHSRKTVPTFRSRALLESLVA




KEEIALVSVDLGQTNPASMKVSRVRAQEGALVAEPVHRMFLS




DVLLGELSSYRKRMDAFEDAIRAQAFETMTPEQQAEITRVCD




VSVEVARRRVCEKYSISPQDVPWGEMTGHSTFIVDAVLRKGG




DESLVYFKNKEGETLKFRDLRISRMEGVRPRLTKDTRDALNK




AVLDLKRAHPTFAKLAKQKLELARRCVNFIEREAKRYTQCER




VVFVIEDLNVGFFHGKGKRDRGWDAFFTAKKENRWVIQALH




KAFSDLGLHRGSYVIEVTPQRTSMTCPRCGHCDKGNRNGEKF




VCLQCGATLHADLEVATDNIERVALTGKAMPKPPVRERSGD




VQKAGTARKARKPLKPKQKTEPSVQEGSSDDGVDKSPGDAS




RNPVYNPSDTLSI





CasΦ.27
SEQ ID
MAKAKTLAALLRELLPGQHLAPHHRWVANKLLMTSGDAAA



NO: 247
FVIGKSVSDPVRGSFRKDVITKAGRIFKKDGPDAAAAFLDGK




WEDRPPNFQPPAKAAIVAISRSFDEWPIVKVSCAIQQYLYALP




VQEFESSVPEARAQAHAAWFQDTGVDDCNFKSTQGLNAIFN




HGKRTYEGVLKKAQNRNDKKNLRLERINAKRAEAGQAPLVA




GPDESPTDDAGCLLHPPGINANIYCYQQVSPRPYEQSCGIQLPP




EYAGYNRLSNVAIPPMPNRLDIPQGQPGYVPEHHRHGIKKFG




RVRKRYGVVPGRNRDADGKRTRQVLTEAGAAAKARDSVLA




VIRIGDDWTVVDLRGLLRNAQWRKLVPDGGITVQGLLDLFTG




DPVIDPRRGVVTFIYKADSVGIHSEKVCRGKQSKNLLERLCA




MPEKSSTRLDCARQAVALVSVDLGQRNPVAARFSRVSLAEG




QLQAQLVSAQFLDDAMVAMIRSYREEYDRFESLVREQAKAA




LSPEQLSEIVRHEADSAESVKSCVCAKFGIDPAGLSWDKMTSG




TWRIADHVQAAGGDVEWFFFKTCGKGKEIKTVRRSDFNVAK




QFRLRLSPETRKDWNDAIWELKRGNPAYVSFSKRKSEFARRV




VNDLVHRARRAVRCDEVVFAIEDLNISFFHGKGQRQMGWDA




FFEVKQENRWFIQALHKAFVERATHKGGYVLEVAPARTSTTC




PECRHCDPESRRGEQFCCIKCRHTCHADLEVATFNIEQVALTG




VSLPKRLSSTLL





CasΦ.28
SEQ ID
MSKEKTPPSAYAILKAKHFPDLDFEKKHKMMAGRMFKNGAS



NO: 248
EQEVVQYLQGKGSESLMDVKPPAKSPILAQSRPFDEWEMVRT




SRLIQETIFGIPKRGSIPKRDGLSETQFNELVASLEVGGKPMLN




KQTRAIFYGLLGIKPPTFHAMAQNILIDLAINIRKGVLKKVDNL




NEKNRKKVKRIRDAGEQDVMVPAEVTAHDDRGYLNHPPGV




NPTIPGYQGVVIPFPEGFEGLPSGMTPVDWSHVLVDYLPHDRL




SIPKGSPGYIPEWQRPLLNRHKGRRHRSWYANSLNKPRKSRT




EEAKDRQNAGKRTALIEAERLKGVLPVLMRFKEDWLIIDARG




LLRNARYRGVLPEGSTLGNLIDLFSDSPRVDTRRGICTFLYRK




GRAYSTKPVKRKESKETLLKLTEKSTIALVSIDLGQTNPLTAK




LSKVRQVDGCLVAEPVLRKLIDNASEDGKEIARYRVAHDLLR




ARILEDAIDLLGIYKDEVVRARSDTPDLCKERVCRFLGLDSQA




IDWDRMTPYTDFIAQAFVAKGGDPKVVTIKPNGKPKMFRKD




RSIKNMKGIRLDISKEASSAYREAQWAIQRESPDFQRLAVWQS




QLTKRIVNQLVAWAKKCTQCDTVVLAFEDLNIGMMHGSGK




WANGGWNALFLHKQENRWFMQAFHKALTELSAHKGIPTIEV




LPHRTSITCTQCGHCHPGNRDGERFKCLKCEFLANTDLEIATD




NIERVALTGLPMPKGERSSAKRKPGGTRKTKKSKHSGNSPLA




AE





CasΦ.29
SEQ ID
MEKAGPTSPLSVLIHKNFEGCRFQIDHLKIAGRKLAREGEAAA



NO: 249
IEYLLDKKCEGLPPNFQPPAKGNVIAQSRPFTEWAPYRASVAI




QKYIYSLSVDERKVCDPGSSSDSHEKWFKQTGVQNYGYTHV




QGLNLIFKHALARYDGVLKKVDNRNEKNRKKAERVNSFRRE




EGLPEEVFEEEKATDETGHLLQPPGVNHSIYCYQSVRPKPFNP




RKPGGISLPEAYSGYSLKPQDELPIGSLDRLSIPPGQPGYVPEW




QRSQLTTQKHRRKRSWYSAQKWKPRTGRTSTFDPDRLNCAR




AQGAILAVVRIHEDWVVFDVRGLLRNALWRELAGKGLTVRD




LLDFFTGDPVVDTKRGVVTFTYKLGKVDVHSLRTVRGKRSK




KVLEDLTLSSDVGLVTIDLGQTNVLAADYSKVTRSENGELLA




VPLSKSFLPKHLLHEVTAYRTSYDQMEEGFRRKALLTLTEDQ




QVEVTLVRDFSVESSKTKLLQLGVDVTSLPWEKMSSNTTYIS




DQLLQQGADPASLFFDGERDGKPCRHKKKDRTWAYLVRPKV




SPETRKALNEALWALKNTSPEFESLSKRKIQFSRRCMNYLLNE




AKRISGCGQVVFVIEDLNVRVHHGRGKRAIGWDNFFKPKREN




RWFMQALHKAASELAIHRGMHIIEACPARSSITCPKCGHCDPE




NRCSSDREKFLCVKCGAAFHADLEVATFNLRKVALTGTALPK




SIDHSRDGLIPKGARNRKLKEPQANDEKACA





CasΦ.30
SEQ ID
MKEQSPLSSVLKSNFPGKKFLSADIRVAGRKLAQLGEAAAVE



NO: 250
YLSPRQRDSVPNFRPPAFCTVVAKSRPFEEWPIYKASVLLQEQ




IYGMTGQEFEERCGSIPTSLSGLRQWASSVGLGAAMEGLHVQ




GMNLMVKNAINRYKGVLVKVENRNKKLVEANEAKNSSREE




RGLPPLRPPELGSAFGPDGRLVNPPGIDKSIRLYQGVSPVPVVK




TTGRPTVHRLDIPAGEKGHVPLWQREAGLVKEGPRRRRMWY




SNSNLKRSRKDRSAEASEARKADSVVVRVSVKEDWVDIDVR




GLLRNVAWRGIERAGESTEDLLSLFSGDPVVDPSRDSVVFLY




KEGVVDVLSKKVVGAGKSRKQLEKMVSEGPVALVSCDLGQT




NYVAARVSVLDESLSPVRSFRVDPREFPSADGSQGVVGSLDRI




RADSDRLEAKLLSEAEASLPEPVRAEIEFLRSERPSAVAGRLCL




KLGIDPRSIPWEKMGSTTSFISEALSAKGSPLALHDGAPIKDSR




FAHAARGRLSPESRKALNEALWERKSSSREYGVISRRKSEASR




RMANAVLSESRRLTGLAVVAVNLEDLNMVSKFFHGRGKRAP




GWAGFFTPKMENRWFIRSIHKAMCDLSKHRGITVIESRPERTS




ISCPECGHCDPENRSGERFSCKSCGVSLHADFEVATRNLERVA




LTGKPMPRRENLHSPEGATASRKTRKKPREATASTFLDLRSVL




SSAENEGSGPAARAG





CasΦ.31
SEQ ID
MLPPSNKIGKSMSLKEFINKRNFKSSIIKQAGKILKKEGEEAVK



NO: 251
KYLDDNYVEGYKKRDFPITAKCNIVASNRKIEDFDISKFSSFIQ




NYVFNLNKDNFEEFSKIKYNRKSFDELYKKIANEIGLEKPNYE




NIQGEIAVIRNAINIYNGVLKKVENRNKKIQEKNQSKDPPKLL




SAFDDNGFLAERPGINETIYGYQSVRLRHLDVEKDKDIIVQLP




DIYQKYNKKSTDKISVKKRLNKYNVDEYGKLISKRRKERINK




DDAILCVSNFGDDWIIFDARGLLRQTYRYKLKKKGLCIKDLL




NLFTGDPIINPTKTDLKEALSLSFKDGIINNRTLKVKNYKKCPE




LISELIRDKGKVAMISIDLGQTNPISYRLSKFTANNVAYIENGVI




SEDDIVKMKKWREKSDKLENLIKEEAIASLSDDEQREVRLYE




NDIADNTKKKILEKFNIREEDLDFSKMSNNTYFIRDCLKNKNI




DESEFTFEKNGKKLDPTDACFAREYKNKLSELTRKKINEKIWE




IKKNSKEYHKISIYKKETIRYIVNKLIKQSKEKSECDDIIVNIEK




LQIGGNFFGGRGKRDPGWNNFFLPKEENRWFINACHKAFSEL




APHKGIIVIESDPAYTSQTCPKCENCDKENRNGEKFKCKKCNY




EANADIDVATENLEKIAKNGRRLIKNFDQLGERLPGAEMPGG




ARKRKPSKSLPKNGRGAGVGSEPELINQSPSQVIA





CasΦ.32
SEQ ID
VPDKKETPLVALCKKSFPGLRFKKHDSRQAGRILKSKGEGAA



NO: 252
VAFLEGKGGTTQPNFKPPVKCNIVAMSRPLEEWPIYKASVVIQ




KYVYAQSYEEFKATDPGKSEAGLRAWLKATRVDTDGYFNV




QGLNLIFQNARATYEGVLKKVENRNSKKVAKIEQRNEHRAER




GLPLLTLDEPETALDETGHLRHRPGINCSVFGYQHMKLKPYV




PGSIPGVTGYSRDPSTPIAACGVDRLEIPEGQPGYVPPWDREN




LSVKKHRRKRASWARSRGGAIDDNMLLAVVRVADDWALLD




LRGLLRNTQYRKLLDRSVPVTIESLLNLVTNDPTLSVVKKPGK




PVRYTATLIYKQGVVPVVKAKVVKGSYVSKMLDDTTETFSL




VGVDLGVNNLIAANALRIRPGKCVERLQAFTLPEQTVEDFFRF




RKAYDKHQENLRLAAVRSLTAEQQAEVLALDTFGPEQAKMQ




VCGHLGLSVDEVPWDKVNSRSSILSDLAKERGVDDTLYMFPF




FKGKGKKRKTEIRKRWDVNWAQHFRPQLTSETRKALNEAK




WEAERNSSKYHQLSIRKKELSRHCVNYVIRTAEKRAQCGKVI




VAVEDLHHSFRRGGKGSRKSGWGGFFAAKQEGRWLMDALF




GAFCDLAVHRGYRVIKVDPYNTSRTCPECGHCDKANRDRVN




REAFICVCCGYRGNADIDVAAYNIAMVAITGVSLRKAARASV




ASTPLESLAAE





CasΦ.33
SEQ ID
MSKTKELNDYQEALARRLPGVRHQKSVRRAARLVYDRQGE



NO: 253
DAMVAFLDGKEVDEPYTLQPPAKCHILAVSRPIEEWPIARVT




MAVQEHVYALPVHEVEKSRPETTEGSRSAWFKNSGVSNHGV




THAQTLNAILKNAYNVYNGVIKKVENRNAKKRDSLAAKNKS




RERKGLPHFKADPPELATDEQGYLLQPPSPNSSVYLVQQHLR




TPQIDLPSGYTGPVVDPRSPIPSLIPIDRLAIPPGQPGYVPLHDR




EKLTSNKHRRMKLPKSLRAQGALPVCFRVFDDWAVVDGRGL




LRHAQYRRLAPKNVSIAELLELYTGDPVIDIKRNLMTFRFAEA




VVEVTARKIVEKYHNKYLLKLTEPKGKPVREIGLVSIDLNVQ




RLIALAIYRVHQTGESQLALSPCLHREILPAKGLGDFDKYKSK




FNQLTEEILTAAVQTLTSAQQEEYQRYVEESSHEAKADLCLK




YSITPHELAWDKMTSSTQYISRWLRDHGWNASDFTQITKGRK




KVERLWSDSRWAQELKPKLSNETRRKLEDAKHDLQRANPEW




QRLAKRKQEYSRHLANTVLSMAREYTACETVVIAIENLPMKG




GFVDGNGSRESGWDNFFTHKKENRWMIKDIHKALSDLAPNR




GVHVLEVNPQYTSQTCPECGHRDKANRDPIQRERFCCTHCGA




QRHADLEVATHNIAMVATTGKSLTGKSLAPQRLQEAAE





CasΦ.41
SEQ ID
VLLSDRIQYTDPSAPIPAMTVVDRRKIKKGEPGYVPPFMRKNL



NO: 254
STNKHRRMRLSRGQKEACALPVGLRLPDGKDGWDFIIFDGRA




LLRACRRLRLEVTSMDDVLDKFTGDPRIQLSPAGETIVTCMLK




PQHTGVIQQKLITGKMKDRLVQLTAEAPIAMLTVDLGEHNLV




ACGAYTVGQRRGKLQSERLEAFLLPEKVLADFEGYRRDSDEH




SETLRHEALKALSKRQQREVLDMLRTGADQARESLCYKYGL




DLQALPWDKMSSNSTFIAQHLMSLGFGESATHVRYRPKRKAS




ERTILKYDSRFAAEEKIKLTDETRRAWNEAIWECQRASQEFRC




LSVRKLQLARAAVNWTLTQAKQRSRCPRVVVVVEDLNVRF




MHGGGKRQEGWAGFFKARSEKRWFIQALHKAYTELPTNRGI




HVMEVNPARTSITCTKCGYCDPENRYGEDFHCRNPKCKVRG




GHVANADLDIATENLARVALSGPMPKAPKLK





CasΦ.34
SEQ ID
MTPSFGYQMIIVTPIHHASGAWATLRLLFLNPKTSGVMLGMT



NO: 255
KTKSAFALMREEVFPGLLFKSADLKMAGRKFAKEGREAAIEY




LRGKDEERPANFKPPAKGDIIAQSRPFDQWPIVQVSQAIQKYIF




GLTKAEFDATKTLLYGEGNHPTTESRRRWFEATGVPDFGFTS




AQGLNAIFSSALARYEGVIQKVENRNEKRLKKLSEKNQRLVE




EGHAVEAYVPETAFHTLESLKALSEKSLVPLDDLMDKIDRLA




QPPGINPCLYGYQQVAPYIYDPENPRGVVLPDLYLGYCRKPD




DPITACPNRLDIPKGQPGYIPEHQRGQLKKHGRVRRFRYTNPQ




AKARAKAQTAILAVLRIDEDWVVMDLRGLLRNVYFREVAAP




GELTARTLLDTFTGCPVLNLRSNVVTFCYDIESKGALHAEYV




RKGWATRNKLLDLTKDGQSVALLSVDLGQRHPVAVMISRLK




RDDKGDLSEKSIQVVSRTFADQYVDKLKRYRVQYDALRKEIY




DAALVSLPPEQQAEIRAYEAFAPGDAKANVLSVMFQGEVSPD




ELPWDKMNTNTHYISDLYLRRGGDPSRVFFVPQPSTPKKNAK




KPPAPRKPVKRTDENVSHMPEFRPHLSNETREAFQKAKWTM




ERGNVRYAQLSRFLNQIVREANNWLVSEAKKLTQCQTVVWA




IEDLHVPFFHGKGKYHETWDGFFRQKKEDRWFVNVFHKAISE




RAPNKGEYVMEVAPYRTSQRCPVCGFVDADNRHGDHFKCLR




CGVELHADLEVATWNIALVAVQGHGIAGPPREQSCGGETAG




TARKGKNIKKNKGLADAVTVEAQDSEGGSKKDAGTARNPVY




IPSESQVNCPAP





CasΦ.35
SEQ ID
MKPKTPKPPKTPVAALIDKHFPGKRFRASYLKSVGKKLKNQG



NO: 256
EDVAVRFLTGKDEERPPNFQPPAKSNIVAQSRPIEEWPIHKVS




VAVQEYVYGLTVAEKEACSDAGESSSSHAAWFAKTGVENFG




YTSVQGLNKIFPPTFNRFDGVIKKVENRNEKKRQKATRINEAK




RNKGQSEDPPEAEVKATDDAGYLLQPPGINHSVYGYQSITLCP




YTAEKFPTIKLPEEYAGYHSNPDAPIPAGVPDRLAIPEGQPGH




VPEEHRAGLSTKKHRRVRQWYAMANWKPKPKRTSKPDYDR




LAKARAQGALLIVIRIDEDWVVVDARGLLRNVRWRSLGKREI




TPNELLDLFTGDPVLDLKRGVVTFTYAEGVVNVCSRSTTKGK




QTKVLLDAMTAPRDGKKRQIGMVAVDLGQTNPIAAEYSRVG




KNAAGTLEATPLSRSTLPDELLREIALYRKAHDRLEAQLREEA




VLKLTAEQQAENARYVETSEEGAKLALANLGVDTSTLPWDA




MTGWSTCISDHLINHGGDTSAVFFQTIRKGTKKLETIKRKDSS




WADIVRPRLTKETREALNDFLWELKRSHEGYEKLSKRLEELA




RRAVNHVVQEVKWLTQCQDIVIVIEDLNVRNFHGGGKRGGG




WSNFFTVKKENRWFMQALHKAFSDLAAHRGIPVLEVYPART




SITCLGCGHCDPENRDGEAFVCQQCGATFHADLEVATRNIAR




VALTGEAMPKAPAREQPGGAKKRGTSRRRKLTEVAVKSAEP




TIHQAKNQQLNGTSRDPVYKGSELPAL





CasΦ.43
SEQ ID
MSEITDLLKANFKGKTFKSADMRMAGRILKKSGAQAVIKYLS



NO: 257
DKGAVDPPDFRPPAKCNIIAQSRPFDEWPICKASMAIQQHIYG




LTKNEFDESSPGTSSASHEQWFAKTGVDTHGFTHVQGLNLIF




QHAKKRYEGVIKKVENYNEKERKKFEGINERRSKEGMPLLEP




RLRTAFGDDGKFAEKPGVNPSIYLYQQTSPRPYDKTKHPYVH




APFELKEITTIPTQDDRLKIPFGAPGHVPEKHRSQLSMAKHKR




RRAWYALSQNKPRPPKDGSKGRRSVRDLADLKAASLADAIPL




VSRVGFDWVVIDGRGLLRNLRWRKLAHEGMTVEEMLGFFSG




DPVIDPRRNVATFIYKAEHATVKSRKPIGGAKRAREELLKATA




SSDGVIRQVGLISVDLGQTNPVAYEISRMHQANGELVAEHLE




YGLLNDEQVNSIQRYRAAWDSMNESFRQKAIESLSMEAQDEI




MQASTGAAKRTREAVLTMFGPNATLPWSRMSSNTTCISDALI




EVGKEEETNFVTSNGPRKRTDAQWAAYLRPRVNPETRALLN




QAVWDLMKRSDEYERLSKRKLEMARQCVNFVVARAEKLTQ




CNNIGIVLENLVVRNFHGSGRRESGWEGFFEPKRENRWFMQV




LHKAFSDLAQHRGVMVFEVHPAYSSQTCPACRYVDPKNRSS




EDRERFKCLKCGRSFNADREVATFNIREIARTGVGLPKPDCER




SRGVQTTGTARNPGRSLKSNKNPSEPKRVLQSKTRKKITSTET




QNEPLATDLKT





CasΦ.44
SEQ ID
MTPKTESPLSALCKKHFPGKRFRTNYLKDAGKILKKHGEDAV



NO: 258
VAFLSDKQEDEPANFCPPAKVHILAQSRPFEDWPINLASKAIQ




TYVYGLTADERKTCEPGTSKESHDRWFKETGVDHHGFTSVQ




GLNLIFKHTLNRYDGVIKKVETRNEKRRSSVVRINEKKAAEG




LPLIAAEAEETAFGEDGRLLQPPGVNHSIYCFQQVSPQPYSSK




KHPQVVLPHAVQGVDPDAPIPVGRPNRLDIPKGQPGYVPEWQ




RPHLSMKCKRVRMWYARANWRRKPGRRSVLNEARLKEASA




KGALPIVLVIGDDWLVMDARGLLRSVFWRRVAKPGLSLSELL




NVTPTGLFSGDPVIDPKRGLVTFTSKLGVVAVHSRKPTRGKKS




KDLLLKMTKPTDDGMPRHVGMVAIDLGQTNPVAAEYSRVV




QSDAGTLKQEPVSRGVLPDDLLKDVARYRRAYDLTEESIRQE




AIALLSEGHRAEVTKLDQTTANETKRLLVDRGVSESLPWEKM




SSNTTYISDCLVALGKTDDVFFVPKAKKGKKETGIAVKRKDH




GWSKLLRPRTSPEARKALNENQWAVKRASPEYERLSRRKLEL




GRRCVNHIIQETKRWTQCEDIVVVLEDLNVGFFHGSGKRPDG




WDNFFVSKRENRWFIQVLHKAFGDLATHRGTHVIEVHPARTS




ITCIKCGHCDAGNRDGESFVCLASACGDRRHADLEVATRNVA




RVAITGERMPPSEQARDVQKAGGARKRKPSARNVKSSYPAV




EPAPASP





CasΦ.36
SEQ ID
MSDNKMKKLSKEEKPLTPLQILIRKYIDKSQYPSGFKTTIIKQA



NO: 259
GVRIKSVKSEQDEINLANWIISKYDPTYIKRDFNPSAKCQIIATS




RSVADFDIVKMSNKVQEIFFASSHLDKNVFDIGKSKSDHDSW




FERNNVDRGIYTYSNVQGMNLIFSNTKNTYLGVAVKAQNKFS




SKMKRIQDINNFRITNHQSPLPIPDEIKIYDDAGFLLNPPGVNP




NIFGYQSCLLKPLENKEIISKTSFPEYSRLPADMIEVNYKISNRL




KFSNDQKGFIQFKDKLNLFKINSQELFSKRRRLSGQPILLVASF




GDDWVVLDGRGLLRQVYYRGIAKPGSITISELLGFFTGDPIVD




PIRGVVSLGFKPGVLSQETLKTTSARIFAEKLPNLVLNNNVGL




MSIDLGQTNPVSYRLSEITSNMSVEHICSDFLSQDQISSIEKAKT




SLDNLEEEIAIKAVDHLSDEDKINFANFSKLNLPEDTRQSLFEK




YPELIGSKLDFGSMGSGTSYIADELIKFENKDAFYPSGKKKFD




LSFSRDLRKKLSDETRKSYNDALFLEKRINDKYLKNAKRRKQ




IVRTVANSLVSKIEELGLTPVINIENLAMSGGFFDGRGKREKG




WDNFFKVKKENRWVMKDFHKAFSELSPHHGVIVIESPPYCTS




VTCTKCNFCDKKNRNGHKFTCQRCGLDANADLDIATENLEK




VAISGKRMPGSERSSDERKVAVARKAKSPKGKAIKGVKCTIT




DEPALLSANSQDCSQSTS





CasΦ.37
SEQ ID
MALSLAEVRERHFKGLRFRSSYLKRAGKILKKEGEAACVAYL



NO: 260
TGKDEESPPNFKPPAKCDVVAQSRPFEEWPIVQASVAVQSYV




YGLTKEAFEAFNPGTTKQSHEACLAATGIDTCGYSNVQGLNL




IFRQAKNRYEGVITKVENRNKKAKKKLTRKNEWRQKNGHSE




LPEAPEELTFNDEGRLLQPPGINPSLYTYQQISPTPWSPKDSSIL




PPQYAGYERDPNAPIPFGVAKDRLTIASGCPGYIPEWMRTAGE




KTNPRTQKKFMHPGLSTRKNKRMRLPRSVRSAPLGALLVTIH




LGEDWLVLDVRGLLRNARWRGVAPKDISTQGLLNLFTGDPVI




DTRRGVVTFTYKPETVGIHSRTWLYKGKQTKEVLEKLTQDQT




VALVAIDLGQTNPVSAAASRVSRSGENLSIETVDRFFLPDELIK




ELRLYRMAHDRLEERIREESTLALTEAQQAEVRALEHVVRDD




AKNKVCAAFNLDAASLPWDQMTSNTTYLSEAILAQGVSRDQ




VFFTPNPKKGSKEPVEVMRKDRAWVYAFKAKLSEETRKAKN




EALWALKRASPDYARLSKRREELCRRSVNMVINRAKKRTQC




QVVIPVLEDLNIGFFHGSGKRLPGWDNFFVAKKENRWLMNG




LHKSFSDLAVHRGFYVFEVMPHRTSITCPACGHCDSENRDGE




AFVCLSCKRTYHADLDVATHNLTQVAGTGLPMPEREHPGGT




KKPGGSRKPESPQTHAPILHRTDYSESADRLGS





CasΦ.45
SEQ ID
QAVIKYLSDKGAVDPPDFRPPAKCNIIAQSRPFDEWPICKASM



NO: 261
AIQQHIYGLTKNEFDESSPGTSSASHEQWFAKTGVDTHGFTH




VQGLNLIFQHAKKRYEGVIKKVENYNEKERKKFEGINERRSK




EGMPLLEPRLRTAFGDDGKFAEKPGVNPSIYLYQQTSPRPYD




KTKHPYVHAPFELKEITTIPTQDDRLKIPFGAPGHVPEKHRSQL




SMAKHKRRRAWYALSQNKPRPPKDGSKGRRSVRDLADLKA




ASLADAIPLVSRVGFDWVVIDGRGLLRNLRWRKLAHEGMTV




EEMLGFFSGDPVIDPRRNVATFIYKAEHATVKSRKPIGGAKRA




REELLKATASSDGVIRQVGLISVDLGQTNPVAYEISRMHQAN




GELVAEHLEYGLLNDEQVNSIQRYRAAWDSMNESFRQKAIES




LSMEAQDEIMQASTGAAKRTREAVLTMFGPNATLPWSRMSS




NTTCISDALIEVGKEEETNFVTSNGPRKRTDAQWAAYLRPRV




NPETRALLNQAVWDLMKRSDEYERLSKRKLEMARQCVNFV




VARAEKLTQCNNIGIVLENLVVRNFHGSGRRESGWEGFFEPK




RENRWFMQVLHKAFSDLAQHRGVMVFEVHPAYSSQTCPACR




YVDPKNRSSEDRERFKCLKCGRSFNADREVATFNIREIARTGV




GLPKPDCERSRDVQTPGTARKSGRSLKSQDNLSEPKRVLQSK




TRKKITSTETQNEPLATDLKT





CasΦ.38
SEQ ID
MIKEQSELSKLIEKYYPGKKFYSNDLKQAGKHLKKSEHLTAK



NO: 262
ESEELTVEFLKSCKEKLYDFRPPAKALIISTSRPFEEWPIYKASE




SIQKYIYSLTKEELEKYNISTDKTSQENFFKESLIDNYGFANVS




GLNLIFQHTKAIYDGVLKKVNNRNNKILKKYKRKIEEGIEIDSP




ELEKAIDESGHFINPPGINKNIYCYQQVSPTIFNSFKETKIICPFN




YKRNPNDIIQKGVIDRLAIPFGEPGYIPDHQRDKVNKHKKRIR




KYYKNNENKNKDAILAKINIGEDWVLFDLRGLLRNAYWRKL




IPKQGITPQQLLDMFSGDPVIDPIKNNITFIYKESIIPIHSESIIKTK




KSKELLEKLTKDEQIALVSIDLGQTNPVAARFSRLSSDLKPEH




VSSSFLPDELKNEICRYREKSDLLEIEIKNKAIKMLSQEQQDEI




KLVNDISSEELKNSVCKKYNIDNSKIPWDKMNGFTTFIADEFI




NNGGDKSLVYFTAKDKKSKKEKLVKLSDKKIANSFKPKISKE




TREILNKITWDEKISSNEYKKLSKRKLEFARRATNYLINQAKK




ATRLNNVVLVVEDLNSKFFHGSGKREDGWDNFFIPKKENRW




FIQALHKSLTDVSIHRGINVIEVRPERTSITCPKCGCCDKENRK




GEDFKCIKCDSVYHADLEVATFNIEKVAITGESMPKPDCERLG




GEESIG





CasΦ.39
SEQ ID
VAFLDGKEVDEPYTLQPPAKCHILAVSRPIEEWPIARVTMAVQ



NO: 263
EHVYALPVHEVEKSRPETTEGSRSAWFKNSGVSNHGVTHAQ




TLNAILKNAYNVYNGVIKKVENRNAKKRDSLAAKNKSRERK




GLPHFKADPPELATDEQGYLLQPPSPNSSVYLVQQHLRTPQID




LPSGYTGPVVDPRSPIPSLIPIDRLAIPPGQPGYVPLHDREKLTS




NKHRRMKLPKSLRAQGALPVCFRVFDDWAVVDGRGLLRHA




QYRRLAPKNVSIAELLELYTGDPVIDIKRNLMTFRFAEAVVEV




TARKIVEKYHNKYLLKLTEPKGKPVREIGLVSIDLNVQRLIAL




AIYRVHQTGESQLALSPCLHREILPAKGLGDFDKYKSKFNQLT




EEILTAAVQTLTSAQQEEYQRYVEESSHEAKADLCLKYSITPH




ELAWDKMTSSTQYISRWLRDHGWNASDFTQITKGRKKVERL




WSDSRWAQELKPKLSNETRRKLEDAKHDLQRANPEWQRLA




KRKQEYSRHLANTVLSMAREYTACETVVIAIENLPMKGGFVD




GNGSRESGWDNFFTHKKENRWMIKDIHKALSDLAPNRGVHV




LEVNPQYTSQTCPECGHRDKANRDPIQRERFCCTHCGAQRHA




DLEVATHNIAMVATTGKSLTGKSLAPQRLQ





CasΦ.42
SEQ ID
LEIPEGEPGHVPWFQRMDIPEGQIGHVNKIQRFNFVHGKNSGK



NO: 264
VKFSDKTGRVKRYHHSKYKDATKPYKFLEESKKVSALDSILA




IITIGDDWVVFDIRGLYRNVFYRELAQKGLTAVQLLDLFTGDP




VIDPKKGIITFSYKEGVVPVFSQKIVSRFKSRDTLEKLTSQGPV




ALLSVDLGQNEPVAARVCSLKNINDKIALDNSCRIPFLDDYKK




QIKDYRDSLDELEIKIRLEAINSLDVNQQVEIRDLDVFSADRAK




ASTVDMFDIDPNLISWDSMSDARFSTQISDLYLKNGGDESRV




YFEINNKRIKRSDYNISQLVRPKLSDSTRKNLNDSIWKLKRTSE




EYLKLSKRKLELSRAVVNYTIRQSKLLSGINDIVIILEDLDVKK




KFNGRGIRDIGWDNFFSSRKENRWFIPAFHKSFSELSSNRGLC




VIEVNPAWTSATCPDCGFCSKENRDGINFTCRKCGVSYHADI




DVATLNIARVAVLGKPMSGPADRERLGGTKKPRVARSRKDM




KRKDISNGTVEVMVTA





CasΦ.46
SEQ ID
IPSFGYLDRLKIAKGQPGYIPEWQRETINPSKKVRRYWATNHE



NO: 265
KIRNAIPLVVFIGDDWVIIDGRGLLRDARRRKLADKNTTIEQL




LEMVSNDPVIDSTRGIATLSYVEGVVPVRSFIPIGEKKGREYLE




KSTQKESVTLLSVDIGQINPVSCGVYKVSNGCSKIDFLDKFFL




DKKHLDAIQKYRTLQDSLEASIVNEALDEIDPSFKKEYQNINS




QTSNDVKKSLCTEYNIDPEAISWQDITAHSTLISDYLIDNNITN




DVYRTVNKAKYKTNDFGWYKKFSAKLSKEAREALNEKIWEL




KIASSKYKKLSVRKKEIARTIANDCVKRAETYGDNVVVAMES




LTKNNKVMSGRGKRDPGWHNLGQAKVENRWFIQAISSAFED




KATHHGTPVLKVNPAYTSQTCPSCGHCSKDNRSSKDRTIFVC




KSCGEKFNADLDVATYNIAHVAFSGKKLSPPSEKSSATKKPRS




ARKSKKSRKS





CasΦ.47
SEQ ID
SPIEKLLNGLLVKITFGNDWIICDARGLLDNVQKGIIHKSYFTN



NO: 266
KSSLVDLIDLFTCNPIVNYKNNVVTFCYKEGVVDVKSFTPIKS




GPKTQENLIKKLKYSRFQNEKDACVLGVGVDVGVTNPFAING




FKMPVDESSEWVMLNEPLFTIETSQAFREEIMAYQQRTDEMN




DQFNQQSIDLLPPEYKVEFDNLPEDINEVAKYNLLHTLNIPNN




FLWDKMSNTTQFISDYLIQIGRGTETEKTITTKKGKEKILTIRD




VNWFNTFKPKISEETGKARTEIKRDLQKNSDQFQKLAKSREQ




SCRTWVNNVTEEAKIKSGCPLIIFVIEALVKDNRVFSGKGHRA




IGWHNFGKQKNERRWWVQAIHKAFQEQGVNHGYPVILCPPQ




YTSQTCPKCNHVDRDNRSGEKFKCLKYGWIGNADLDVGAYN




IARVAITGKALSKPLEQKKIKKAKNKT





CasΦ.48
SEQ ID
LLDNVQKGIIHKSYFTNKSSLVDLIDLFTCNPIVNYKNNVVTF



NO: 267
CYKEGVVDVKSFTPIKSGPKTQENLIKKLKYSRFQNEKDACV




LGVGVDVGVTNPFAINGFKMPVDESSEWVMLNEPLFTIETSQ




AFREEIMAYQQRTDEMNDQFNQQSIDLLPPEYKVEFDNLPEDI




NEVAKYNLLHTLNIPNNFLWDKMSNTTQFISDYLIQIGRGTET




EKTITTKKGKEKILTIRDVNWFNTFKPKISEETGKARTEIKRDL




QKNSDQFQKLAKSREQSCRTWVNNVTEEAKIKSGCPLIIFVIE




ALVKDNRVFSGKGHRAIGWHNFGKQKNERRWWVQAIHKAF




QEQGVNHGYPVILCPPQYTSQTCPKCNHVDRDNRSGEKFKCL




KYGWIGNADLDVGAYNIARVAITGKALSKPLEQKKIKKAKN




KT





CasΦ.49
SEQ ID
MIKPTVSQFLTPGFKLIRNHSRTAGLKLKNEGEEACKKFVREN



NO: 268
EIPKDECPNFQGGPAIANIIAKSREFTEWEIYQSSLAIQEVIFTLP




KDKLPEPILKEEWRAQWLSEHGLDTVPYKEAAGLNLIIKNAV




NTYKGVQVKVDNKNKNNLAKINRKNEIAKLNGEQEISFEEIK




AFDDKGYLLQKPSPNKSIYCYQSVSPKPFITSKYHNVNLPEEYI




GYYRKSNEPIVSPYQFDRLRIPIGEPGYVPKWQYTFLSKKENK




RRKLSKRIKNVSPILGIICIKKDWCVFDMRGLLRTNHWKKYH




KPTDSINDLFDYFTGDPVIDTKANVVRFRYKMENGIVNYKPV




REKKGKELLENICDQNGSCKLATVDVGQNNPVAIGLFELKKV




NGELTKTLISRHPTPIDFCNKITAYRERYDKLESSIKLDAIKQLT




SEQKIEVDNYNNNFTPQNTKQIVCSKLNINPNDLPWDKMISGT




HFISEKAQVSNKSEIYFTSTDKGKTKDVMKSDYKWFQDYKPK




LSKEVRDALSDIEWRLRRESLEFNKLSKSREQDARQLANWISS




MCDVIGIENLVKKNNFFGGSGKREPGWDNFYKPKKENRWWI




NAIHKALTELSQNKGKRVILLPAMRTSITCPKCKYCDSKNRN




GEKFNCLKCGIELNADIDVATENLATVAITAQSMPKPTCERSG




DAKKPVRARKAKAPEFHDKLAPSYTVVLREAVKRPAATKK





AGQAKKKKEF





(Bold sequence is Nuclear Localization Signal)









In some embodiments, any of the programmable CasΦ nuclease of the present disclosure (e.g., any one of SEQ ID NO: 221-SEQ ID NO: 268 or fragments or variants thereof) may include a nuclear localization signal (NLS). In some cases, said NLS may have a sequence of KRPAATKKAGQAKKKKEF (SEQ ID NO: 269).


A CasΦ polypeptide or a variant thereof can comprise at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, at least 99%, or 100% sequence identity with any one of SEQ ID NO: 221-SEQ ID NO: 268.


In some embodiments, the Type VI CRISPR/Cas enzyme is a programmable Cas13 nuclease. The general architecture of a Cas13 protein includes an N-terminal domain and two HEPN (higher eukaryotes and prokaryotes nucleotide-binding) domains separated by two helical domains (Liu et al., Cell 2017 Jan. 12; 168 (1-2):121-134.el2). The HEPN domains each comprise aR-X4—H motif. Shared features across Cas13 proteins include that upon binding of the crRNA of the guide nucleic acid to a target nucleic acid, the protein undergoes a conformational change to bring together the HEPN domains and form a catalytically active RNase. (Tambe et al., Cell Rep. 2018 Jul. 24; 24(4): 1025-1036). Thus, two activatable HEPN domains are characteristic of a programmable Cas13 nuclease of the present disclosure. However, programmable Cas13 nucleases also consistent with the present disclosure include Cas13 nucleases comprising mutations in the HEPN domain that enhance the Cas13 proteins cleavage efficiency or mutations that catalytically inactivate the HEPN domains. Programmable Cas13 nucleases consistent with the present disclosure also Cas13 nucleases comprising catalytic


A programmable Cas13 nuclease can be a Cas13a protein (also referred to as “c2c2”), a Cas13b protein, a Cas13c protein, a Cas13d protein, or a Cas13e protein. Example C2c2 proteins are set forth as SEQ ID NO: 153-SEQ ID NO: 160. In some cases, a subject C2c2 protein includes an amino acid sequence having 80% or more (e.g., 85% or more, 90% or more, 95% or more, 98% or more, 99% or more, 99.5% or more, or 100%) amino acid sequence identity with the amino acid sequence set forth in any one of SEQ ID NO: 153-SEQ ID NO: 160. In some cases, a suitable C2c2 polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the Listeria seeligeri C2c2 amino acid sequence set forth in SEQ ID NO: 153. In some cases, a suitable C2c2 polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the Leptotrichia buccalis C2c2 amino acid sequence set forth in SEQ ID NO: 154. In some cases, a suitable C2c2 polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the Rhodobacter capsulatus C2c2 amino acid sequence set forth in SEQ ID NO: 156. In some cases, a suitable C2c2 polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the Carnobacterium gallinarum C2c2 amino acid sequence set forth in SEQ ID NO: 157. In some cases, a suitable C2c2 polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the Herbinix hemicellulosilytica C2c2 amino acid sequence set forth in SEQ ID NO: 158. In some cases, the C2c2 protein includes an amino acid sequence having 80% or more amino acid sequence identity with the Leptotrichia buccalis (Lbu) C2c2 amino acid sequence set forth in SEQ ID NO: 154. In some cases, the C2c2 protein is a Leptotrichia buccalis (Lbu) C2c2 protein (e.g., see SEQ ID NO: 154). In some cases, the C2c2 protein includes the amino acid sequence set forth in any one of SEQ ID NO: 153, SEQ ID NO: 154 and SEQ ID NO: 156-SEQ ID NO: 160. In some cases, a C2c2 protein used in a method of the present disclosure is not a Leptotrichia shahii (Lsh) C2c2 protein. In some cases, a C2c2 protein used in a method of the present disclosure is not a C2c2 polypeptide having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the Lsh C2c2 polypeptide set forth in SEQ ID NO: 155. Other Cas13 protein sequences are set forth in SEQ ID NO: 153-SEQ ID NO: 170.









TABLE 4







Cas13 Protein Sequences










SEQ





ID





NO

Description
Sequence





SEQ


Listeria

MWISIKTLIHHLGVLFFCDYMYNRREKKIIEVKTMRITKVEVDRKKVLIS


ID


seeligeri

RDKNGGKLVYENEMQDNTEQIMHHKKSSFYKSVVNKTICRPEQKQMK


NO:

C2c2 amino
KLVHGLLQENSQEKIKVSDVTKLNISNFLNHRFKKSLYYFPENSPDKSEE


153

acid
YRIEINLSQLLEDSLKKQQGTFICWESFSKDMELYINWAENYISSKTKLI




sequence
KKSIRNNRIQSTESRSGQLMDRYMKDILNKNKPFDIQSVSEKYQLEKLT





SALKATFKEAKKNDKEINYKLKSTLQNHERQIIEELKENSELNQFNIEIR





KHLETYFPIKKTNRKVGDIRNLEIGEIQKIVNHRLKNKIVQRILQEGKLA





SYEIESTVNSNSLQKIKIEEAFALKFINACLFASNNLRNMVYPVCKKDIL





MIGEFKNSFKEIKHKKFIRQWSQFFSQEITVDDIELASWGLRGAIAPIRNE





IIHLKKHSWKKFFNNPTFKVKKSKIINGKTKDVTSEFLYKETLFKDYFYS





ELDSVPELIINKMESSKILDYYSSDQLNQVFTIPNFELSLLTSAVPFAPSF





KRVYLKGFDYQNQDEAQPDYNLKLNIYNEKAFNSEAFQAQYSLFKMVY





YQVFLPQFTTNNDLFKSSVDFILTLNKERKGYAKAFQDIRKMNKDEKPS





EYMSYIQSQLMLYQKKQEEKEKINHFEKFINQVFIKGFNSFIEKNRLTYI





CHPTKNTVPENDNIEIPFHTDMDDSNIAFWLMCKLLDAKQLSELRNEMI





KFSCSLQSTEEISTFTKAREVIGLALLNGEKGCNDWKELFDDKEAWKK





NMSLYVSEELLQSLPYTQEDGQTPVINRSIDLVKKYGTETILEKLFSSSD





DYKVSAKDIAKLHEYDVTEKIAQQESLHKQWIEKPGLARDSAWTKKY





QNVINDISNYQWAKTKVELTQVRHLHQLTIDLLSRLAGYMSIADRDFQF





SSNYILERENSEYRVTSWILLSENKNKNKYNDYELYNLKNASIKVSSKN





DPQLKVDLKQLRLTLEYLELFDNRLKEKRNNISHFNYLNGQLGNSILEL





FDDARDVLSYDRKLKNAVSKSLKEILSSHGMEVTFKPLYQTNHHLKID





KLQPKKIHHLGEKSTVSSNQVSNEYCQLVRTLLTMK





SEQ


Leptotrichia

MKVTKVGGISHKKYTSEGRLVKSESEENRTDERLSALLNMRLDMYIKN


ID


buccalis

PSSTETKENQKRIGKLKKFFSNKMVYLKDNTLSLKNGKKENIDREYSET


NO:

(Lbu) C2c2
DILESDVRDKKNFAVLKKIYLNENVNSEELEVFRNDIKKKLNKINSLKY


154

amino acid
SFEKNKANYQKINENNIEKVEGKSKRNIIYDYYRESAKRDAYVSNVKE




sequence
AFDKLYKEEDIAKLVLEIENLTKLEKYKIREFYHEIIGRKNDKENFAKIIY





EEIQNVNNMKELIEKVPDMSELKKSQVFYKYYLDKEELNDKNIKYAFC





HFVEIEMSQLLKNYVYKRLSNISNDKIKRIFEYQNLKKLIENKLLNKLDT





YVRNCGKYNYYLQDGEIATSDFIARNRQNEAFLRNIIGVSSVAYFSLRNI





LETENENDITGRMRGKTVKNNKGEEKYVSGEVDKIYNENKKNEVKEN





LKMFYSYDFNMDNKNEIEDFFANIDEAISSIRHGIVHFNLELEGKDIFAF





KNIAPSEISKKMFQNEINEKKLKLKIFRQLNSANVFRYLEKYKILNYLKR





TRFEFVNKNIPFVPSFTKLYSRIDDLKNSLGIYWKTPKTNDDNKTKEIID





AQIYLLKNIYYGEFLNYFMSNNGNFFEISKEIIELNKNDKRNLKTGFYKL





QKFEDIQEKIPKEYLANIQSLYMINAGNQDEEEKDTYIDFIQKIFLKGFM





TYLANNGRLSLIYIGSDEETNTSLAEKKQEFDKFLKKYEQNNNIKIPYEI





NEFLREIKLGNILKYTERLNMFYLILKLLNHKELTNLKGSLEKYQSANK





EEAFSDQLELINLLNLDNNRVTEDFELEADEIGKFLDFNGNKVKDNKEL





KKFDTNKIYFDGENIIKHRAFYNIKKYGMLNLLEKIADKAGYKISIEELK





KYSNKKNEIEKNHKMQENLHRKYARPRKDEKFTDEDYESYKQAIENIE





EYTHLKNKVEFNELNLLQGLLLRILHRLVGYTSIWERDLRFRLKGEFPE





NQYIEEIFNFENKKNVKYKGGQIVEKYIKFYKELHQNDEVKINKYSSAN





IKVLKQEKKDLYIRNYIAHFNYIPHAEISLLEVLENLRKLLSYDRKLKNA





VMKSVVDILKEYGFVATFKIGADKKIGIQTLESEKIVHLKNLKKKKLMT





DRNSEELCKLVKIMFEYKMEEKKSEN





SEQ


Leptotrichia

MGNLFGHKRWYEVRDKKDFKIKRKVKVKRNYDGNKYILNINENNNKEK


ID


shahii (Lsh)

IDNNKFIRKYINYKKNDNILKEFTRKFHAGNILFKLKGKEGIIRIENNDDF


NO:

C2c2
LETEEVVLYIEAYGKSEKLKALGITKKKIIDEAIRQGITKDDKKIEIKRQE


155

protein
NEEEIEIDIRDEYTNKTLNDCSIILRIIENDELETKKSIYEIFKNINMSLY





KIIEKIIENETEKVFENRYYEEHLREKLLKDDKIDVILTNFMEIREKIKSN





LEILGFVKFYLNVGGDKKKSKNKKMLVEKILNINVDLTVEDIADFVIKELE





FWNITKRIEKVKKVNNEFLEKRRNRTYIKSYVLLDKHEKFKIERENKKD





KIVKFFVENIKNNSIKEKIEKILAEFKIDELIKKLEKELKKGNCDTEIFGI





FKKHYKVNFDSKKFSKKSDEEKELYKIIYRYLKGRIEKILVNEQKVRLKK





MEKIEIEKILNESILSEKILKRVKQYTLEHIMYLGKLRHNDIDMTTVNTD





DFSRLHAKEELDLELITFFASTNMELNKIFSRENINNDENIDFFGGDREK





NYVLDKKILNSKIKIIRDLDFIDNKNNITNNFIRKFTKIGTNERNRILHAI





SKERDLQGTQDDYNKVINIIQNLKISDEEVSKALNLDVVFKDKKNIITKIN





DIKISEENNNDIKYLPSFSKVLPEILNLYRNNPKNEPFDTIETEKIVLNAL





IYVNKELYKKLILEDDLEENESKNIFLQELKKTLGNIDEIDENIIENYYKN





AQISASKGNNKAIKKYQKKVIECYIGYLRKNYEELFDFSDFKMNIQEIK





KQIKDINDNKTYERITVKTSDKTIVINDDFEYIISIFALLNSNAVINKIRN





RFFATSVWLNTSEYQNIIDILDEIMQLNTLRNECITENWNLNLEEFIQKMK





EIEKDFDDFKIQTKKEIFNNYYEDIKNNILTEFKDDINGCDVLEKKLEKIV





IFDDETKFEIDKKSNILQDEQRKLSNINKKDLKKKVDQYIKDKDQEIKSK





ILCRIIFNSDFLKKYKKEIDNLIEDMESENENKFQEIYYPKERKNELYIYK





KNLFLNIGNPNFDKIYGLISNDIKMADAKFLFNIDGKNIRKNKISEIDAIL





KNLNDKLNGYSKEYKEKYIKKLKENDDFFAKNIQNKNYKSFEKDYNR





VSEYKKIRDLVEFNYLNKIESYLIDINWKLAIQMARFERDMHYIVNGLR





ELGIIKLSGYNTGISRAYPKRNGSDGFYTTTAYYKFFDEESYKKFEKICY





GFGIDLSENSEINKPENESIRNYISHFYIVRNPFADYSIAEQIDRVSNLLS





YSTRYNNSTYASVFEVFKKDVNLDYDELKKKFKLIGNNDILERLMKPKK





VSVLELESYNSDYIKNLIIELLTKIENTNDTL





SEQ


Rhodobacter

MQIGKVQGRTISEFGDPAGGLKRKISTDGKNRKELPAHLSSDPKALIGQ


ID


capsulatus

WISGIDKIYRKPDSRKSDGKAIHSPTPSKMQFDARDDLGEAFWKLVSEA


NO:

C2c2 amino
GLAQDSDYDQFKRRLHPYGDKFQPADSGAKLKFEADPPEPQAFHGRW


156

acid
YGAMSKRGNDAKELAAALYEHLHVDEKRIDGQPKRNPKTDKFAPGLV




sequence
VARALGIESSVLPRGMARLARNWGEEEIQTYFVVDVAASVKEVAKAA





VSAAQAFDPPRQVSGRSLSPKVGFALAEHLERVTGSKRCSFDPAAGPSV





LALHDEVKKTYKRLCARGKNAARAFPADKTELLALMRHTHENRVRNQ





MVRMGRVSEYRGQQAGDLAQSHYWTSAGQTEIKESEIFVRLWVGAFA





LAGRSMKAWIDPMGKIVNTEKNDRDLTAAVNIRQVISNKEMVAEAMA





RRGIYFGETPELDRLGAEGNEGFVFALLRYLRGCRNQTFHLGARAGFLK





EIRKELEKTRWGKAKEAEHVVLTDKTVAAIRAIIDNDAKALGARLLAD





LSGAFVAHYASKEHFSTLYSEIVKAVKDAPEVSSGLPRLKLLLKRADGV





RGYVHGLRDTRKHAFATKLPPPPAPRELDDPATKARYIALLRLYDGPFR





AYASGITGTALAGPAARAKEAATALAQSVNVTKAYSDVMEGRSSRLRP





PNDGETLREYLSALTGETATEFRVQIGYESDSENARKQAEFIENYRRDM





LAFMFEDYIRAKGFDWILKIEPGATAMTRAPVLPEPIDTRGQYEHWQA





ALYLVMHFVPASDVSNLLHQLRKWEALQGKYELVQDGDATDQADAR





REALDLVKRFRDVLVLFLKTGEARFEGRAAPFDLKPFRALFANPATFDR





LFMATPTTARPAEDDPEGDGASEPELRVARTLRGLRQIARYNHMAVLS





DLFAKHKVRDEEVARLAEIEDETQEKSQIVAAQELRTDLHDKVMKCHP





KTISPEERQSYAAAIKTIEEHRFLVGRVYLGDHLRLHRLMMDVIGRLID





YAGAYERDTGTFLINASKQLGAGADWAVTIAGAANTDARTQTRKDLA





HFNVLDRADGTPDLTALVNRAREMMAYDRKRKNAVPRSILDMLARLG





LTLKWQMKDHLLQDATITQAAIKHLDKVRLTVGGPAAVTEARFSQDY





LQMVAAVFNGSVQNPKPRRRDDGDAWHKPPKPATAQSQPDQKPPNKA





PSAGSRLPPPQVGEVYEGVVVKVIDTGSLGFLAVEGVAGNIGLHISRLR





RIREDAIIVGRRYRFRVEIYVPPKSNTSKLNAADLVRID





SEQ


Carnobacterium

MRITKVKIKLDNKLYQVTMQKEEKYGTLKLNEESRKSTAEILRLKKASF


ID


gallinarum

NKSFHSKTINSQKENKNATIKKNGDYISQIFEKLVGVDTNKNIRKPKMS


NO:

C2c2 amino
LTDLKDLPKKDLALFIKRKFKNDDIVEIKNLDLISLFYNALQKVPGEHFT


157

acid
DESWADFCQEMMPYREYKNKFIERKIILLANSIEQNKGFSINPETFSKRK




sequence
RVLHQWAIEVQERGDFSILDEKLSKLAEIYNFKKMCKRVQDELNDLEK





SMKKGKNPEKEKEAYKKQKNFKIKTIWKDYPYKTHIGLIEKIKENEELN





QFNIEIGKYFEHYFPIKKERCTEDEPYYLNSETIATTVNYQLKNALISYL





MQIGKYKQFGLENQVLDSKKLQEIGIYEGFQTKFMDACVFATSSLKNII





EPMRSGDILGKREFKEAIATSSFVNYHHFFPYFPFELKGMKDRESELIPF





GEQTEAKQMQNIWALRGSVQQIRNEIFHSFDKNQKFNLPQLDKSNFEFD





ASENSTGKSQSYIETDYKFLFEAEKNQLEQFFIERIKSSGALEYYPLKSL





EKLFAKKEMKFSLGSQVVAFAPSYKKLVKKGHSYQTATEGTANYLGLS





YYNRYELKEESFQAQYYLLKLIYQYVFLPNFSQGNSPAFRETVKAILRIN





KDEARKKMKKNKKFLRKYAFEQVREMEFKETPDQYMSYLQSEMREE





KVRKAEKNDKGFEKNITMNFEKLLMQIFVKGFDVFLTTFAGKELLLSSEE





KVIKETEISLSKKINEREKTLKASIQVEHQLVATNSAISYWLFCKLLDSR





HLNELRNEMIKFKQSRIKFNHTQHAELIQNLLPIVELTILSNDYDEKNDS





QNVDVSAYFEDKSLYETAPYVQTDDRTRVSFRPILKLEKYHTKSLIEAL





LKDNPQFRVAATDIQEWMHKREEIGELVEKRKNLHTEWAEGQQTLGA





EKREEYRDYCKKIDRFNWKANKVTLTYLSQLHYLITDLLGRMVGFSAL





FERDLVYFSRSFSELGGETYHISDYKNLSGVLRLNAEVKPIKIKNIKVID





NEENPYKGNEPEVKPFLDRLHAYLENVIGIKAVHGKIRNQTAHLSVLQLE





LSMIESMNNLRDLMAYDRKLKNAVTKSMIKILDKHGMILKLKIDENHK





NFEIESLIPKEIIHLKDKAIKTNQVSEEYCQLVLALLTTNPGNQLN





SEQ


Herbinix

MKLTRRRISGNSVDQKITAAFYRDMSQGLLYYDSEDNDCTDKVIESMD


ID


hemicellulos

FERSWRGRILKNGEDDKNPFYMFVKGLVGSNDKIVCEPIDVDSDPDNL


NO:


ilytica C2c2

DILINKNLTGFGRNLKAPDSNDTLENLIRKIQAGIPEEEVLPELKKIKEMI


158

amino acid
QKDIVNRKEQLLKSIKNNRIPFSLEGSKLVPSTKKMKWLFKLIDVPNKTF




sequence
NEKMLEKYWEIYDYDKLKANITNRLDKTDKKARSISRAVSEELREYHK





NLRTNYNRFVSGDRPAAGLDNGGSAKYNPDKEEFLLFLKEVEQYFKKY





FPVKSKHSNKSKDKSLVDKYKNYCSYKVVKKEVNRSIINQLVAGLIQQ





GKLLYYFYYNDTWQEDFLNSYGLSYIQVEEAFKKSVMTSLSWGINRLT





SFFIDDSNTVKFDDITTKKAKEAIESNYFNKLRTCSRMQDHFKEKLAFFY





PVYVKDKKDRPDDDIENLIVLVKNAIESVSYLRNRTFHFKESSLLELLKE





LDDKNSGQNKIDYSVAAEFIKRDIENLYDVFREQIRSLGIAEYYKADMIS





DCFKTCGLEFALYSPKNSLMPAFKNVYKRGANLNKAYIRDKGPKETGD





QGQNSYKALEEYRELTWYIEVKNNDQSYNAYKNLLQLIYYHAFLPEVR





ENEALITDFINRTKEWNRKETEERLNTKNNKKHKNFDENDDITVNTYR





YESIPDYQGESLDDYLKVLQRKQMARAKEVNEKEEGNNNYIQFIRDVV





VWAFGAYLENKLKNYKNELQPPLSKENIGLNDTLKELFPEEKVKSPFNI





KCRFSISTFIDNKGKSTDNTSAEAVKTDGKEDEKDKKNIKRKDLLCFYL





FLRLLDENEICKLQHQFIKYRCSLKERRFPGNRTKLEKETELLAELEELM





ELVRFTMPSIPEISAKAESGYDTMIKKYFKDFIEKKVFKNPKTSNLYYHS





DSKTPVTRKYMALLMRSAPLHLYKDIFKGYYLITKKECLEYIKLSNIIKD





YQNSLNELHEQLERIKLKSEKQNGKDSLYLDKKDFYKVKEYVENLEQV





ARYKHLQHKINFESLYRIFRIHVDIAARMVGYTQDWERDMHFLFKALV





YNGVLEERRFEAIFNNNDDNNDGRIVKKIQNNLNNKNRELVSMLCWN





KKLNKNEFGAIIWKRNPIAHLNHFTQTEQNSKSSLESLINSLRILLAYDR





KRQNAVTKTINDLLLNDYHIRIKWEGRVDEGQIYFNIKEKEDIENEPIIH





LKHLHKKDCYIYKNSYMFDKQKEWICNGIKEEVYDKSILKCIGNLFKFD





YEDKNKSSANPKHT





SEQ


Paludibacter

MRVSKVKVKDGGKDKMVLVHRKTTGAQLVYSGQPVSNETSNILPEKK


ID


propionicigenes

RQSFDLSTLNKTIIKFDTAKKQKLNVDQYKIVEKIFKYPKQELPKQIKAE


NO:

C2c2
EILPFLNHKFQEPVKYWKNGKEESFNLTLLIVEAVQAQDKRKLQPYYD


159

amino acid
WKTWYIQTKSDLLKKSIENNRIDLTENLSKRKKALLAWETEFTASGSID




sequence
LTHYHKVYMTDVLCKMLQDVKPLTDDKGKINTNAYHRGLKKALQNH





QPAIFGTREVPNEANRADNQLSIYHLEVVKYLEHYFPIKTSKRRNTADDI





AHYLKAQTLKTTIEKQLVNAIRANIIQQGKTNHHELKADTTSNDLIRIKT





NEAFVLNLTGTCAFAANNIRNMVDNEQTNDILGKGDFIKSLLKDNTNS





QLYSFFFGEGLSTNKAEKETQLWGIRGAVQQIRNNVNHYKKDALKTVF





NISNFENPTITDPKQQTNYADTIYKARFINELEKIPEAFAQQLKTGGAVS





YYTIENLKSLLTTFQFSLCRSTIPFAPGFKKVFNGGINYQNAKQDESFYE





LMLEQYLRKENFAEESYNARYFMLKLIYNNLFLPGFTTDRKAFADSVG





FVQMQNKKQAEKVNPRKKEAYAFEAVRPMTAADSIADYMAYVQSEL





MQEQNKKEEKVAEETRINFEKFVLQVFIKGFDSFLRAKEFDFVQMPQPQ





LTATASNQQKADKLNQLEASITADCKLTPQYAKADDATHIAFYVFCKLL





DAAHLSNLRNELIKFRESVNEFKFHHLLEIIEICLLSADVVPTDYRDLYS





SEADCLARLRPFIEQGADITNWSDLFVQSDKHSPVIHANIELSVKYGTTK





LLEQIINKDTQFKTTEANFTAWNTAQKSIEQLIKQREDHHEQWVKAKN





ADDKEKQERKREKSNFAQKFIEKHGDDYLDICDYINTYNWLDNKMHF





VHLNRLHGLTIELLGRMAGFVALFDRDFQFFDEQQIADEFKLHGFVNLHS





IDKKLNEVPTKKIKEIYDIRNKIIQINGNKINESVRANLIQFISSKRNYY





NNAFLHVSNDEIKEKQMYDIRNHIAHFNYLTKDAADFSLIDLINELRELL





HYDRKLKNAVSKAFIDLFDKHGMILKLKLNADHKLKVESLEPKKIYHLG





SSAKDKPEYQYCTNQVMMAYCNMCRSLLEMKK





SEQ


Leptotrichia

MYMKITKIDGVSHYKKQDKGILKKKWKDLDERKQREKIEARYNKQIESK


ID


wadei (Lwa)

IYKEFFRLKNKKRIEKEEDQNIKSLYFFIKELYLNEKNEEWELKNINLEI


NO:

C2c2 amino
LDDKERVIKGYKFKEDVYFFKEGYKEYYLRILFNNLIEKVQNENREKVR


160

acid
KNKEFLDLKEIFKKYKNRKIDLLLKSINNNKINLEYKKENVNEEIYGINP




sequence
TNDREMTFYELLKEIIEKKDEQKSILEEKLDNFDITNFLENIEKIFNEET





EINIIKGKVLNELREYIKEKEENNSDNKLKQIYNLELKKYIENNFSYKKQ





KSKSKNGKNDYLYLNFLKKIMFIEEVDEKKEINKEKFKNKINSNFKNLFV





QHILDYGKLLYYKENDEYIKNTGQLETKDLEYIKTKETLIRKMAVLVSFA





ANSYYNLFGRVSGDILGTEVVKSSKTNVIKVGSHIFKEKMLNYFFDFEIF





DANKIVEILESISYSIYNVRNGVGHFNKLILGKYKKKDINTNKRIEEDLN





NNEEIKGYFIKKRGEIERKVKEKFLSNNLQYYYSKEKIENYFEVYEFEIL





KRKIPFAPNFKRIIKKGEDLFNNKNNKKYEYFKNFDKNSAEEKKEFLKT





RNFLLKELYYNNFYKEFLSKKEEFEKIVLEVKEEKKSRGNINNKKSGVS





FQSIDDYDTKINISDYIASIHKKEMERVEKYNEEKQKDTAKYIRDFVEEI





FLTGFINYLEKDKRLHFLKEEFSILCNNNNNVVDFNININEEKIKEFLKE





NDSKTLNLYLFFNMIDSKRISEFRNELVKYKQFTKKRLDEEKEFLGIKIE





LYETLIEFVILTREKLDTKKSEEIDAWLVDKLYVKDSNEYKEYEEILKLF





VDEKILSSKEAPYYATDNKTPILLSNFEKTRKYGTQSFLSEIQSNYKYSK





VEKENIEDYNKKEEIEQKKKSNIEKLQDLKVELHKKWEQNKITEKEIEKY





NNTTRKINEYNYLKNKEELQNVYLLHEMLSDLLARNVAFFNKWERDF





KFIVIAIKQFLRENDKEKVNEFLNPPDNSKGKKVYFSVSKYKNTVENID





GIHKNFMNLIFLNNKFMNRKIDKMNCAIWVYFRNYIAHFLHLHTKNEKI





SLISQMNLLIKLFSYDKKVQNHILKSTKTLLEKYNIQINFEISNDKNEVF





KYKIKNRLYSKKGKMLGKNNKFEILENEFLENVKAMLEYSE





SEQ


Bergeyella

MENKTSLGNNIYYNPFKPQDKSYFAGYFNAAMENTDSVFRELGKRLKG


ID


zoohelcum

KEYTSENFFDAIFKENISLVEYERYVKLLSDYFPMARLLDKKEVPIKERK


NO:

Cas13b
ENFKKNFKGIIKAVRDLRNFYTHKEHGEVEITDEIFGVLDEMLKSTVLT


161


VKKKKVKTDKTKEILKKSIEKQLDILCQKKLEYLRDTARKIEEKRRNQR





ERGEKELVAPFKYSDKRDDLIAAIYNDAFDVYIDKKKDSLKESSKAKY





NTKSDPQQEEGDLKIPISKNGVVFLLSLFLTKQEIHAFKSKIAGFKATVI





DEATVSEATVSHGKNSICFMATHEIFSHLAYKKLKRKVRTAEINYGEAEN





AEQLSVYAKETLMMQMLDELSKVPDVVYQNLSEDVQKTFIEDWNEYL





KENNGDVGTMEEEQVIHPVIRKRYEDKFNYFAIRFLDEFAQFPTLRFQV





HLGNYLHDSRPKENLISDRRIKEKITVFGRLSELEHKKALFIKNTETNED





REHYWEIFPNPNYDFPKENISVNDKDFPIAGSILDREKQPVAGKIGIKVK





LLNQQYVSEVDKAVKAHQLKQRKASKPSIQNIIEEIVPINESNPKEAIVF





GGQPTAYLSMNDIHSILYEFFDKWEKKKEKLEKKGEKELRKEIGKELEK





KIVGKIQAQIQQIIDKDTNAKILKPYQDGNSTAIDKEKLIKDLKQEQNIL





QKLKDEQTVREKEYNDFIAYQDKNREINKVRDRNHKQYLKDNLKRKYP





EAPARKEVLYYREKGKVAVWLANDIKRFMPTDFKNEWKGEQHSLLQK





SLAYYEQCKEELKNLLPEKVFQHLPFKLGGYFQQKYLYQFYTCYLDKR





LEYISGLVQQAENFKSENKVFKKVENECFKFLKKQNYTHKELDARVQSI





LGYPIFLERGFMDEKPTIIKGKTFKGNEALFADWFRYYKEYQNFQTFYD





TENYPLVELEKKQADRKRKTKIYQQKKNDVFTLLMAKHIFKSVFKQDS





IDQFSLEDLYQSREERLGNQERARQTGERNTNYIWNKTVDLKLCDGKIT





VENVKLKNVGDFIKYEYDQRVQAFLKYEENIEWQAFLIKESKEEENYP





YVVEREIEQYEKVRREELLKEVHLIEEYILEKVKDKEILKKGDNQNFKY





YILNGLLKQLKNEDVESYKVFNLNTEPEDVNINQLKQEATDLEQKAFV





LTYIRNKFAHNQLPKKEFWDYCQEKYGKIEKEKTYAEYFAEVFKKEKE





ALIK





SEQ


Prevotella

MEDDKKTTDSIRYELKDKHFWAAFLNLARHNVYITVNHINKILEEGEIN


ID


intermedia

RDGYETTLKNTWNEIKDINKKDRLSKLIIKHFPFLEAATYRLNPTDTTKQ


NO:

Cas13b
KEEKQAEAQSLESLRKSFFVFIYKLRDLRNHYSHYKHSKSLERPKFEEG


162


LLEKMYNIFNASIRLVKEDYQYNKDINPDEDFKHLDRTEEEFNYYFTKD





NEGNITESGLLFFVSLFLEKKDAIWMQQKLRGFKDNRENKKKMTNEVF





CRSRMLLPKLRLQSTQTQDWILLDMLNELIRCPKSLYERLREEDREKFR





VPIEIADEDYDAEQEPFKNTLVRHQDRFPYFALRYFDYNEIFTNLRFQID





LGTYHFSIYKKQIGDYKESHHLTHKLYGFERIQEFTKQNRPDEWRKFVK





TFNSFETSKEPYIPETTPHYHLENQKIGIRFRNDNDKIWPSLKTNSEKNE





KSKYKLDKSFQAEAFLSVHELLPMMFYYLLLKTENTDNDNEIETKKKENK





NDKQEKHKIEEIIENKITEIYALYDTFANGEIKSIDELEEYCKGKDIEIG





HLPKQMIAILKDEHKVMATEAERKQEEMLVDVQKSLESLDNQINEEIENV





ERKNSSLKSGKIASWLVNDMMRFQPVQKDNEGKPLNNSKANSTEYQL





LQRTLAFFGSEHERLAPYFKQTKLIESSNPHPFLKDTEWEKCNNILSFYR





SYLEAKKNFLESLKPEDWEKNQYFLKLKEPKTKPKTLVQGWKNGFNLP





RGIFTEPIRKWFMKHRENITVAELKRVGLVAKVIPLFFSEEYKDSVQPFY





NYHFNVGNINKPDEKNFLNCEERRELLRKKKDEFKKMTDKEKEENPSY





LEFKSWNKFERELRLVRNQDIVTWLLCMELFNKKKIKELNVEKIYLKNI





NTNTTKKEKNTEEKNGEEKNIKEKNNILNRIMPMRLPIKVYGRENFSKN





KKKKIRRNTFFTVYIEEKGTKLLKQGNFKALERDRRLGGLFSFVKTPSK





AESKSNTISKLRVEYELGEYQKARIEIIKDMLALEKTLIDKYNSLDTDNF





NKMLTDWLELKGEPDKASFQNDVDLLIAVRNAFSHNQYPMRNRIAFA





NINPFSLSSANTSEEKGLGIANQLKDKTHKTIEKIIEIEKPIETKE





SEQ


Prevotella

MQKQDKLFVDRKKNAIFAFPKYITIMENKEKPEPIYYELTDKHFWAAFL


ID


buccae

NLARHNVYTTINHINRRLEIAELKDDGYMMGIKGSWNEQAKKLDKKV


NO:

Cas13b
RLRDLIMKHFPFLEAAAYEMTNSKSPNNKEQREKEQSEALSLNNLKNV


163


LFIFLEKLQVLRNYYSHYKYSEESPKPIFETSLLKNMYKVFDANVRLVK





RDYMHHENIDMQRDFTHLNRKKQVGRTKNIIDSPNFHYHFADKEGNM





TIAGLLFFVSLFLDKKDAIWMQKKLKGFKDGRNLREQMTNEVFCRSRIS





LPKLKLENVQTKDWMQLDMLNELVRCPKSLYERLREKDRESFKVPFDI





FSDDYNAEEEPFKNTLVRHQDRFPYFVLRYFDLNEIFEQLRFQIDLGTYH





FSIYNKRIGDEDEVRHLTHHLYGFARIQDFAPQNQPEEWRKLVKDLDHF





ETSQEPYISKTAPHYHLENEKIGIKFCSAHNNLFPSLQTDKTCNGRSKFN





LGTQFTAEAFLSVHELLPMMFYYLLLTKDYSRKESADKVEGIIRKEISNI





YAIYDAFANNEINSIADLTRRLQNTNILQGHLPKQMISILKGRQKDMGK





EAERKIGEMIDDTQRRLDLLCKQTNQKIRIGKRNAGLLKSGKIADWLVN





DMMRFQPVQKDQNNIPINNSKANSTEYRMLQRALALFGSENFRLKAYF





NQMNLVGNDNPHPFLAETQWEHQTNILSFYRNYLEARKKYLKGLKPQ





NWKQYQHFLILKVQKTNRNTLVTGWKNSFNLPRGIFTQPIREWFEKHN





NSKRIYDQILSFDRVGFVAKAIPLYFAEEYKDNVQPFYDYPFNIGNRLKP





KKRQFLDKKERVELWQKNKELFKNYPSEKKKTDLAYLDFLSWKKFER





ELRLIKNQDIVTWLMFKELFNMATVEGLKIGEIHLRDIDTNTANEESNNI





LNRIMPMKLPVKTYETDNKGNILKERPLATFYIEETETKVLKQGNFKAL





VKDRRLNGLFSFAETTDLNLEEHPISKLSVDLELIKYQTTRISIFEMTLG





LEKKLIDKYSTLPTDSFRNMLERWLQCKANRPELKNYVNSLIAVRNAFS





HNQYPMYDATLFAEVKKFTLFPSVDTKKIELNIAPQLLEIVGKAIKEIEK





SENKN





SEQ


Porphyromonas

MNTVPASENKGQSRTVEDDPQYFGLYLNLARENLIEVESHVRIKFGKK


ID


gingivalis

KLNEESLKQSLLCDHLLSVDRWTKVYGHSRRYLPFLHYFDPDSQIEKD


NO:

Cas13b
HDSKTGVDPDSAQRLIRELYSLLDFLRNDFSHNRLDGTTFEHLEVSPDIS


164


SFITGTYSLACGRAQSRFAVFFKPDDFVLAKNRKEQLISVADGKECLTV





SGFAFFICLFLDREQASGMLSRIRGFKRTDENWARAVHETFCDLCIRHP





HDRLESSNTKEALLLDMLNELNRCPRILYDMLPEEERAQFLPALDENSM





NNLSENSLDEESRLLWDGSSDWAEALTKRIRHQDRFPYLMLRFIEEMDL





LKGIRFRVDLGEIELDSYSKKVGRNGEYDRTITDHALAFGKLSDFQNEE





EVSRMISGEASYPVRFSLFAPRYAIYDNKIGYCHTSDPVYPKSKTGEKR





ALSNPQSMGFISVHDLRKLLLMELLCEGSFSRMQSDFLRKANRILDETA





EGKLQFSALFPEMRHRFIPPQNPKSKDRREKAETTLEKYKQEIKGRKDK





LNSQLLSAFDMDQRQLPSRLLDEWMNIRPASHSVKLRTYVKQLNEDCR





LRLRKFRKDGDGKARAIPLVGEMATFLSQDIVRMIISEETKKLITSAYYN





EMQRSLAQYAGEENRRQFRAIVAELRLLDPSSGHPFLSATMETAHRYTE





GFYKCYLEKKREWLAKIFYRPEQDENTKRRISVFFVPDGEARKLLPLLIR





RRMKEQNDLQDWIRNKQAHPIDLPSHLFDSKVMELLKVKDGKKKWNE





AFKDWWSTKYPDGMQPFYGLRRELNIHGKSVSYIPSDGKKFADCYTHL





MEKTVRDKKRELRTAGKPVPPDLAADIKRSFHRAVNEREFMLRLVQED





DRLMLMAINKMMTDREEDILPGLKNIDSILDEENQFSLAVHAKVLEKE





GEGGDNSLSLVPATIEIKSKRKDWSKYIRYRYDRRVPGLMSHFPEHKAT





LDEVKTLLGEYDRCRIKIFDWAFALEGAIMSDRDLKPYLHESSSREGKS





GEHSTLVKMLVEKKGCLTPDESQYLILIRNKAAHNQFPCAAEMPLIYRD





VSAKVGSIEGSSAKDLPEGSSLVDSLWKKYEMIIRKILPILDPENRFFGKL





LNNMSQPINDL





SEQ


Bacteroides

MESIKNSQKSTGKTLQKDPPYFGLYLNMALLNVRKVENHIRKWLGDV


ID


pyogenes

ALLPEKSGFHSLLTTDNLSSAKWTRFYYKSRKFLPFLEMFDSDKKSYEN


NO:

Cas13b
RRETAECLDTIDRQKISSLLKEVYGKLQDIRNAFSHYHIDDQSVKHTALI


165


ISSEMHRFIENAYSFALQKTRARFTGVFVETDFLQAEEKGDNKKFFAIG





GNEGIKLKDNALIFLICLFLDREEAFKFLSRATGFKSTKEKGFLAVRETF





CALCCRQPHERLLSVNPREALLMDMLNELNRCPDILFEMLDEKDQKSF





LPLLGEEEQAHILENSLNDELCEAIDDPFEMIASLSKRVRYKNRFPYLML





RYIEEKNLLPFIRFRIDLGCLELASYPKKMGEENNYERSVTDHAMAFGR





LTDFHNEDAVLQQITKGITDEVRFSLYAPRYAIYNNKIGFVRTSGSDKIS





FPTLKKKGGEGHCVAYTLQNTKSFGFISIYDLRKILLLSFLDKDKAKNIV





SGLLEQCEKHWKDLSENLFDAIRTELQKEFPVPLIRYTLPRSKGGKLVSS





KLADKQEKYESEFERRKEKLTEILSEKDFDLSQIPRRMIDEWLNVLPTSR





EKKLKGYVETLKLDCRERLRVFEKREKGEHPLPPRIGEMATDLAKDIIR





MVIDQGVKQRITSAYYSEIQRCLAQYAGDDNRRHLDSIIRELRLKDTKN





GHPFLGKVLRPGLGHTEKLYQRYFEEKKEWLEATFYPAASPKRVPRFV





NPPTGKQKELPLIIRNLMKERPEWRDWKQRKNSHPIDLPSQLFENEICRL





LKDKIGKEPSGKLKWNEMFKLYWDKEFPNGMQRFYRCKRRVEVFDKV





VEYEYSEEGGNYKKYYEALIDEVVRQKISSSKEKSKLQVEDLTLSVRRV





FKRAINEKEYQLRLLCEDDRLLFMAVRDLYDWKEAQLDLDKIDNMLG





EPVSVSQVIQLEGGQPDAVIKAECKLKDVSKLMRYCYDGRVKGLMPYF





ANHEATQEQVEMELRHYEDHRRRVFNWVFALEKSVLKNEKLRRFYEE





SQGGCEHRRCIDALRKASLVSEEEYEFLVHIRNKSAHNQFPDLEIGKLPP





NVTSGFCECIWSKYKAIICRIIPFIDPERRFFGKLLEQK





SEQ

Cas13c
MTEKKSIIFKNKSSVEIVKKDIFSQTPDNMIRNYKITLKISEKNPRVVEAE


ID


IEDLMNSTILKDGRRSARREKSMTERKLIEEKVAENYSLLANCPMEEVD


NO:


SIKIYKIKRFLTYRSNMLLYFASINSFLCEGIKGKDNETEEIWHLKDNDV


166


RKEKVKENFKNKLIQSTENYNSSLKNQIEEKEKLLRKESKKGAFYRTIIK





KLQQERIKELSEKSLTEDCEKIIKLYSELRHPLMHYDYQYFENLFENKEN





SELTKNLNLDIFKSLPLVRKMKLNNKVNYLEDNDTLFVLQKTKKAKTL





YQIYDALCEQKNGFNKFINDFFVSDGEENTVFKQIINEKFQSEMEFLEKR





ISESEKKNEKLKKKFDSMKAHFHNINSEDTKEAYFWDIHSSSNYKTKYN





ERKNLVNEYTELLGSSKEKKLLREEITQINRKLLKLKQEMEEITKKNSLF





RLEYKMKIAFGFLFCEFDGNISKFKDEFDASNQEKIIQYHKNGEKYLTYF





LKEEEKEKFNLEKMQKIIQKTEEEDWLLPETKNNLFKFYLLTYLLLPYE





LKGDFLGFVKKHYYDIKNVDFMDENQNNIQVSQTVEKQEDYFYHKIRL





FEKNTKKYEIVKYSIVPNEKLKQYFEDLGIDIKYLTGSVESGEKWLGEN





LGIDIKYLTVEQKSEVSEEKIKKFL





SEQ

Cas13c
MEKDKKGEKIDISQEMIEEDLRKILILFSRLRHSMVHYDYEFYQALYSG


ID


KDFVISDKNNLENRMISQLLDLNIFKELSKVKLIKDKAISNYLDKNTTIH


NO:


VLGQDIKAIRLLDIYRDICGSKNGFNKFINTMITISGEEDREYKEKVIEH


167


FNKKMENLSTYLEKLEKQDNAKRNNKRVYNLLKQKLIEQQKLKEWFGG





PYVYDIHSSKRYKELYIERKKLVDRHSKLFEEGLDEKNKKELTKINDEL





SKLNSEMKEMTKLNSKYRLQYKLQLAFGFILEEFDLNIDTFINNFDKDK





DLIISNFMKKRDIYLNRVLDRGDNRLKNIIKEYKFRDTEDIFCNDRDNNL





VKLYILMYILLPVEIRGDFLGFVKKNYYDMKHVDFIDKKDKEDKDTFF





HDLRLFEKNIRKLEITDYSLSSGFLSKEHKVDIEKKINDFINRNGAMKLP





EDITIEEFNKSLILPIMKNYQINFKLLNDIEISALFKIAKDRSITFKQAI





DEIKNEDIKKNSKKNDKNNHKDKNINFTQLMKRALHEKIPYKAGMYQIRN





NISHIDMEQLYIDPLNSYMNSNKNNITISEQIEKIIDVCVTGGVTGKELN





NNIINDYYMKKEKLVFNLKLRKQNDIVSIESQEKNKREEFVFKKYGLDYK





DGEINIIEVIQKVNSLQEELRNIKETSKEKLKNKETLFRDISLINGTIRK





NINFKIKEMVLDIVRMDEIRHINIHIYYKGENYTRSNIIKFKYAIDGENK





KYYLKQHEINDINLELKDKFVTLICNMDKHPNKNKQTINLESNYIQNVKF





IIP





SEQ

Cas13c
MENKGNNKKIDFDENYNILVAQIKEYFTKEIENYNNRIDNIIDKKELLKY


ID


SEKKEESEKNKKLEELNKLKSQKLKILTDEEIKADVIKIIKIFSDLRHSL


NO:


MHYEYKYFENLFENKKNEELAELLNLNLFKNLTLLRQMKIENKTNYLEG


168


REEFNIIGKNIKAKEVLGHYNLLAEQKNGFNNFINSFFVQDGTENLEFKK





LIDEHFVNAKKRLERNIKKSKKLEKELEKMEQHYQRLNCAYVWDIHTS





TTYKKLYNKRKSLIEEYNKQINEIKDKEVITAINVELLRIKKEMEEITKS





NSLFRLKYKMQIAYAFLEIEFGGNIAKFKDEFDCSKMEEVQKYLKKGVK





YLKYYKDKEAQKNYEFPFEEIFENKDTHNEEWLENTSENNLFKFYILTY





LLLPMEFKGDFLGVVKKHYYDIKNVDFTDESEKELSQVQLDKMIGDSFFH





KIRLFEKNTKRYEIIKYSILTSDEIKRYFRLLELDVPYFEYEKGTDEIGI





FNKNIILTIFKYYQIIFRLYNDLEIHGLFNISSDLDKILRDLKSYGNKNI





NFREFLYVIKQNNNSSTEEEYRKIWENLEAKYLRLHLLTPEKEEIKTKTK





EELEKLNEISNLRNGICHLNYKEIIEEILKTEISEKNKEATLNEKIRKVI





NFIKENELDKVELGFNFINDFFMKKEQFMFGQIKQVKEGNSDSITTERER





KEKNNKKLKETYELNCDNLSEFYETSNNLRERANSSSLLEDSAFLKKIGL





YKVKNNKVNSKVKDEEKRIENIKRKLLKDSSDIMGMYKAEVVKKLKEKLI





LIFKHDEEKRIYVTVYDTSKAVPENISKEILVKRNNSKEEYFFEDNNKKY





VTEYYTLEITETNELKVIPAKKLEGKEFKTEKNKENKLMLNNHYCFNVKI





IY





SEQ

Cas13c
MEEIKHKKNKSSIIRVIVSNYDMTGIKEIKVLYQKQGGVDTFNLKTIINL


ID


ESGNLEIISCKPKEREKYRYEFNCKTEINTISITKKDKVLKKEIRKYSLE


NO:


LYFKNEKKDTVVAKVTDLLKAPDKIEGERNHLRKLSSSTERKLLSKTLCK


169


NYSEISKTPIEEIDSIKIYKIKRFLNYRSNFLIYFALINDFLCAGVKEDD





INEVWLIQDKEHTAFLENRIEKITDYIFDKLSKDIENKKNQFEKRIKKYK





TSLEELKTETLEKNKTFYIDSIKTKITNLENKITELSLYNSKESLKEDLI





KIISIFTNLRHSLMHYDYKSFENLFENIENEELKNLLDLNLFKSIRMSDE





FKTKNRTNYLDGTESFTIVKKHQNLKKLYTYYNNLCDKKNGFNTFINSFF





VTDGIENTDFKNLIILHFEKEMEEYKKSIEYYKIKISNEKNKSKKEKLKE





KIDLLQSELINMREHKNLLKQIYFFDIHNSIKYKELYSERKNLIEQYNLQ





INGVKDVTAINHINTKLLSLKNKMDKITKQNSLYRLKYKLKIAYSFLMIE





FDGDVSKFKNNFDPTNLEKRVEYLDKKEEYLNYTAPKNKFNFAKLEEELQ





KIQSTSEMGADYLNVSPENNLFKFYILTYIMLPVEFKGDFLGFVKNHYYN





IKNVDFMDESLLDENEVDSNKLNEKIENLKDSSFFNKIRLFEKNIKKYEI





VKYSVSTQENMKEYFKQLNLDIPYLDYKSTDEIGIFNKNMILPIFKYYQN





VFKLCNDIEIHALLALANKKQQNLEYAIYCCSKKNSLNYNELLKTFNRK





TYQNLSFIRNKIAHLNYKELFSDLFNNELDLNTKVRCLIEFSQNNKFDQI





DLGMNFINDYYMKKTRFIFNQRRLRDLNVPSKEKIIDGKRKQQNDSNN





ELLKKYGLSRTNIKDIFNKAWY





SEQ

Cas13c
MKVRYRKQAQLDTFIIKTEIVNNDIFIKSIIEKAREKYRYSFLFDGEEKY


ID


HFKNKSSVEIVKNDIFSQTPDNMIRNYKITLKISEKNPRVVEAEIEDLMN


NO:


STILKDGRRSARREKSMTERKLIEEKVAENYSLLANCPIEEVDSIKIYKI


170


KRFLTYRSNMLLYFASINSFLCEGIKGKDNETEEIWHLKDNDVRKEKVKE





NFKNKLIQSTENYNSSLKNQIEEKEKLSSKEFKKGAFYRTIIKKLQQERI





KELSEKSLTEDCEKIIKLYSELRHPLMHYDYQYFENLFENKENSELTKNL





NLDIFKSLPLVRKMKLNNKVNYLEDNDTLFVLQKTKKAKTLYQIYDALCE





QKNGFNKFINDFFVSDGEENTVFKQIINEKFQSEMEFLEKRISESEKKNE





KLKKKLDSMKAHFRNINSEDTKEAYFWDIHSSRNYKTKYNERKNLVNE





YTKLLGSSKEKKLLREEITKINRQLLKLKQEMEEITKKNSLFRLEYKMKI





AFGFLFCEFDGNISKFKDEFDASNQEKIIQYHKNGEKYLTSFLKEEEKEK





FNLEKMQKIIQKTEEEDWLLPETKNNLFKFYLLTYLLLPYELKGDFLGF





VKKHYYDIKNVDFMDENQNNIQVSQTVEKQEDYFYHKIRLFEKNTKK





YEIVKYSIVPNEKLKQYFEDLGIDIKYLTGSVESGEKWLGENLGIDIKYL





TVEQKSEVSEEKNKKVSLKNNGMFNKTILLFVFKYYQIAFKLFNDIELY





SLFFLREKSEKPFEVFLEELKDKMIGKQLNFGQLLYVVYEVLVKNKDLDK





ILSKKIDYRKDKSFSPEIAYLRNFLSHLNYSKFLDNFMKINTNKSDENKE





VLIPSIKIQKMIQFIEKCNLQNQIDFDFNFVNDFYMRKEKMFFIQLKQIF





PDINSTEKQKKSEKEEILRKRYHLINKKNEQIKDEHEAQSQLYEKILSLQ





KIFSCDKNNFYRRLKEEKLLFLEKQGKKKISMKEIKDKIASDISDLLGIL





KKEITRDIKDKLTEKFRYCEEKLLNISFYNHQDKKKEEGIRVFLIRDKNS





DNFKFESILDDGSNKIFISKNGKEITIQCCDKVLETLMIEKNTLKISSNG





KIISLIPHYSYSIDVKY









In some cases, a suitable programmable nuclease for use in the compositions and methods herein comprises an amino acid sequence having at least 60% amino acid sequence identity to any one of SEQ ID NO: 397-SEQ ID NO: 424. In some cases, a suitable programmable nuclease for use in the compositions and methods herein comprises an amino acid sequence having at least 80% amino acid sequence identity to any one of SEQ ID NO: 397-SEQ ID NO: 424. In some cases, a subject programmable nuclease for use in the compositions and methods herein may include an amino acid sequence having 80% or more (e.g., 85% or more, 90% or more, 95% or more, 98% or more, 99% or more, 99.5% or more, or 100%) amino acid sequence identity with the amino acid sequence set forth in any one of SEQ ID NO: 397-SEQ ID NO: 424. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 424. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 397. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 398. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 399. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 400. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 401. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 402. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 403. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 404. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 405. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 406. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 407. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 408. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 409. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 410. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 411. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 412. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 413. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 414. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 415. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 416. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 417. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 418. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 419. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 420. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 421. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 422. In some cases, a suitable programmable nuclease polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 423.









TABLE 20







Amino Acid Sequences of Exemplary Programmable Nucleases








SEQ ID



NO:
Programmable Nuclease Amino Acid Sequence





SEQ ID
MAGKKKDKDVINKTLSVRIIRPRYSDDIEKEISDEKAKRKQDGKTGELDR


NO: 424
AFFSELKSRNPDIITNDELFPLFTEIQKNLTEIYNKSISLLYMKLIVEEEGGST



ASALSAGPYKECKARFNSYISLGLRQKIQSNFRRKELKGFQVSLPTAKSDR



FPIPFCHQVENGKGGFKVYETGDDFIFEVPLIKYTATNKKSTSGKNYTKVQ



LNNPPVPMNVPLLLSTMRRRQTKKGMQWNKDEGTNAELRRVMSGEYK



VSYAEIIRRTRFGKHDDWFVNFSIKFKNKTDELNQNVRGGIDIGVSNPLVC



AVTNGLDRYIVANNDIMAFNERAMARRRTLLRKNRFKRSGHGAKNKLEP



ITVLTEKNERFRKSILQRWAREVAEFFKRTSASVVNMEDLSGITEREDFFST



KLRTTWNYRLMQTTIENKLKEYGIAVNYISPKYTSQTCHSCGKRNDYFTF



SYRSENNYPPFECKECNKVKCNADFNAAKNIALKVVL





SEQ ID
MCMKITKIDGISHKKYKEKGKLIKNNDTAKDIIEERFNDIEKKTKELFQKT


NO: 397
LDFYVKNYEKCKEQNKERREKAKNYFSKVKILVDNKKITICNENTEKMEI



EDFNEYDVRSGKYFNVLNKILNGENYTEEDLEVFENDLQKRTGRIKSIKN



SLEENKAHFKKESINNNIIYDRVKGNNKKSLFYEYYRISSKHQEYVNNIFE



AFDKLYSNSHEAMNNLFSEITKDSKDRNIRKIREAYHEILNKNKTEFGEEL



YKKIQDNRNNFDKLLEIEPEIKELTKSQIFYKYYIDKVNLDETSIKHCFCHL



VEIEVNQLLKNYVYSKRNINKEKLENIFEYCKLKNLIKNKLVNKLNNYIR



NCGKYNAYISNNDVVVNSEKISEIRTKEAFLRSIIGVSSSAYFSLRNILNTD



NTQDITNKVDKEVDKLYQENKKIELEERLKLFFGNYFDINNQQEIKVFLM



NIDKIISSIRHEIIHFKMETNAQNIFDENNVNLGNTAKNIFSNEINEEKIKFKI



FKQLNSANVFDYLSNKDITEYMDKVVFSFTNRNVSFVPSFTKIYNRVQDL



ANSLEIKKWKIPDKSEGKDAQIYLLKNIYYGKFLDEFLNEENGIFISIKDKII



ELNRNQNKRTGFYKLEKFEKIEETNPKKYLEIIQSLYMINIEEIDSEGKNIFL



DFIQKIFLKGFFEFIKNNYNYLLELKKIQDKKNIFDSEMSEYIAGEKTLEDI



GEINEIIQDIKITEIDKILNQTDKINCFYLLLKLLNYKEITELKGNLEKYQILS



KTNVYEKELMLLNIVNLDNNKVKIENFKILAEEIGKFIEKINIEEINKNKKI



KTFEELRNFEKGENTGEYYNIYSDDKNIKNIRNLYNIKKYGMLDLLEKISE



KTNYCIKKKDLEEYSELRKQLEDEKTNFYKIQEYLHSKYQQKPKKILLKN



NKNDYEKYKKSIENIEKYVHLKNKIEFNELNLLQSLLLKILHRLVGFTSIW



ERDLRFRLIGEFPDELDVEDIFDHRKRYKGTGKGICKKYDRFINTHTEYKN



NNKMENVKFADNNPVRNYIAHFNYLPNPKYSILKMMEKLRKLLDYDRK



LKNAVMKSIKDILEEYGFKAEFIINSDKEIILNLVKSVEIIHLGKEDLKSRRN



SEDLCKLVKAMLEYSK*





SEQ ID
MEKIKKPSNRNSIPSIIISDYDANKIKEIKVKYLKLARLDKITIQDMEIVDNI


NO: 398
VEFKKILLNGVEHTIIDNQKIEFDNYEITGCIKPSNKRRDGRISQAKYVVTI



TDKYLRENEKEKRFKSTERELPNNTLLSRYKQISGFDTLTSKDIYKIKRYID



FKNEMLFYFQFIEEFFNPLLPKGKNFYDLNIEQNKDKVAKFIVYRLNDDFK



NKSLNSYITDTCMIINDFKKIQKILSDFRHALAHFDFDFIQKFFDDQLDKN



KFDINTISLIETLLDQKEEKNYQEKNNYIDDNDILTIFDEKGSKFSKLHNFY



TKISQKKPAFNKLINSFLSQDGVPNEEFKSYLVTKKLDFFEDIHSNKEYKKI



YIQHKNLVIKKQKEESQEKPDGQKLKNYNDELQKLKDEMNTITKQNSLN



RLEVKLRLAFGFIANEYNYNFKNFNDEFTNDVKNEQKIKAFKNSSNEKLK



EYFESTFIEKRFFHFSVNFFNKKTKKEETKQKNIFNSIENETLEELVKESPLL



QIITLLYLFIPRELQGEFVGFILKIYHHTKNITSDTKEDEISIEDAQNSFSLKF



KILAKNLRGLQLFHYSLSHNTLYNNKQCFFYEKGNRWQSVYKSFQISHNQ



DEFDIHLVIPVIKYYINLNKLMGDFEIYALLKYADKNSITVKLSDITSRDDL



KYNGHYNFATLLFKTFGIDTNYKQNKVSIQNIKKTRNNLAHQNIENMLKA



FENSEIFAQREEIVNYLQTEHRMQEVLHYNPINDFTMKTVQYLKSLSVHS



QKEGKIADIHKKESLVPNDYYLIYKLKAIELLKQKVIEVIGESEDEKKIKN



AIAKEEQIKKGNN





SEQ ID
MKDYIRKTLSLRILRPYYGEEIEKEIAAAKKKSQAEGGDGALDNKFWDRL


NO: 399
KAEHPEIISSREFYDLLDAIQRETTLYYNRAISKLYHSLIVEREQVSTAKALS



AGPYHEFREKFNAYISLGLREKIQSNFRRKELARYQVALPTAKSDTFPIPIY



KGFDKNGKGGFKVREIENGDFVIDLPLMAYHRVGGKAGREYIELDRPPAV



LNVPVILSTSRRRANKTWFRDEGTDAEIRRVMAGEYKVSWVEILQRKRF



GKPYGGWYVNFTIKYQPRDYGLDPKVKGGIDIGLSSPLVCAVTNSLARLTI



RDNDLVAFNRKAMARRRTLLRQNRYKRSGHGSANKLKPIEALTEKNELY



RKAIMRRWAREAADFFRQHRAATVNMEDLTGIKDREDYFSQMLRCYWN



YSQLQTMLENKLKEYGIAVKYIEPKDTSKTCHSCGHVNEYFDFNYRSAH



KFPMFKCEKCGVECGADYNAARNIAQA





SEQ ID
MKITKIDGILHKKYIKEGKLVKSTSEENKTDERLSELLTIRLDTYIKNPDNA


NO: 400
SEEENRIRRETLKEFFSNKVLYLKDSILYLKDRREKNQLQNKNYSEEDISE



YDLKNKNSFLVLKKILLNEDINSEELEIFRNDFEKKLDKINSLKYSLEENK



ANYQKINENNIKKVEGKSKRNIFYNYYKDSAKRNDYINNIQEAFDKLYKK



EDIENLFFLIENSKKHEKYKIRECYHKIIGRKNDKENFATIIYEEIQNVNNM



KELIEKVPNVSELKKSQVFYKYYLNKEKLNDENIKYVFCHFVEIEMSKLL



KNYVYKKPSNISNDKVKRIFEYQSLKKLIENKLLNKLDTYVRNCGKYSFY



LODGEIATSDFIVGNRQNEAFLRNIIGVSSTAYFSLRNILETENENDITGRIK



GKTVKNKKGEEKYISGEIDKLYDNNKQNEVKKNLKMFYSYDFNMNRKK



EIEDFFSNIDEAISSIRHGIVHFNLELEGKDIFTFKNIVPSQISKKMFQNEINE



KKLKLKIFRQLNSANVFRYLEKYKILNYLNRTRFEFVNKNIPFVPSFTKLY



SRIDDLKNSLCIYWKIPKANDNNKTKEITDAQIYLLKNIYYGEFLNYFMSN



NGNFFEIIKEIIELNKNDKRNLKTGFYKLQKFENLQEKTPKEYLANIQSFY



MIDAGNKDEEEKDAYIDFIQKIFLKGFMTYLANNGRLSLMYIGNDEQINT



SLAGKKQEFDKFLKKYEQNNNIEIPHEINEFVREIKLGKILKYTESLNMFY



LILKLLNHKELTNLKGSLEKYQSANKEEAFSDQLELINLLNLDNNRVTEDF



ELEADEIGKFLDFNGNKVKDNKELKKFDTNKIYFDGENIIKHRAFYNIKK



YGILNLLEKISDEAKYKISIEELKNYSNKKIEIEKNHTTQENLHRKYARPRK



DEKFNDEDYKKYEKTIRNIQQYTHLKNKVEFNELNLLQSLLLRILHRLVG



YTSIWERDLRFRLKGEFPENQYIEEIFNFDNSKNVKYKNGQIVEKYISFYK



ELYKDDMEKISIYSDKKVKELKKEKKDLYIRNYIAHFNYIPNAEVSLLEVL



ENLRKLLSYDRKLKNAIMKSIVDILKEYGFVVTFKIEKDKKIRIESLKSEEV



VHLKKLKLKDNDKKKEPIKTYRNSKELCKLVKVMFEYKMKEKKSEN*





SEQ ID
MKITKIDGISHKKYIKEGKLVKSTSEENKTDERLSELLTIRLDTYIKNPDNA


NO: 401
SEEENRIRRENLKEFFSNKVLYLKDGILYLKDRREKNQLQNKNYSEEDISE



YDLKNKNSFLVLKKILLNEDINSEELEIFRKDVEAKLNKINSLKYSFEENK



ANYQKINENNVEKVGGKSKRNIIYDYYRESAKRNDYINNVQEAFDKLYK



KEDIEKLFFLIENSKKHEKYKIRECYHKIIGRKNDKENFAKIIYEEIQNVNNI



KELIEKVPDMSELKKSQVFYKYYLDKEELNDKNIKYAFCHFVEIEMSQLL



KNYVYKRLSNISNDKIKRIFEYQNLKKLIENKLLNKLDTYVRNCGKYNYY



LODGEIATSDFIAGNRQNEAFLRNIIGVSSVAYFSLRNILETENKDDITGKM



RGKTRIDSKTGEEKYIPGEVDQIYYENKQNEVKNKLKMFYGYDFDMDN



KKEIEDFFANIDEAISSIRHGIVHFNLDLDGKDIFAFKNIVPSEISKKMFQNE



INEKKLKLKIFRQLNSANVFRYLEKYKILNYLKRTRFEFVNKNIPFVPSFTK



LYSRIDDLKNSLGIYWKTPKTNDDNKTKEIIDAQIYLLKNIYYGEFLNYFM



SNNGNFFEISREIIELNKNDKRNLKTGFYKLQKFEDIQEKTPKKYLANIQSL



YMINAGNQDEEEKDTYIDFIQKIFLKGFMTYLANNGRLSLMYIGNDEQIN



TSLAGKKQEFDKFLKKYEQNNNIEIPHEINEFLREIKLGKILKYTESLNMF



YLILKLLNHKELTNLKGSLEKYQSANKEETFSDELELINLLNLDNNRVTED



FELEANEIGKFLDFNGNKIKDRKELKKFDTKKIYFDGENIIKHRAFYNIKK



YGMLNLLEKIADKAKYKISLKELKEYSNKKNEIEKNYTMQQNLHRKYAR



PKKDEKFNDEDYKEYEKAIGNIQKYTHLKNKVEFNELNLLQGLLLKILHR



LVGYTSIWERDLRFRLKGEFPENQYIEEIFNFDNSKNVKYKSGQIVEKYIN



FYKELYKDNVEKRSIYSDKKVKKLKQEKKDLYIRNYIAHFNYIPHAEISLL



EVLENLRKLLSYDRKLKNAIMKSVVDILKEYGFVATFKIGADKKIGIQTLE



SEKIVHLKNLKKKKLMTDRNSEELCKLVKVMFEYKMEEKNLKTKKCKVI*





SEQ ID
MKNQNTLPSNPTDILKDKPFWAAFFNLARHNVYLTVNHINKLLDLEKLY


NO: 402
NKDKHKEIFEHEDIFNISDDVMNDVNSNGKKRKLDIKKIWANLDTDLTRK



YQLRELILKHFPFIQPAIIGAQTKERTTIDKDKRSTSTSNDSLKPTGEGDIND



PLSLSNVKSIFFRLLQMLEQLRNYYSHVKHSKSATMPNFDEGLLKSMYNI



FIDSVNKVKEDYSSNSVIDPNTSFSHLISKDEQGEIKPCRYSFTSKDGSINAS



GLLFFVSLFLEKQDSIWMQKKIPGFKKTSENYMKMTNEVFCRNHILLPKM



RLETVYDKDWMLLDMLNEVVRCPLSLYKRLAPADQNKFKVPEKSSDNA



NRQEDDNPFSRILVRHQNRFPYFALRFFDLNEVFTTLRFQINLGCYHFAIC



KKQIGDKKEVHHLTRTLYGFSRLQNFTQNTRPEEWNTLVKTTEPSSGNDG



KTVQGVPLPYISYTIPHYQIENEKIGIKIFDGDTAVDTDIWPSVSTEKQLNK



PDKYTLTPGFKADVFLSVHELLPMMFYYQLLLCEGMLKTDAGNAVEKVL



IDTRNAIFNLYDAFVQEKINTITDLENYLQDKPILIGHLPKQMIDLLKGHQR



DMLKAVEQKKAMLIKDTERRLERLNKQPEQKPNVAAKNTGTLLRNGQIA



DWLVKDMMRFQPVKRDKEGNPINCSKANSTEYQMLQRAFAFYTTDSYR



LPRYFEQLHLINCDNSHLFLSRFEYDKQPNLIAFYAAYLEAKLEFLNELQP



QNWASDNYFLLLRAPKNDRQKLAEGWKNGFNLPRGLFTEKIKTWFNEH



KTIVDISDCDIFKNRVGQVARLIPVFFDKKFKDHSQPFYTYNFNVGNVSKI



TEANYLSKEKRENLFKSYQNKFKNNIPAEKTKEYREYKNFSSWKKFEREL



RLIKNQDILTWLMCKNLFDEKIKPKKDILEPRIAVSYIKLDSLQTNTSTAGS



LNALAKVVPMTLAIHIDSPKPKGKAGNNEKENKEFTVYIKEEGTKLLKW



GNFKTLLADRRIKGLFSYIEHDDINLEKYPLTKYQVDSELDLYQKYRIDIF



KQTLDLEAQLLDKYSDLNTDNFNQMLSGWSEKEGIPRNIKQDVAFLIGVR



NGFSHNQYPDSKRIAFSRIKKFNPKTSSLQESEGLNIAKQMYEEAQQVVN



KIKNIESFD*





SEQ ID
MKVTKIDGISHKKFEDEGKLVKFTGHFNIKNEMKERLEKLKELKLSNYIK


NO: 403
NPENVKNKDKNKEKETKSRRENLKKYFSEIILRKKEEKYLLKKTRKFKNI



TEEINYDDIKKRENQQKIFDVLKELLEQRINENDKEEILNFDSVKLKEAFE



EDFIKKELKIKAIEESLEKNRADYRKDYVELENEKYEDVKGQNKRSLVFE



YYKNPENREKFKENIKYAFENLYTEENIKNLYSEIKEIFEKVHLKSKVRYF



YQNEIIGESEFSEKDEEGISILYKQIINSVEKKEKFIEFLQKVKIKDLTRSQIF



YKYFLENEELNDENIKYVFSYFVEIEVNKLLKENVYKTKKFNEGNKYRV



KNIFNYDKLKNLVVYKLENKLNNYVRNCGKYNYHMENGDIATSDINMK



NRQTEAFLRSILGVSSFGYFSLRNILGVNDDDFYKIEKDERKNENFILKKA



KEDFTSKNIFEKVVDKSFEKKGIYQIKENLKMFYGNSFDKVDKDELKKFF



VNMLEAITSVRHRIVHYNINTNSENIFDFSNIEVSKLLKNIFEKEIDTRELKL



KIFRQLNSAGVFDYWESWVIKKYLENVKFEFVNKNVPFVPSFKKLYDRID



NLKGWNALKLGNNINIPKRKEAKDSQIYLLKNIYYGEFVEKFVNDNKNF



EKIVKEIIEINRGAGTNKKTGFYKLEKFETLKANTPTKYLEKLQSLHKISY



DKEKIEEDKDVYVDFVQKIFLKGFVNYLKKLDSLKSLNLLNLRKDETITD



KKSVHDEKLKLWENSGSNLSKMPEEIYEYVKKIKISNINYNDRMSIFYLLL



KLIDYRELTNLRGNLEKYESMNKNKIYSEELTIINLVNLDNNKVRTNFSLE



AEDIGKFLKSSITIKNIAQLNNFSKIFADGENVIKHRSFYNIKKYGILDLLEK



IVAKADLKITKEEIKKYENLQNELKRNDFYKIQEQIHRNYNQKPFSIKKIEN



KKDFEKYKKVIEKIQDYTQLKNKIEFNDLNLLQSLIFRILHRLAGYTSLWE



RDLQFKLKGEFPEDKYIDEIFNSDGNNNQKYKHGGIADKYANFLIEKKEE



KSGEILNKKQRKKKIKEDLEIRNYIAHFNYLPNAEKSILEILEELRELLKHD



RKLKNAVMKSIKDIFREYGFIVEFTISHTKNGKKIKVCSVKSEKIKHLKNN



ELITTRNSEDLCELVKIMLEHKELQK*





SEQ ID
MRISKTLSLRIVRPFYTPEVEAGIKAEKDKREAQGQTRSLDAKFFNELKK


NO: 404
KHSEIILSSEFYSLLSEVQRQLTSIYNHAMSNLYHKIIVEGEKTSTSKALSNI



GYDECKAIFPSYMALGLRQKIQSNFRRRDLKNFRMAVPTAKSDKFPIPIYR



QVDGSKGGFKISENDGKDFIVELPLVDYVAEEVKTAKGRFTKINISKPPKI



KNIPVILSTLRRRQSGQWFSDDGTNAEIRRVISGEYKVSWIEIVRRTRFGK



HDDWFVNMVIKYDKPEEGLDSKVVGGIDVGVSSPLVCALNNSLDRYFVK



SSDIIAFNKRAMARRRTLLRQNKYKRSGHGSKNKLEPITVLTEKNERFKK



SIMQRWAKEVAEFFRGKGASVVRMEELSGLKEKDNFFSSYLRMYWNYG



QLQQIIENKLKEYGIKVNYVSPKDTSKKCHSCTHINEFFTFEYRQKNNFPL



FKCEKCGVECSADYNAAKNMAIA





SEQ ID
VKISKTLSLRIIRPYYTPEVESAIKAEKDKREAQGQTRNLDAKFFNELKKK


NO: 405
HPQIILSGEFYSLLFEMQRQLTSIYNRAMSSLYHKIIVEGEKTSTSKALSDIG



YDECKSVFPSYIALGLRQKIQSNFRRKELKGFRMAVPTAKSDKFPIPIYKQ



VDDGKGGFKISENKEGDFIVELPLVEYTAEDVKTAKGKFTKINISKPPKIK



NIPVILSTLRRKQSGQWFSDEGTNAEIRRVISGEYKVSWIEVVRRTRFGKH



DDWFLNIVIKYDKTEDGLDPEVVGGIDVGVSTPLVCAVNNSLDRYFVKSS



DIIAFKKRAMARRRTLLRQNRFKRSGHGSKSKLEPITILTEKNERFKKSIM



QRWAKEVAEFFKGERASVVQMEELSGLKEKDNFFGSYLRMYWNYGQLQ



QIIENKLKEYGIKVNYVSPKDTSKKCHSCGYINEFFTFEFRQKNNFPLFKC



KKCGVECNADYNAAKNIAIA





SEQ ID
VKLPILKPLHKQILSEEYSTSFKIKAFENDNEVLKAIDTFWNEHIEKSIHPV


NO: 406
TGNKFNILSKIENLCDQLQKYKDKDLEKLFIERKNLSTVSHQVYGQWNII



RDALRMHLEMNNKNIKEKDIDKYLDNDAFSWKEIKDSIKIYKEHVEDAK



ELNENGIIKYFSAMSINEEDDEKEYSISLIKNINEKYNNVKSILQEDRTGKS



DLHQDKEKVGIIKEFLDSLKQLQWFLRLLYVTVPLDEKDYEFYNELEVYY



EALLPLNSLYNKVRNYMTRKPYSVEKFKLNFNSPTLLDGWDKNKETANL



SIILRKNGKYYLGIMNKENNTIFEYYPGTKSNDYYEKMIYKLLPGPNKML



PKVFFSKKGLEYYNPPKEILNIYEKGEFKKDKSGNFKKESLHTLIDFYKEA



IAKNEDWEVFNFKFKNTKEYEDISQFYRDVEEQGYLITFEKVDANYVDK



LVKEGKLYLFQIYNKDFSENKKSKGNPNLHTIYWKGLYDSENLKNVVYK



LNGEAEVFYRKKSIDYPEEIYNHGHHKEELLGKFNYPIIKDRRYTQDKFLF



HVPITMNFISKEEKRVNQLACEYLSATKEDVHIIGIDRGERHLLYLSLIDKE



GNIKKQLSLNTIKNENYDKEIDYRVKLDEKEKKRDEARKNWDVIENIKEL



KEGYMSQVIHIIAKMMVEEKAILIMEDLNIGFKRGRFKVEKQVYQKFEK



MLIDKLNYLVFKNKNPLEPGGSLNAYQLTSKFDSFKKLGKQSGFIFYVPSA



YTSKIDPTTGFYNFIQVDVPNLEKGKEFFSKFEKIIYNTKEDYFEFHCKYG



KFVSEPKNKDNDRKTKESLTYYNAIKDTVWVVCSTNHERYKIVRNKAGY



YESHPVDVTKNLKDIFSQANINYNEGKDIKPIIIESNNAKLLKSIAEQLKLIL



AMRYNNGKHGDDEKDYILSPVKNKQGKFFCTLDGNQTLPINADANGAY



NIALKGLLLIEKIKKQQGKIKDLYISNLEWFMFMMSR





SEQ ID
MIKNPSNRHSLPKVIISEVDHEKILEFKIKYEKLARLDRFEVKAMHYEGKE


NO: 407
IVFDEVLVNGGLIEVEYQDDNKTLFVKVGEKSYSIRGKKVGGKQRLLEDR



VSKTKVQLELSDGVVDNKGNLRKSRTERELIVADNIKLYSQIVGREVTTT



KEIYLVKRFLAYRSDLLFYYSFVDNFFKVAGNEKELWKINFDDATSAQFM



GYIPFMVNDNLKNDNAYLKDYVRNDVQIKDDLKKVQTIFSALRHTLLHF



NYEFFEKLFNGEDVGFDFDIGFLNLLIENIDKLNIDAKKEFIDNEKIRLFGE



NLSLAKVYRLYSDICVNRVGFNKFINSMLIKDGVENQVLKAEFNRKFGGN



AYTIDIHSNQEYKRIYNEHKKLVIKVSTLKDGQAIRRGNKKISELKEQMKS



MTKKNSLARLECKMRLAFGFLYGEYNNYKAFKNNFDTNIKNSQFDVND



VEKSKAYFLSTYERRKPRTREKLEKVAKDIESLELKTVIANDTLLKFILLM



FVFMPQELKGDFLGFVKKYYHDVHSIDDDTKEQEEDVVEAMSTSLKLKI



LGRNIRSLTLFKYALSSQVNYNSTDNIFYVEGNRYGKIYKKLGISHNQEEF



DKTLVVPLLRYYSSLFKLMNDFEIYSLAKANPTAVSLQELVDDETSPYKQ



GNYFNFNKMLRDIYGLTSDEIKSGQVVFMRNKIAHFDTEVLLSKPLLGQT



KMNLQRKDIVSFIEARGDIKELLGYDAINDFRMKVIHLRTKMRVYSDKLQ



TMMDLLRNAKTPNDFYNVYKVKGVESINKHLLEVLAQTAEERTVEKQIR



DGNEKYDL





SEQ ID
LNSIEKIKKPSNRNSIPSIIISDYDENKIKEIKVKYLKLARLDKITIQDMEIRD


NO: 408
NIVEFKKILLNGIEHTIKDNQKIEFDNYEITAYVRASKQRRDGKITQAKYV



VTITDKYLRDNEKEKRFKSTERELPNDTLLMRYKQISGFDTLTSKDIYKIK



RYIDFKNEMLFYFQFIEEFFSPLLPKGTNFYSLNIEQNKDKVVKYIVYRLN



DDFKNQSLNQFIKKTDTIKYDFLKIQKILSDFRHALAHFDFDFIQKFFDDEL



DKNRFDISTISLIKTMLQEKEEKYYQEKNNYIEDSDTLTLFDEKESNFSKIH



NFYIKISQKKPAFNKLINSFLSKDGVPNEELKSYLATKKIDFFEDIHSNKEY



KKIYIKHKNLVVEKQKEESQEKPNGQKLKNYNDELQKLKDEMNKITKQN



SLNRLEVKLRLAFGFIANEYNYNFKNFNDKFTLDVKKEQKIKVFKNSSNE



KLKEYFESTFIEKRFFHFCVKFFNKKTKKEETKQKNIFNLIENETLEELVKE



SPLLQIITLLYLFIPKELQGEFVGFILKIYHHTKNITNDTKEDEKSIEDTQNSF



SLKLKILAKNLRGLQLFNYSLSHNTLYNTKEHFFYEKGNRWQSVYKSLEI



SHNQDEFDIHLVIPVIKYYINLNKLIGDFEIYALLTYADKNSITEKLSDITKR



DDLKFRGYYNFSTLLFKTFMINTNYEQNQKSTQYIKQTRNDIAHQNIENM



LKAFENNEIFAQREEIVNYLQKEHKMQEILHYNPINDFTMKTVQYLKSLNI



HSQKESKIADIHKKESLVPNDYYLIYKLKVIELLKQKVIEAIGETKDEEKIK



NAIAKEEQIKKGYNK





SEQ ID
MLKHKRKNKNSLARVVLSNYDSNNIYEIKIKYEKLAKLDKINIIEMDYDA


NO: 409
DNNVMFKKVLFNNKEIDLSHKDKTKINIELDNKKYNISAKKQIGKTHLVV



RNKQTSKISRIKKIQDTYYRGKDVFILDNNIEILDKKQTKDKFIVTLNDITN



NKTTSTEAELIDDTKDIFKKISAKKDLKSSDIYKIKRFISIRSNFSFYYTFVD



NYFKIFHAKKDKNKEELYKIKFKDEINIKPYLENILDNMKNKNGILYNYAN



DRKKVLNDLRNIQYVFKEFRHKLAHFDYNFLDNFFSNSVEEKYKQKVNE



IKLLDILLDNIDSLNVVPKQNYIEDETISVFDAKDIKLKRLYTYYIKLTINYP



GFKKLINSFFIQDGIENQELKEYINNKEKDTQVLKELDNKAYYMDISQYR



KYKNIYNKHKELVSEKELSSDGKKINSLNQKINKLKIDMKNITKPNALNR



LIYRLRVAFGFIYKEYATINNFNKSFLQDTKTKRFENISQQDIKSYLDISYQD



KGKFFVKSKKTFKNKTTVKYTFEDLDLTLNEIITQDDIFVKVIFLFSIFMPK



ELNGDFFGFINMYYHKMKNISYDTKDIDMLDTISQNMKLKILEQNIKKTY



VFKYYLDLDSSIYSKLVQNIKITEDIDSKKYLYAKIFKYYQHLYKLISDVEI



YLLYKYNSKENLSITIDKDELKHRGYYNFQSLLIKNNINKDDAYWSIVNM



RNNLSHQNIDELVGHFCKGCLRKSTTDIAELWLRKDILTITNEIINKIESFK



DIKITLGYDCVNDFTQKVKQYKQKLKASNERLAKKIEEKQNQVVDEKNK



EELEKNILNMKNIQKINRYILDIL





SEQ ID
MLKHKRKNKNSLARVVLSNYDSNNIYEIKIKYEKLAKLDKINIIEMDYDA


NO: 410
DNNVMFKKVLFNNKEIDLSHKDKTKINIELDNKKYNISAKKQIGKTHLVV



RDKQTSKISRIKKIQDTYYRGKDVFILDNNIEILDKKQTKDKFIVTLNDITN



DKTTSTEAELIDDTKDIFKKISAKKDLKSSDIYKIKRFISIRSNFSFYYTFVD



NYFKIFHAKKDKNKEELYKIKFKDEINIKPYLENILDNMKNKNGILYDYAD



DREKVLNDLKNIQYVFTEFRHKLAHFDYNFLDNFFSNSVTDQYKQKVNE



IKLLDILLDNIDSLNVVPKQNYIEDETISVFDAKDIKLKRLYTYYIKLTINYP



GFKKLINSFFIQDGIENQELKEYINNKEKDTQVLKELDNKAYYMDISQYR



KYKNIYNKHKELVSEKELSSDGQKINSLNQKINKLKIEMKNITKPNALNR



LIYRLRVAFGFIYKEYATINNENKSFLQDTKIKRFENISQQDIKNYLDISYQD



KGKFFVKSKKTFKNKTTIKYTFEDLDLTLNEIITQDDIFVKVIFLFSIFMPKE



LNGDFFGFINMYYHKMKNISYDTKDIDMLDTISQNMKLKILEQNIKKTYV



FKYYLDLDSSIYSKLVQNIKITEDIDSKKYLYAKIFKYYQHLYKLISDVEIY



LLYKYNSKENLSITIDKDELKHRGYYNFQSLLIKNNINKDDAYWSIVNMR



NNLSHQNIDELVGHFCKGCLRKSTTDIAELWLRKDILTITNEIINKIESFKDI



KITLGYDCVNDFTQKVKQYKQKLKASNERLAKKIEEKQNQVVDEKNKE



ELEKKILNMKNIQKINRYILDIL





SEQ ID
MSQLKNPSNKNSLPRIIISDFNEIKINEIKIKYHKLDRLDKIIVKEMEIINNKI


NO: 411
FFKKILFNNQIKDINSENIELENYILAGEVKPSNTKIILNRDGKEKSFIVYDG



FTFKYKPNDKRISETKTNAKYILTIKDKTRHRESSTQRDILKSSIIETYKQIS



GFENITSKDIYTIKRYIDFKNEMMFYYTFIDDFFFPITGKNKQDKKNNFYN



YKIKENAKKFISLINYRINDDFKNKNGILYDYLSNKEEIIINDFIHIQTILKDV



RHAIAHFNFDFIQKLFDNEQAFNSKFDGIEILNILFNQKQEKYFEAQTNYIE



EETIKILDEKELSFKKLHSFYSQICQKKPAFNKLINSFIIQDGIENKELKDYIS



QKYNSKFDYYLDIHTCKIYKDIYNQHKKFVADKQFLENQKTDGQKIKKL



NDQINQLKTKMNNLTKKNSLKRLEIKFRLAFGFIFTEYQTFKNFNERFIEDI



KANKYSTKIELLDYGKIKEYISITHEEKRFFNYKTFNKKTNKNINKTIFQSL



EKETFENLVKNDNLIKMMFLFQLLLPRELKGEFLGFILKIYHDLKNIDNDT



KPDEKSLSELNISTALKLKILVKNIRQINLFNYTISNNTKYEEKEKRFYEEG



NQWKDIYKKLYISHDFDIFDIHLIIPIIKYNINLYKLIGDFEVYLLLKYLERN



TNYKTLDKLIEAEELKYKGYYNFTTLLSKAINIALNDKEYHNITHLRNNTS



HQDIQNIISSFKNNKLLEQRENIIELISKESLKKKLHFDPINDFTMKTLQLLK



SLEVHSDKSEKIENLLKKEPLLPNDVYLLYKLKGIEFIKKELISNIGITKYEE



KIQEKIAKGVEK





SEQ ID
MVKNPANRHALPKVIISEVDNNNILEFKIKYEKLARLDKVEVKSMHFDNN


NO: 412
KQVVFDEVVINGGLIEPTYEDKHKKLVVTAGEKSYSIVGQKVGGKPRLLE



DRVSKTKVQLELTNYVEDKEGKKRVSKTERELIVADNIELYSQIVGREVKT



TKEIYLIKRFLEYRSDLLFYYGFVDNFFKVAGNGKELWKIDFTNSDSLHLI



EYFKFSINDNLKNDENYLKNYVSDNTKIENDLVKCQNNFNSLRHALMHF



DYDFFEKLFNGEDVGFDFDIEFLNIMIDKVDKLNIDTKKEFIDDEEVTLFG



EALSLKKLYGLFSHIAINRVAFNKLINSFIIEDGIENKELKDFFNNKKESQAY



EIDIHSNAEYKALYVQHKKLVMATSAMTDGDEIAKKNQEISDLKEKMKVI



TKENSLARLEHKLRLAFGFIYTEYKDYKTFKKHFDQDIKGAKYKGLNVE



KLKEYYETTLKNSKPKTDEKLEDVAKKIDKLSLKELIDDDTLLKFVLLLFI



FMPQELKGDFLGFIKKYYHDKKHIDQDTKDKDTEIEELSTGLKLKVLDKN



IRSLSILKHSFSFQVKYNRKDKNFYEDGNLHGKFYKKLSISHNQEEFNKSV



YAPLFRYYSALYKLINDFEIYALAQHVENHETLADQVNKSQFIQKSYFNFR



KLLDNTDSISQSSSYNTLIVMRNDISHLSYEPLFNYPLDERKSYKKKTQKG



VKTFHVELLYISRAKIIELISLQTDMKKLLGYDAVNDFNMKVVHLRKRLS



VYANKEESIRKMQADAKTPNDFYNIYKVKGVESINQHLLKVIGVTEAEKS



IEKQINEGNKKHNT





SEQ ID
MTKKPSNRNSLPKVIINKVDESSILEFKIKYEKLARLDRFEVRSMRYDGDG


NO: 413
RIIFDEVVANAGLLDVDYEDDNRTIVVKIENKAYNIYGKKVGGEKRLNGK



ISKAKVQLILTDSIRKNANDTHRHSLTERELINKNEVDLYSKIAEREISTTK



DIYLVKRFLAYRSDLLLYYAFINHYVRVNGNKKEFWKTEIDDKIIDYFIYTI



NDTLKNKEGYLEKYIVDRDQIKKDLEKIKQIFSHLRHKLMHYDFRFFTDL



FDGKDVDIKVDNSIQKISELLDIEFLNIVIDKLEKLNIDAKKEFIDDEKITLF



GQEIELKKLYSLYAHTSINRVAFNKLINSFLIKDGVENKELKEYFNAHNQG



KESYYIDIHQNQEYKKLYIEHKNLVAKLSATTDGKEIAKINRELADKKEQ



MKQITKANSLKRLEYKLRLAFGFIYTEYKDYERFKNSFDTDTKKKKFDAI



DNAKIIEYFEATNKAKKIEKLEEILKGIDKLSLKTLIQDDILLKFLLLFFTFL



PQEIKGEFLGFIKKYYHDITSLDEDTKDKDDEITELPRSLKLKIFSKNIRKLS



ILKHSLSYQIKYNKKESSYYEAGNVFNKMFKKQAISHNLEEFGKSIYLPM



LKYYSALYKLINDFEIYALYKDMDTSETLSQQVDKQEYKRNEYFNFETLL



RKKFGNDIEKVLVTYRNKIAHLDFNFLYDKPINKFISLYKSREKIVNYIKNH



DIQAVLKYDAVNDFVMKVIQLRTKLKVYADKEQTIESMIQNTQNPNGFYN



IYKVKAVENINRHLLKVIGYTESEKAVEEKIRAGNTSKS





SEQ ID
MIKNPSNRYALPKVIISKIDNQNILEFKIKYKKLSKLDIVKVKSMHYDDRAI


NO: 414
IFDEVIVNDGLIDVEYRDNHKTIFVKVGNKSYSISGQKVGGKERLLENRVS



KTKVQLELKDKATNRVSKTERELIVDDNIKIYSQIVGRDVKTTKDIYLIKR



FLAYRSDLLFYYGFVNNFFHVANNRSEFWKIDFNDSNNSKLIEYFKFTIND



HLKNDENYLKDYISDNEKLKNDLIKVKNSFEKIRHALMHFDYDFFVKLF



NGEDVGLELDIEFLDIMIDKLDKLNIDTKKEFIDDEKITIFGEELSLAKLYRF



YAHTAINRVAFNKLINSFIIENGVENQSLKEYFNQQAGGIAYEIDIHQNREY



KNLYNEHKKLVSRVLSISDGQEIAILNQKIAKLKDQMKQITKANSIKRLEY



KLRLALGFIYTEYENYEEFKNNFDTDIKNGRFTPKDNDGNKRAFDSRELE



QLKGYYEATIQTQKPKTDEKIEEVSKKIDRLSLKSLIADDILLKFILLMFTF



MPQELKGEFLGFIKKYYHDTKHIDQDTISDSDDTIETLSIGLKLKILDKNIR



SLSILKHSLSFQTKYNKKDRNYYEDGNIHGKFFKKLGISHNQEEFNKSVY



APLFRYYSALYKLINDFEIYTLSLHIVGSETLTDQVNKSQFLSGRYFNFRKL



LTQSYHINNNSTHSTIFNAVINMRNDISHLSYEPLFDCPLNGKKSYKRKIRN



QFKTINIKPLVESRKIIIDFITLQTDMQKVLGYDAVNDFTMKIVQLRTRLKA



YANKEQTIQKMITEAKTPNDFYNIYKVQGVEEINKYLLEVIGETQAEKEIR






EKIERGNIANF


SEQ ID
MKKSIFDQFVNQYALSKTLRFELKPVGETGRMLEEAKVFAKDETIKKKYE


NO: 415
ATKPFFNKLHREFVEEALNEVELAGLPEYFEIFKYWKRYKKKFEKDLQKK



EKELRKSVVGFFNAQAKEWAKKYETLGVKKKDVGLLFEENVFAILKERY



GNEEGSQIVDESTGKDVSIFDSWKGFTGYFIKFQETRKNFYKDDGTATAL



ATRIIDQNLKRFCDNLLIFESIRDKIDFSEVEQTMGNSIDKVFSVIFYSSCLL



QEGIDFYNCVLGGETLPNGEKRQGINELINLYRQKTSEKVPFLKLLDKQIL



SEKEKFMDEIENDEALLDTLKIFRKSAEEKTTLLKNIFGDFVMNQGKYDL



AQIYISRESLNTISRKWTSETDIFEDSLYEVLKKSKIVSASVKKKDGGYAFP



EFIALIYVKSALEQIPTEKFWKERYYKNIGDVLNKGFLNGKEGVWLQFLLI



FDFEFNSLFEREIIDENGDKKVAGYNLFAKGFDDLLNNFKYDQKAKVVIK



DFADEVLHIYQMGKYFAIEKKRSWLADYDIDSFYTDPEKGYLKFYENAY



EEIIQVYNKLRNYLTKKPYSEDKWKLNFENPTLADGWDKNKEADNSTVI



LKKDGRYYLGLMARGRNKLFDDRNLPKILEGVENGKYEKVVYKYFPDQ



AKMFPKVCFSTKGLEFFQPSEEVITIYKNSEFKKGYTFNVRSMQRLIDFYK



DCLVRYEGWQCYDFRNLRKTEDYRKNIEEFFSDVAMDGYKISFQDVSES



YIKEKNQNGDLYLFEIKNKDWNEGANGKKNLHTIYFESLFSADNIAMNFP



VKLNGQAEIFYRPRTEGLEKERIITKKGNVLEKGDKAFHKRRYTENK VFF



HVPITLNRTKKNPFQFNAKINDFLAKNSDINVIGVDRGEKQLAYFSVISQR



GKILDRGSLNVINGVNYAEKLEEKARGREQARKDWQQIEGIKDLKKGYIS



QVVRKLADLAIQYNAIIVFEDLNMRFKQIRGGIEKSVYQQLEKALIDKLTF



LVEKEEKDVEKAGHLLKAYQLAAPFETFQKMGKQTGIVFYTQAAYTSRI



DPVTGWRPHLYLKYSSAEKAKADLLKFKKIKFVDGRFEFTYDIKSFREQK



EHPKATVWTVCSCVERFRWNRYLNSNKGGYDHYSDVTKFLVELFQEYGI



DFERGDIVGQIEVLETKGNEKFFKNFVFFFNLICQIRNTNASELAKKDGKD



DFILSPVEPFFDSRNSEKFGEDLPKNGDDNGAFNIARKGLVIMDKITKFAD



ENGGCEKMKWGDLYVSNVEWDNFVANK





SEQ ID
MFNNFIKKYSLQKTLRFELKPVGETADYIEDFKSEYLKDTVLKDEQRAKD


NO: 416
YQEIKTLIDDYHREYIEECLREPVDKKTGEILDFTQDLEDAFSYYQKLKEN



PTENRVGWEKEQESLRKKLVTSFVGNDGLFKKEFITRDLPEWLQKKGLW



GEYKDTVENFKKFTTYFSGFHENRKNMYTAEAQSTAIANRLMNDNLPKF



FNNYLAYQTIKEKHPDLVFRLDDALLQAAGVEHLDEAFQPRYFSRLFAQS



GITAFNELIGGRTTENGEKIQGLNEQINLYRQQNPEKAKGFPRFMPLFKQIL



SDRETHSFLPDAFENDKELLQALRDYVDAATSEEGMISQLNKAMNQFVT



ADLKRVYIKSAALTSLSQELFHFFGVISDAIAWYAEKRLSPKKAQESFLKQ



EVYAIEELNQAVVGYIDQLEDQSELQQLLVDLPDPQKPVSSFILTHWQKSQ



EPLQAVIAKVEPLFELEELSKNKRAPKHDKDQGGEGFQQVDAIKNMLDA



FMEVSHAIKPLYLVKGRKAIDMPDVDTGFYADFAEAYSAYEQVTVSLYNK



TRNHLSKKPFSKDKIKINFDAPTLLNGWDLNKESDNKSIILRKDGNFYLAI



MHPKHTKVFDCYSASEAAGKCYEKMNYKLLSGANKMLPKVFFSKKGIE



TFSPPQEILDLYKNNEHKKGATFKLESCHKLIDFFKRNIPKYKVHPTDNFG



WDVFGFHFSPTSSYGDLSGFYREVEAQGYKLWFSDVSEAYINKCVEEGK



LFLFQIYNKDFSPNSTGKPNLHTLYWKGLFEPENLKDVVLKLNGEAEIFY



RKHSIKHEDKTIHRAKDPIANKNADNPKKQSVFDYDIIKDKRYTQDKFFF



HVPISLNFKSQGVVRFNDKINGLLAAQDDVHVIGIDRGERHLLYYTVVNG



KGEVVEQGSLNQVATDQGYVVDYQQKLHAKEKERDQARKNWSTIENIK



ELKAGYLSQVVHKLAQLIVKHNAIVCLEDLNFGFKRGRFKVEKQVYQKF



EKALIDKLNYLVFKERGATQAGGYLNAYQLAAPFESFEKLGKQTGILYYV



RSDYTSKIDPATGFVDFLKPKYESMAKSKVFFESFERIQWNQAKGYFEFEF



DYKKMCPSRKFGDYRTRWVVCTFGDTRYQNRRNKSSGQWETETIDVTA



QLKALFAAYGITYNQEDNIKDAIAAVKYTKFYKQLYWLLRLTLSLRHSVT



GTDEDFILSPVADENGVFFDSRKATDKQPKDADANGAYHIALKGLWNLQ



QIR QHDWNVEKPKKLNLAMKNEEWFGFAQKKKFRA





SEQ ID
MIKNPSNRYALPKVIISKIDNQNILEFKIKYKKLSKLDIVKVKSMHYDDRAI


NO: 417
IFDEVIVNDGLIDVEYRDNHKTIFVKVGNKSYSISGQKVGGKERLLENRVS



KTKVQLELKDKATNRVSKTERELIVDDNIKIYSQIVGRDVKTTKDIYLIKR



FLAYRSDLLFYYGFVNNFFHVANNRSEFWKIDFNDSNNSKLIEYFKFTIND



HLKNDENYLKDYISDNEKLKNDLIKVKNSFEKIRHALMHFDYDFFVKLF



NGEDVGLELDIEFLDIMIDKLDKLNIDTKKEFIDDEKITIFGEELSLAKLYRF



YAHTAINRVAFNKLINSFIIENGVENQSLKEYFNQQAGGIAYEIDIHQNREY



KNLYNEHKKLVSRVLSISDGQEIAILNQKIAKLKDQMKQITKANSIKRLEY



KLRLALGFIYTEYENYEEFKNNFDTDIKNGRFTPKDNDGNKRAFDSRELE



QLKGYYEATIQTQKPKTDEKIEEVSKKIDRLSLKSLIADDILLKFILLMFTF



MPQELKGEFLGFIKKYYHDTKHIDQDTISDSDDTIETLSIGLKLKILDKNIR



SLSILKHSLSFQTKYNKKDRNYYEDGNIHGKFFKKLGISHNQEEFNKSVY



APLFRYYSALYKLINDFEIYTLSLHIVGSETLTDQVNKSQFLSGRYFNFRKL



LTQSYHINNNSTHSTIFNAVINMRNDISHLSYEPLFDCPLNGKKSYKRKIRN



QFKTINIKPLVESRKIIIDFITLQTDMQKVLGYDAVNDFTMKIVQLRTRLKA



YANKEQTIQKMITEAKTPNDFYNIYKVQGVEEINKYLLEVIGETQAEKEIR



EKIERGNIANF





SEQ ID
MIKNPSNRHSLPKVIISEVDHEKILEFKIKYEKLARLDRFEVKAMHYEGKE


NO: 418
IVFDEVLVNGGLIEVEYQDDNKTLFVKVGEKSYSIRGKKVGGKQRLLEDR



VSKTKVQLELSDGVVDNKGNLRKSRTERELIVADNIKLYSQIVGREVTTT



KEIYLVKRFLAYRSDLLFYYSFVDNFFKVAGNEKELWKINFDDATSAQFM



GYIPFMVNDNLKNDNAYLKDYVRNDVQIKDDLKKVQTIFSALRHTLLHF



NYEFFEKLFNGEDVGFDFDIGFLNLLIENIDKLNIDAKKEFIDNEKIRLFGE



NLSLAKVYRLYSDICVNRVGFNKFINSMLIKDGVENQVLKAEFNRKFGGN



AYTIDIHSNQEYKRIYNEHKKLVIKVSTLKDGQAIRRGNKKISELKEQMKS



MTKKNSLARLECKMRLAFGFLYGEYNNYKAFKNNFDTNIKNSQFDVND



VEKSKAYFLSTYERRKPRTREKLEKVAKDIESLELKTVIANDTLLKFILLM



FVFMPQELKGDFLGFVKKYYHDVHSIDDDTKEQEEDVVEAMSTSLKLKI



LGRNIRSLTLFKYALSSQVNYNSTDNIFYVEGNRYGKIYKKLGISHNQEEF



DKTLVVPLLRYYSSLFKLMNDFEIYSLAKANPTAVSLQELVDDETSPYKQ



GNYFNFNKMLRDIYGLTSDEIKSGQVVFMRNKIAHFDTEVLLSKPLLGQT



KMNLQRKDIVSFIEARGDIKELLGYDAINDFRMKVIHLRTKMRVYSDKLQ



TMMDLLRNAKTPNDFYNVYKVKGVESINKHLLEVLAQTAEERTVEKQIR



DGNEKYDL





SEQ ID
MEEKMLKSYDYFTKLYSLQKTLRFELKPIGKTLEHIKNSGIIESDETLEEQ


NO: 419
YAIVKNIIDKLHRKHIDEALSLVDFTKHLDTLKTFQELYLKRGKTDKEKEE



LEKLSADLRKLIVSYLKGNVKEKTQHNLNPIKERFEILFGKELFTNEEFFLL



AENEKEKKAIQAFKGFTTYFKGFQENRKNMYSEEGNSTSIAYRIINENLPL



FIENIARFQKVMSTIEKTTIKKLEQNLKTELKKHNLPGIFTIEYFNNVLTQE



GISRYNTIIGGKTTHEGVKIQGLNEIINLYNQQSKDVKLPILKPLHKQILSEE



YSTSFKIKAFENDNEVLKAIDTFWNEHIEKSIHPVTGNKFNILSKIENLCDQ



LQKYKDKDLEKLFIERKNLSTVSHQVYGQWNIIRDALRMHLEMNNKNIK



EKDIDKYLDNDAFSWKEIKDSIKIYKEHVEDAKELNENGIIKYFSAMSINE



EDDEKEYSISLIKNINEKYNNVKSILQEDRTGKSDLHQDKEKVGIIKEFLDS



LKQLQWFLRLLYVTVPLDEKDYEFYNELEVYYEALLPLNSLYNKVRNYM



TRKPYSVEKFKLNFNSPTLLDGWDKNKETANLSIILRKNGKYYLGIMNKE



NNTIFEYYPGTKSNDYYEKMIYKLLPGPNKMLPKVFFSKKGLEYYNPPKE



ILNIYEKGEFKKDKSGNFKKESLHTLIDFYKEAIAKNEDWEVFNFKFKNT



KEYEDISQFYRDVEEQGYLITFEKVDANYVDKLVKEGKLYLFQIYNKDFS



ENKKSKGNPNLHTIYWKGLYDSENLKNVVYKLNGEAEVFYRKKSIDYPE



EIYNHGHHKEELLGKFNYPIIKDRRYTQDKFLFHVPITMNFISKEEKRVNQ



LACEYLSATKEDVHIIGIDRGERHLLYLSLIDKEGNIKKQLSLNTIKNENYD



KEIDYRVKLDEKEKKRDEARKNWDVIENIKELKEGYMSQVIHIIAKMMV



EEKAILIMEDLNIGFKRGRFKVEKQVYQKFEKMLIDKLNYLVFKNKNPLE



PGGSLNAYQLTSKFDSFKKLGKQSGFIFYVPSAYTSKIDPTTGFYNFIQVDV



PNLEKGKEFFSKFEKIIYNTKEDYFEFHCKYGKFVSEPKNKDNDRKTKES



LTYYNAIKDTVWVVCSTNHERYKIVRNKAGYYESHPVDVTKNLKDIFSQ



ANINYNEGKDIKPIIIESNNAKLLKSIAEQLKLILAMRYNNGKHGDDEKDY



ILSPVKNKQGKFFCTLDGNQTLPINADANGAYNIALKGLLLIEKIKKQQGK



IKDLYISNLEWFMFMMSR





SEQ ID
MNTQKKEFNPKSFKDFTNLYSLNKTLRFSLTPNKKTAEILEFNKQKEVKCF


NO: 420
SNDRKIAGAYQEIKKYLNKLHQEFIQEAMKFFAFSEEELKGFEKEYLNLL



NFTDKDNFKKKNKIRNEYEQERKILTIKIATYFSKFKSEKYQSFNLANITG



KKVFSILEQKYKEDKKTLKIIHIFKYKPTKDEKKEGEAVNFSTYLTGFNEN



RKNFYKSEDKAGQFATRTIDNLAQFIKNKKLFEDKYQKNYSKIGILDEQIK



IFNLDYFNNLFLQEGLDEYNGILGNNKGEENKSNEGINQKINIFKQKEKAR



LKKEKENFNKSDFPLFKELYKQIGSIRKENDVYVEIKTDKELVEELNNFPK



NVENYLKDIQSFYKTFFEKLQNEEYELDKIYLPKSVGTYFSYIAFSDWNK



LAFIYNKRYKNEKIKIVEGGDVNVQYRSLEVLKNRIDELKDEDNLNFNKF



FIDKLKFNEAKKENNWQNFWFCIEYYINSQFIGGEKNILNKEKNEYEILPF



GSLKELKEKYFEAVKKYKEKMVDTESGLIDDEEKEIKETLKNYLDRIKEI



ERIAKYFDLKKSFEEIKQEDLDSNFYGEYQKVVDKTNELKIYQYYSEFRN



YLTQNNSVEEKIKLNFNSGLLLDGWDLNKEKVKFSIIFQENGKYYLGIINK



EKDKTILDKDKHPEIFTKNSDFRKMEYKLFPSPSKMLPKISFSETAKKGDE



DVGWSEEIQKIKDEFAEFQEYKKKSKDNWKDEFNRGKLNKLIDYYKQVL



EKHSEGYMNTYNFELKDSSKYKNLGEFNDDIARQNYKVKFVGIDKNYID



EKVANGELFLFQIYNKDFSEDKKEGSTNNLETIYFKELFSKENLENPVFKL



SGGAEMFFRNKIEKKKEKKKLDKDGKPMISKKGEKVVDKRRFSENKILF



HLPIEINYGKGKMPNFNKKINEYISKNPENIKIIGIDRGEKHLLYYSIIDQNG



NNIESMSLNAVDEFGNFVNPEKLEEYEIDNNGKKERRWKYIVNDKEIKVT



NYQRKLDELEKERQKSRQSWQNINKIKNLKKGYISFVVKKIVDLAIENNA



IIILEDLNFGFKSFRQKIEKNVYQQFEKALIDKLGFVVDKQKQNQRFAPQL



SAPFESFQKIGKQTGIVYYVLANNTSKVCPSCQWIKNFYLKYEKKNTIFNL



QKNQKLKVFFEQEKNRFRFEYQMSKEYISVYSDVDRQRYDKTKNQNKG



GYLEYKNSNQKEIIDKDGVIQKQSITLQLKELFKENHIDLEKEILKQLDNK



KEKNSGYTGVYNKFIYLFNLILQIRNAISFREKDYIQCPSCHFDTRKENYL



KINDGDGNGAYNIALRGLYLLKGKNGIINNLEKIKLIFSNNDYFQWAKKL



KNKK





SEQ ID
MQNKQSFADFTNLYSLSKTLRFELKPIGQTQAMLDENKIFEVDENRKKAY


NO: 421
DKTKPYFDRLHREFINESLSNAQLKGISEYFETFKQFRSNQNNKDLKELIN



KQQKFLRHQIVTLFDENGKHWATTKYAHLKIKKKNLDILFDEQVFYILKE



RYGSEKETQLVDKETGAVTSIFDNWKGFTGYFTKFFETRKNFYKSDGTST



ALATRIIDQNLNRFFDNLETFHKIKDKIDVKEVEIFFKLKADNVFSIDFYNQ



CLLQNGIDKYNDFLGGQTLENGEKQKGINEIINKYRQDNKDQKLPFLKKL



DKQILSEKDRFINEIESKEEFFQVLTEFYQSATVKVTIIKTLLNDFVHNTDK



YKLEKIYLTKEAFNTIANKWTDETQIFEDNLDLVLKNKKITAKQDFIPLAYI



KEALEVIEKDRKFFKDRYYNDPQIGFFPDQSYWEQFLAILNFEFMTHFQR



VAKDKITGKKIELGYFVFEKRIKELLDSDPSLNSQSKIIIKEFADEVLHIFQM



AKYFALEKKREWKGDYYQLDDQFYNHIDYGFKDQFYENAYEKIVQPYN



KIRNYLTKKPYSDVKWKLNFGNPTLANGWDKNKEADNTAVILKKDGNY



YLGVMKKGKNKIFSDQNKEKYKAYNSAYYEKLVYKLFPDPSKMFPKVCF



SKKGLNFFQPSEEILRIYKNNEFKKGNTFSISSMQKLIAFYIDCLGLYEGWK



HYEFKNIKDVRQYKENIGEFYADVAESGYKLWFEKISEEYITQKNQLGELF



LFQIYNKDFAKKTTGRKNLHTIYFEELFSQTNIDNNFPFKLNGQAELFYRP



KSLEKIEEKRNFKRSIVNKKRYTQNKIFFHVPITLNRTSENIGRFNVRVNNF



LANNSNVNIVGVDRGEKNLAYYSIIKQNGEVLKSGSLNIINGVDYHALLT



DRAQRREQERRNWQDVESIKDLKRGYISQVVHELVSLAIKYNAIIVMEDL



NMRFKQIRGGIEKSTYQQLEKALIEKLNFLVNKEETDSNQAGNLLNAYQL



TAPFKTFKDMGKQTGIIFYTQASYTSKIDPLTGWRPNIYLRYSNAKQAKA



DILMFTNIYFSEKKDRFEFTYDLEKIDDKRKDLPIKTEWTVCSNVERFSWE



KSLNNNKGGYVHYPIQDSNGEESITSKLKKLFMDFGIDLTDIKTQIESLDT



NKKDNANFFRKFIFYFQLICQIRNTQVNKSDDGNDFIFSPVEPFFDSRFAD



KFRKNLPKNGDENGAYNIARKGLIILHKISDYFVKEGSTDKISWKDLSISQ



TEWDNFTTDK





SEQ ID
MKKEKEFKSFGDFTNLYEISKTLRFELKPVENTQTMLDEADVFGKDKVIK


NO: 422
DKYTKTKPFIDKLHREFVDESLKDVSLSGLKKYSEVLENWKKNKKDKDI



VKELKKEEERLRKEVVEFFDNTAKKWANEKYKELGLKKKDIGILFEESVF



DLLKEKYGEEQDSFLKEEKGDFLKNEKGEKVSIFDEWKGFVGYFTKFQE



TRKNFYKNDGTETALATRIIDQNLKRFCDNIDDFKKIKNKIDFSEVEKNFN



KTADVFSLDFYNQCLLQKGIDSYNEFIGGKTLENGKKLKGVNELVNEYR



QKNKNEKVSFLKLLDKQILSEKEKLSFGIENDEQLLVVLNSFYETAEEKTK



ILRTLFGDFVEHNENYDLDKTYISKVAFNTISHKWTNETHKFEELLYGAM



KEDKPIGLNYDKKEDSYKFPDFIALGYLKKCLNNLDCDTKFWKEKYYEN



NADKKDKDKGFLTGGQNAWDQFLQIFIFEFNQLFNSEAFDNKGKEIKIGY



DNFRKDFEEIINQKDFKNDENLKIAIKNFADSVLWIYQMAKYFAIEKKRG



WDDDFELSEFYTNPSNGYSLFYDRAYEEIVQKYNDLRNYLTKKPYKEDK



WKLNFENPTLANGFDKNKESDNSTVILRKKRKYYLGLMKKGNNKIFEDR



NKAEFIRNIESGAYEKMAYKYLPDVAKMIPKCSTQLNEAKNHFRNSADDL



EIKKSFSNPLKITKRIFDLNNIQYDKTNVSKKISGDNKGIKIFQKEYYKISG



DFDVYKSALNDWIDFCKDFLSKYDSTKDFDFSILRKTKDYKSLDEFYVDV



AKITYKISFTPVSESYIDQKNKNGELYLFEIYNQDFAKGKMGAKNLHTLYF



ENVFSPENISKNFPIKLNGNAELFFRPKSIESKKEKRNFVREIVNKKRYSED



KIFFHCPITLNRETGSIYRENNYVNNFLSENNINIIGVDRGEKHLAYYSVID



KNGVKIGGGSFNEINKVDYAKKLEERAGEREQSRKDWQVVEGIKDLKKG



YISQVVRELADLAIKHNAIIVLEDLNMRFKQIRGGIEKSIYQQLEKALIDKL



SFLVEKGEKDPNQAGHILKAYQLAAPFTSFKDMGKQTGIVFYTQASYTSK



TCPNCGFRKNNNKFYFENNIGKAQDALKKLKTFEYDSENKCFGLSYCLS



DFANKEEVEKNKNKKRNNAPYSDIEKKDCFELSTKDAVRYRWHDKNTER



GKTFFEGESVYEEKEEKEIGQTKRGLVKEYDISKCLIGLFEKTGLDYKQNL



LDKINSGKFDGTFYKNLFNYLNLLFEIRNSISGTEIDYISCPECQFHTDKSK



TIKNGDDNGSYNIARKGMIILDKIKQFKKENGSLDKMGWGELFIDLEEWD



KFAQKKNNNIIDK





SEQ ID
MKYTDFTGIYSVSKTLRFELIPQGSTVENMKREGILNNDMHRADSYKEM


NO: 423
KKLIDEYHKAFIERCLSDFSLKYDDTGKHDSLEEYFFYYEQKRNDKTKKI



FEDIQVALRKQISKRFTGDTAFKRLFKKELIKEDLPSFVKNDPVKTELIKEF



SDFTTYFQEFHKNRKNMYTSDAKSTAIAYRIINENLPKFIDNINAFDIVAKV



PEMQEHFKTIADELRSHLQVGNDIDKMFNLQFFNKVLTQSQLDVYNAVIG



GKSEGNKKIQGINEYINLYNQQHKKARLPMLKLLYKQILSDRVAISWLQD



EFDNDQDMLDTIEAFYNKLNSNETGVLGEGKLKQILMGLDGYNLDGVF



MRNDLQLSEVSQRLCGGWNIIKDAMTSDLKRSVQKKKKETDADFEERVS



KLFSAQNSFSIAYINQCLGQAGIRCKIQDYFACLGAKEGENEAETTPDIFD



QIAEAYHGAAPILNARPSSHNLAQDIEKVKAIKALLDALKRLQRFVKPLL



GRGDEGDKDNFFYGDFMPIWEVLDQLTPLYNKVRNRMTRKPYSQEKIKL



NFENSTLLNGWDLNKEHDNTSVILRREGLYYLGIMNKNYNKIFDANNVE



TIGDCYEKMIYKLLPGPNKMLPKVFFSKSRVQEFSPSKKILEIWESKSFKK



GDNFNLDDCHALIDFYKDSIAKHPDWNKFNFKFSDTQSYTNISDFYRDVN



QQGYSLSFTKVSVDYVNRMVDEGKLYLFQIYNKDFSPQSKGTPNMHTLY



WRMLFDERNLHNVIYKLNGEAEVFYRKASLRCDRPTHPAHQPITCKNEN



DSKRVCVFDYDIIKNRRYTVDKFMFHVPITINYKCTGSDNINQQVCDYLR



SAGDDTHIIGIDRGERNLLYLVIIDQHGTIKEQFSLNEIVNEYKGNTYCTNY



HSLLEEKEAGNKKARQDWQTIESIKELKEGYLSQVIHKISMLMQRYHAIV



VLEDLNGSFMRSRQKVEKQVYQKFEHMLINKLNYLVNKQYDATEPGGL



LHALQLTSRMDSFKKLGKQSGFLFYIPAWNTSKIDPVTGFVNLFDTRYCN



EAKAKEFFEKFDDISYNDERDWFEFSFDYRHFTNKPTGTRTQWTLCTQGT



RVRTFRNPEKSNHWDNEEFDLTQAFKDLFNKYGIDIASGLKARIVNGQLT



KETSAVKDFYESLLKLLKLTLQMRNSVTGTDIDYLVSPVADKDGIFFDSRT



CGSLLPANADANGAFNIARKGLMLLRQIQQSSIDAEKIQLAPIKNEDWLEF



AQEKPYL









The programmable nuclease can be Cas13. Sometimes the Cas13 can be Cas13a, Cas13b, Cas13c, Cas13d, or Cas13e. In some cases, the programmable nuclease can be Mad7 or Mad2. In some cases, the programmable nuclease can be Cas12. Sometimes the Cas12 can be Cas12a, Cas12b, Cas12c, Cas12d, or Cas12e. In some cases, the programmable nuclease can be Csm1, Cas9, C2c4, C2c8, C2c5, C2c10, C2c9, or CasZ. Sometimes, the Csm1 can also be also called smCms1, miCms1, obCms1, or suCms1. Sometimes Cas13a can also be also called C2c2. Sometimes CasZ can also be called Cas14a, Cas14b, Cas14c, Cas14d, Cas14e, Cas14f, Cas14g, Cas14h, Cas14i, Cas14j, or Cas14k. Sometimes, the programmable nuclease can be a type V CRISPR-Cas system. In some cases, the programmable nuclease can be a type VI CRISPR-Cas system. Sometimes the programmable nuclease can be a type III CRISPR-Cas system. In some cases, the programmable nuclease can be from at least one of Leptotrichia shahii (Lsh), Listeria seeligeri (Lse), Leptotrichia buccalis (Lbu), Leptotrichia wadeu (Lwa), Rhodobacter capsulatus (Rca), Herbinix hemicellulosilytica (Hhe), Paludibacter propionicigenes (Ppr), Lachnospiraceae bacterium (Lba), [Eubacterium] rectale (Ere), Listeria newyorkensis (Lny), Clostridium aminophilum (Cam), Prevotella sp. (Psm), Capnocytophaga canimorsus (Cca, Lachnospiraceae bacterium (Lba), Bergeyella zoohelcum (Bzo), Prevotella intermedia (Pin), Prevotella buccae (Pbu), Alistipes sp. (Asp), Riemerella anatipestifer (Ran), Prevotella aurantiaca (Pau), Prevotella saccharolytica (Psa), Prevotella intermedia (Pin2), Capnocytophaga canimorsus (Cca), Porphyromonas gulae (Pgu), Prevotella sp. (Psp), Porphyromonas gingivalis (Pig), Prevotella intermedia (Pin3), Enterococcus italicus (Ei), Lactobacillus salivarius (Ls), or Thermus thermophilus (Tt). Sometimes the Cas13 is at least one of LbuCas13a, LwaCas13a, LbaCas13a, HheCas13a, PprCas13a, EreCas13a, CamCas13a, or LshCas13a. The trans cleavage activity of the CRISPR enzyme can be activated when the crRNA is complexed with the target nucleic acid. The trans cleavage activity of the CRISPR enzyme can be activated when the guide nucleic acid comprising a tracrRNA and crRNA are complexed with the target nucleic acid. The target nucleic acid can be RNA or DNA.


In some embodiments, a programmable nuclease as disclosed herein is an RNA-activated programmable RNA nuclease. In some embodiments, a programmable nuclease as disclosed herein is a DNA-activated programmable RNA nuclease. In some embodiments, a programmable nuclease is capable of being activated by a target RNA to initiate trans cleavage of an RNA reporter and is capable of being activated by a target DNA to initiate trans cleavage of an RNA reporter, such as a Type VI CRISPR/Cas enzyme (e.g., Cas13). For example, Cas13a of the present disclosure can be activated by a target RNA to initiate trans cleavage activity of the Cas13a for the cleavage of an RNA reporter and can be activated by a target DNA to initiate trans cleavage activity of the Cas13a for trans cleavage of an RNA reporter. An RNA reporter can be an RNA-based reporter molecule. In some embodiments, the Cas13a recognizes and detects ssDNA to initiate transcleavage of RNA reporters. Multiple Cas13a isolates can recognize, be activated by, and detect target DNA, including ssDNA, upon hybridization of a guide nucleic acid with the target DNA. For example, Lbu-Cas13a and Lwa-Cas13a can both be activated to transcollaterally cleave RNA reporters by target DNA. Thus, Type VI CRISPR/Cas enzyme (e.g., Cas13, such as Cas13a) can be DNA-activated programmable RNA nucleases, and therefore, can be used to detect a target DNA using the methods as described herein. DNA-activated programmable RNA nuclease detection of ssDNA can be robust at multiple pH values. For example, target ssDNA detection by Cas13 can exhibit consistent cleavage across a wide range of pH conditions, such as from a pH of 6.8 to a pH of 8.2. In contrast, target RNA detection by Cas13 may exhibit high cleavage activity of pH values from 7.9 to 8.2. In some embodiments, a DNA-activated programmable RNA nuclease that also is capable of being an RNA-activated programmable RNA nuclease, can have DNA targeting preferences that are distinct from its RNA targeting preferences. For example, the optimal ssDNA targets for Cas13a have different properties than optimal RNA targets for Cas13a. As one example, gRNA performance on ssDNA may not necessarily correlate with the performance of the same gRNAs on RNA. As another example, gRNAs can perform at a high level regardless of target nucleotide identity at a 3′ position on a target RNA sequence. In some embodiments, gRNAs can perform at a high level in the absence of a G at a 3′ position on a target ssDNA sequence. Furthermore, target DNA detected by Cas13 disclosed herein can be directly from organisms, or can be indirectly generated by nucleic acid amplification methods, such as PCR and LAMP or any amplification method described herein. Key steps for the sensitive detection of a target DNA, such as a target ssDNA, by a DNA-activated programmable RNA nuclease, such as Cas13a, can include: (1) production or isolation of DNA to concentrations above about 0.1 nM per reaction for in vitro diagnostics, (2) selection of a target sequence with the appropriate sequence features to enable DNA detection as these features are distinct from those required for RNA detection, and (3) buffer composition that enhances DNA detection. The detection of a target DNA by a DNA-activated programmable RNA nuclease can be connected to a variety of readouts including fluorescence, lateral flow, electrochemistry, or any other readouts described herein. Multiplexing of programmable DNA nuclease, such as a Type V CRISPR-Cas protein, with a DNA-activated programmable RNA nuclease, such as a Type VI protein, with a DNA reporter and an RNA reporter, can enable multiplexed detection of target ssDNAs or a combination of a target dsDNA and a target ssDNA, respectively. Multiplexing of different RNA-activated programmable RNA nucleases that have distinct RNA reporter cleavage preferences can enable additional multiplexing. Methods for the generation of ssDNA for DNA-activated programmable RNA nuclease-based diagnostics can include (1) asymmetric PCR, (2) asymmetric isothermal amplification, such as RPA, LAMP, SDA, etc. (3) NEAR for the production of short ssDNA molecules, and (4) conversion of RNA targets into ssDNA by a reverse transcriptase followed by RNase H digestion. Thus, DNA-activated programmable RNA nuclease detection of target DNA is compatible with the various systems, kits, compositions, reagents, and methods disclosed herein.


Engineered Programmable Nucleases

Disclosed herein are non-naturally occurring compositions and systems comprising at least one of an engineered Cas protein and an engineered guide nucleic acid, which may simply be referred to herein as a Cas protein and a guide nucleic acid, respectively. In general, an engineered Cas protein and an engineered guide nucleic acid refer to a Cas protein and a guide nucleic acid, respectively, that are not found in nature. In some instances, systems and compositions comprise at least one non-naturally occurring component. For example, compositions and systems may comprise a guide nucleic acid, wherein the sequence of the guide nucleic acid is different or modified from that of a naturally-occurring guide nucleic acid. In some instances, compositions and systems comprise at least two components that do not naturally occur together. For example, compositions and systems may comprise a guide nucleic acid comprising a repeat region and a spacer region which do not naturally occur together. Also, by way of example, composition and systems may comprise a guide nucleic acid and a Cas protein that do not naturally occur together. Conversely, and for clarity, a Cas protein or guide nucleic acid that is “natural,” “naturally-occurring,” or “found in nature” includes Cas proteins and guide nucleic acids from cells or organisms that have not been genetically modified by a human or machine.


In some instances, the guide nucleic acid may comprise a non-natural nucleobase sequence. In some instances, the non-natural sequence is a nucleobase sequence that is not found in nature. The non-natural sequence may comprise a portion of a naturally occurring sequence, wherein the portion of the naturally occurring sequence is not present in nature absent the remainder of the naturally-occurring sequence. In some instances, the guide nucleic acid may comprise two naturally occurring sequences arranged in an order or proximity that is not observed in nature. In some instances, compositions and systems comprise a ribonucleotide complex comprising a CRISPR/Cas effector protein and a guide nucleic acid that do not occur together in nature. Engineered guide nucleic acids may comprise a first sequence and a second sequence that do not occur naturally together. For example, an engineered guide nucleic acid may comprise a sequence of a naturally occurring repeat region and a spacer region that is complementary to a naturally occurring eukaryotic sequence. The engineered guide nucleic acid may comprise a sequence of a repeat region that occurs naturally in an organism and a spacer region that does not occur naturally in that organism. An engineered guide nucleic acid may comprise a first sequence that occurs in a first organism and a second sequence that occurs in a second organism, wherein the first organism and the second organism are different. The guide nucleic acid may comprise a third sequence disposed at a 3′ or 5′ end of the guide nucleic acid, or between the first and second sequences of the guide nucleic acid. For example, an engineered guide nucleic acid may comprise a naturally occurring crRNA and tracrRNA coupled by a linker sequence.


In some instances, compositions and systems described herein comprise an engineered Cas protein that is similar to a naturally occurring Cas protein. The engineered Cas protein may lack a portion of the naturally occurring Cas protein. The Cas protein may comprise a mutation relative to the naturally-occurring Cas protein, wherein the mutation is not found in nature. The Cas protein may also comprise at least one additional amino acid relative to the naturally-occurring Cas protein. For example, the Cas protein may comprise an addition of a nuclear localization signal relative to the natural occurring Cas protein. In certain embodiments, the nucleotide sequence encoding the Cas protein is codon optimized (e.g., for expression in a eukaryotic cell) relative to the naturally occurring sequence.


In some instances, compositions and systems provided herein comprise a multi-vector system encoding a Cas protein and a guide nucleic acid described herein, wherein the guide nucleic acid and the Cas protein are encoded by the same or different vectors. In some embodiments, the engineered guide and the engineered Cas protein are encoded by different vectors of the system.


Thermostable Programmable Nuclease

Described herein are various embodiments of thermostable programmable nucleases. In some embodiments, a programmable nuclease is referred to as an effector protein. An effector protein may be thermostable. In some instances, known effector proteins (e.g., Cas12 nucleases) are relatively thermo-sensitive and only exhibit activity (e.g., cis and/or trans cleavage) sufficient to produce a detectable signal in a diagnostic assay at temperatures less than 40° C., and optimally at about 37° C. A thermostable protein may have enzymatic activity, stability, or folding comparable to those at 37° C. In some instances, the trans cleavage activity (e.g., the maximum trans cleavage rate as measured by fluorescent signal generation) of an effector protein in a trans cleavage assay at 40° ° C. may be at least 50% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 55% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 60% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 65% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 70% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 75% of that at 37° ° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 80% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 85% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 90% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 95% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 100% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° ° C. may be at least 1-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 2-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 3-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 4-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° ° C. may be at least 5-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 6-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 7-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 8-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 9-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 10-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 40° C. may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that at 37° C.


In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 50% of that at 37° ° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 55% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 60% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 65% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 70% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 75% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 80% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 85% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° ° C. may be at least 90% of that at 37° ° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 95% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 100% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 1-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 2-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 3-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 4-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 5-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 6-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 7-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 8-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 9-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 10-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 45° C. may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that at 37° C.


In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 50% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 55% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° ° C. may be at least 60% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 65% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 70% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 75% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 80% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 85% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 90% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° ° C. may be at least 95% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 100% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 1-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° ° C. may be at least 2-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 3-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 4-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° ° C. may be at least 5-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 6-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 7-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 8-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 9-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° C. may be at least 10-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 50° ° C. may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that at 37° C.


In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 50% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 55% of that at 37° ° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 60% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 65% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 70% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 75% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 80% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 85% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 90% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 95% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 100% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 1-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 2-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 3-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 4-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 5-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 6-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 7-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 8-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 9-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 10-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 55° C. may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that at 37° C.


In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° ° C. may be at least 50% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 55% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 60% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 65% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 70% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° ° C. may be at least 75% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 80% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 85% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 90% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 95% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° ° C. may be at least 100% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 1-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 2-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° ° C. may be at least 3-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 4-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 5-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 6-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° ° C. may be at least 7-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 8-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 9-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 10-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 60° C. may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that at 37° C.


In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 50% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 55% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 60% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 65% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 70% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 75% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 80% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 85% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 90% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 95% of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° ° C. may be at least 100% of that at 37° ° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 1-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 2-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 3-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 4-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° ° C. may be at least 5-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 6-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 7-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 8-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 9-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 10-fold of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 65° C. may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that at 37° C. In some instances, the trans cleavage activity of an effector protein in a trans cleavage assay at 70° C., 75° C. 80° C., or more may be at least 50, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 95%, at least 100%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that at 37° C.


In some instances, the trans cleavage activity may be measured against a negative control in a trans cleavage assay. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 37° C. may be at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 37° C. may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 40° C. may be at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 40° C. may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 45° C. may be at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 45° C. may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 50° ° C. may be at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 50° C. may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 55° C. may be at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 55° C. may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 60° C. may be at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 60° C. may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 65° C. may be at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 65° C. may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 70° C., 75° C., 80° C., or more may be at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold of that against a negative control nucleic acid. In some instances, the trans cleavage activity of an effector protein against a nucleic acid in a trans cleavage assay at 70° C., 75° C., 80° ° C., or more may be at least 11-fold, at least 12-fold, at least 13-fold, at least 14-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold or more of that against a negative control nucleic acid.


Reporters

Described herein are reagents comprising a reporter comprising a detection moiety, wherein the reporter is capable of being cleaved by the activated nuclease, thereby generating a first detectable signal. As used herein, a reporter is used interchangeably with reporter molecule. In some cases, the reporter comprises a single-stranded nucleic acid comprising deoxyribonucleotides. In other cases, the reporter comprises a single-stranded nucleic acid comprising ribonucleotides. The reporter can comprise a single-stranded nucleic acid comprising at least one deoxyribonucleotide and at least one ribonucleotide. In some cases, the reporter comprises a single-stranded nucleic acid comprising at least one ribonucleotide residue at an internal position that functions as a cleavage site. In some cases, the reporter comprises at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 ribonucleotide residues at an internal position. In some cases, the reporter may comprise from 2 to 10, from 3 to 9, from 4 to 8, or from 5 to 7 ribonucleotide residues at an internal position. In some cases, the reporter may comprise from 3 to 10, from 4 to 10, from 5 to 10, from 6 to 10, from 7 to 10, from 8 to 10, from 9 to 10, from 2 to 8, from 3 to 8, from 5 to 8, from 6 to 8, from 7 to 8, from 2 to 5, from 3 to 5, or from 4 to 5 ribonucleotide residues at an internal position. Sometimes the ribonucleotide residues are continuous. Alternatively, the ribonucleotide residues are interspersed in between non-ribonucleotide residues. In some cases, the reporter has only ribonucleotide residues. In some cases, the reporter has only deoxyribonucleotide residues. In some cases, the reporter comprises nucleotides resistant to cleavage by the programmable nuclease described herein. In some cases, the reporter comprises synthetic nucleotides. In some cases, the reporter comprises at least one ribonucleotide residue and at least one non-ribonucleotide residue. In some cases, the reporter is 5-20, 5-15, 5-10, 7-20, 7-15, or 7-10 nucleotides in length. In some cases, the reporter comprises at least one uracil ribonucleotide. In some cases, the reporter comprises at least two uracil ribonucleotides. Sometimes the reporter has only uracil ribonucleotides. In some cases, the reporter comprises at least one adenine ribonucleotide. In some cases, the reporter comprises at least two adenine ribonucleotide. In some cases, the reporter has only adenine ribonucleotides. In some cases, the reporter comprises at least one cytosine ribonucleotide. In some cases, the reporter comprises at least two cytosine ribonucleotide. In some cases, the reporter comprises at least one guanine ribonucleotide. In some cases, the reporter comprises at least two guanine ribonucleotide. A reporter can comprise only unmodified ribonucleotides, only unmodified deoxyribonucleotides, or a combination thereof. In some cases, the reporter is from 5 to 12 nucleotides in length. In some cases, the reporter is at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length. In some cases, the reporter is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length. For cleavage by a programmable nuclease comprising Cas13, a reporter can be 5, 8, or 10 nucleotides in length. For cleavage by a programmable nuclease comprising Cas12, a reporter can be 10 nucleotides in length. A DETECTR reaction can comprise 100 pM to 1 nM of reporters. A DETECTR reaction can comprise 1 nM to 5 nM of reporters. A DETECTR reaction can comprise 5 nM to 20 nM of reporters. A DETECTR reaction can comprise 20 nM to 50 nM of reporters. A DETECTR reaction can comprise 50 nM to 100 nM of reporters. A DETECTR reaction can comprise 100 nM to 250 nM reporters. A DETECTR reaction can comprise 250 nM to 500 nM reporters. A DETECTR reaction can comprise 500 nM to 1000 nM (1 μM) of reporters.


The reporter can comprise a detection moiety (in addition to the nucleic acid) capable of generating a first detectable signal. Sometimes the reporter may comprise a protein capable of generating a signal. A signal can be a colorimetric, potentiometric, amperometric, optical (e.g., fluorescent, colorimetric, etc.), or piezo-electric signal. The generation of the detectable signal from the release of the detection moiety indicates that cleavage by the programmable nuclease has occurred and that the sample contains the target nucleic acid. A detection moiety can be any moiety capable of generating a colorimetric, potentiometric, amperometric, optical (e.g., fluorescent, colorimetric, etc.), or piezo-electric signal. A reporter, sometimes, is protein-nucleic acid that can generate a calorimetric, potentiometric, amperometric, optical (e.g., fluorescent, colorimetric, etc.), or piezo-electric signal upon cleavage of the nucleic acid. Often a calorimetric signal is heat produced after cleavage of the reporters. Sometimes, a calorimetric signal is heat absorbed after cleavage of the reporters. A potentiometric signal, for example, is electrical potential produced after cleavage of the reporters. An amperometric signal can be movement of electrons produced after the cleavage of a reporter. Often, the signal is an optical signal, such as a colorimetric signal or a fluorescence signal. An optical signal is, for example, a light output produced after the cleavage of the reporters. Sometimes, an optical signal is a change in light absorbance between before and after the cleavage of reporters. Often, a piezo-electric signal is a change in mass between before and after the cleavage of the reporter.


Detecting the presence or absence of a target nucleic acid of interest can involve measuring a signal emitted from a detection moiety present in a reporter, after cleavage of the reporter by an activated programmable nuclease. The signal can be measured using one or more sensors integrated with the device or operatively coupled to the device. Thus, the detecting steps disclosed herein can involve measuring the presence of a target nucleic acid, quantifying how much of the target nucleic acid is present, or, measuring a signal indicating that the target nucleic acid is absent in a sample. In some embodiments, a signal is generated upon cleavage of the reporter by the programmable nuclease. In other embodiments, the signal changes upon cleavage of the reporter by the programmable nuclease. In other embodiments, a signal can be present in the absence of reporter cleavage and disappear upon cleavage of the target nucleic acid by the programmable nuclease. For example, a signal can be produced in a microfluidic device or lateral flow device after contacting a sample with a composition comprising a programmable nuclease.


In some cases, the signal can comprise a colorimetric signal or a signal visible by eye. In some instances, the signal is fluorescent, electrical, chemical, electrochemical, or magnetic. A signal can be a calorimetric, potentiometric, amperometric, optical (e.g., fluorescent, colorimetric, etc.), or piezo-electric signal. In some cases, the detectable signal is a colorimetric signal or a signal visible by eye. In some instances, the detectable signal is fluorescent, electrical, chemical, electrochemical, or magnetic. In some cases, the first detection signal is generated by binding of the detection moiety to the capture molecule in the detection region, where the first detection signal indicates that the sample contained the target nucleic acid. Sometimes the system can detect more than one type of target nucleic acid, wherein the system may comprise more than one type of guide nucleic acid and more than one type of reporter. In some cases, the detectable signal is generated directly by the cleavage event. Alternatively, or in combination, the detectable signal is generated indirectly by the signal event. Sometimes the detectable signal is not a fluorescent signal. In some instances, the detectable signal is a colorimetric or color-based signal. In some cases, the detected target nucleic acid is identified based on its spatial location on the detection region of the support medium.


The reporter can comprise a quenching moiety on the other side of the cleavage site. Sometimes the quenching moiety is a fluorescence quenching moiety. In some cases, the quenching moiety is 5′ to the cleavage site and the detection moiety is 3′ to the cleavage site. In some cases, the detection moiety is 5′ to the cleavage site and the quenching moiety is 3′ to the cleavage site. Sometimes the quenching moiety is at the 5′ terminus of the nucleic acid of the reporter. Sometimes the detection moiety is at the 3′ terminus of the nucleic acid of the reporter. In some cases, the detection moiety is at the 5′ terminus of the nucleic acid of the reporter. In some cases, the quenching moiety is at the 3′ terminus of the nucleic acid of the reporter. In some cases, the reporter comprises at least one population of the single-stranded nucleic acid capable of generating a first detectable signal. In some cases, the reporter comprises a population of the single-stranded nucleic acid capable of generating a first detectable signal. Optionally, there is more than one population of reporter. In some cases, there are 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 30, 40, 50, or greater than 50, or any number spanned by the range of this list of different populations of reporters capable of generating a detectable signal.


In some cases, the one or more detectable signals generated after cleavage can produce an index of refraction change or one or more electrochemical changes. In some cases, real-time detection of the Cas reaction can be achieved using fluorescence, electrochemical detection, and/or electrochemiluminescence. In some cases, the detectable signals can be detected and analyzed in various ways. For example, the detectable signals can be detected using an imaging device. The imaging device can comprise a digital camera, such as a digital camera on a mobile device. The mobile device can have a software program or a mobile application that can capture fluorescence, ultraviolet (UV), infrared (IR), or visible wavelength signals. Any suitable detection or measurement device can be used to detect and/or analyze the colorimetric, fluorescence, amperometric, potentiometric, or electrochemical signals described herein. In some embodiments, the colorimetric, fluorescence, amperometric, potentiometric, or another electrochemical sign can be detected using a measurement device connected to a detection chamber of the device (e.g., a fluorescence measurement device, a spectrophotometer, and/or an oscilloscope).


In certain aspects of this disclosure, multiplexing refers to parallel sensing of multiple target nucleic acid sequences in one sample by multiple probes. In some cases, there are from 2 to 50, from 3 to 40, from 4 to 30, from 5 to 20, or from 6 to 10 different populations of reporters capable of generating a detectable signal. In some cases there are from 2 to 50, from 5 to 50, from 10 to 50, from 15 to 50, from 20 to 50, from 25 to 50, from 30 to 50, from 35 to 50, from 40 to 50, from 2 to 40, from 5 to 40, from 10 to 40, from 15 to 40, from 20 to 40, from 25 to 40, from 30 to 40, from 35 to 40, from 2 to 30, from 5 to 30, from 10 to 30, from 15 to 30, from 20 to 30, from 25 to 30, from 2 to 20, from 5 to 20, from 10 to 20, from 15 to 20, from 2 to 10, or from 5 to 10 different populations of reporters capable of generating a detectable signal.









TABLE 5







Exemplary Single Stranded Reporter









5′ DETECTION

3′


MOIETY*
SEQUENCE (SEQ ID NO:)
QUENCHER*





/56-FAM/
rUrUrUrUrU
/3IABkFQ/





/5IRD700/
rUrUrUrUrU
/3IRQCQN/





/5TYE665/
rUrUrUrUrU
/3IAbRQSp/





/5Alex594N/
rUrUrUrUrU
/3IAbRQSp/





/5ATTO633N/
rUrUrUrUrU
/3IAbRQSp/





/56-FAM/
rUrUrUrUrUrUrUrU
/3IABkFQ/





/5IRD700/
rUrUrUrUrUrUrUrU
/3IRQC1N/





/5TYE665/
rUrUrUrUrUrUrUrU
/3IAbRQSp/





/5Alex594N/
rUrUrUrUrUrUrUrU
/3IAbRQSp/





/5ATTO633N/
rUrUrUrUrUrUrUrU
/3IAbRQSp/





/56-FAM/
rUrUrUrUrUrUrUrUrUrU(SEQ ID NO: 3)
/3IABkFQ/





/5IRD700/
rUrUrUrUrUrUrUrUrUrU(SEQ ID NO: 3)
/3IRQC1N/





/5TYE665/
rUrUrUrUrUrUrUrUrUrU(SEQ ID NO: 3)
/3IAbRQSp/





/5Alex594N/
rUrUrUrUrUrUrUrUrUrU(SEQ ID NO: 3)
/3IAbRQSp/





/5ATTO633N/
rUrUrUrUrUrUrUrUrUrU(SEQ ID NO: 3)
/3IAbRQSp/





/56-FAM/
TTTTrUrUTTTT(SEQ ID NO: 4)
/3IABkFQ/





/5IRD700/
TTTTrUrUTTTT(SEQ ID NO: 4)
/3IRQC1N/





/5TYE665/
TTTTrUrUTTTT(SEQ ID NO: 4)
/3IAbRQSp/





/5Alex594N/
TTTTrUrUTTTT(SEQ ID NO: 4)
/3IAbRQSp/





/5ATTO633N/
TTTTrUrUTTTT(SEQ ID NO: 4)
/3IAbRQSp/





/56-FAM/
TTrUrUTT
/3IABkFQ/





/5IRD700/
TTrUrUTT
/3IRQC1N/





/5TYE665/
TTrUrUTT
/3IAbRQSp/





/5Alex594N/
TTrUrUTT
/3IAbRQSp/





/5ATTO633N/
TTrUrUTT
/3IAbRQSp/





/56-FAM/
TArArUGC
/3IABkFQ/





/5IRD700/
TArArUGC
/3IRQC1N/





/5TYE665/
TArArUGC
/3IAbRQSp/





/5Alex594N/
TArArUGC
/3IAbRQSp/





/5ATTO633N/
TArArUGC
/3IAbRQSp/





/56-FAM/
TArUrGGC
/3IABkFQ/





/5IRD700/
TArUrGGC
/3IRQC1N/





/5TYE665/
TArUrGGC
/3IAbRQSp/





/5Alex594N/
TArUrGGC
/3IAbRQSp/





/5ATTO633N/
TArUrGGC
/3IAbRQSp/





/56-FAM/
rUrUrUrUrU
/3IABkFQ/





/5IRD700/
rUrUrUrUrU
/3IRQC1N/





/5TYE665/
rUrUrUrUrU
/3IAbRQSp/





/5Alex594N/
rUrUrUrUrU
/3IAbRQSp/





/5ATTO633N/
rUrUrUrUrU
/3IAbRQSp/





/56-FAM/
TTATTATT
/3IABkFQ/





/56-FAM/
TTATTATT
/3IABkFQ/





/5IRD700/
TTATTATT
/3IRQC1N/





/5TYE665/
TTATTATT
/3IAbRQSp/





/5Alex594N/
TTATTATT
/3IAbRQSp/





/5ATTO633N/
TTATTATT
/3IAbRQSp/





/56-FAM/
TTTTTT
/3IABkFQ/





/56-FAM/
TTTTTTTT
/3IABkFQ/





/56-FAM/
TTTTTTTTTT (SEQ ID NO: 12)
/3IABkFQ/





/56-FAM/
TTTTTTTTTTTT (SEQ ID NO: 13)
/3IABkFQ/





/56-FAM/
TTTTTTTTTTTTTT (SEQ ID NO: 14)
/3IABkFQ/





/56-FAM/
AAAAAA
/3IABkFQ/





/56-FAM/
CCCCCC
/3IABkFQ/





/56-FAM/
GGGGGG
/3IABkFQ/





/56-FAM/
TTATTATT
/3IABkFQ/





/56-FAM/: 5′ 6-Fluorescein (Integrated DNA Technologies)


/3IABkFQ/: 3′ Iowa Black FQ (Integrated DNA Technologies)


/5IRD700/: 5′ IRDye 700 (Integrated DNA Technologies)


/5TYE665/: 5′ TYE 665 (Integrated DNA Technologies)


/5Alex594N/: 5′ Alexa Fluor 594 (NHS Ester)(Integrated DNA Technologies)


/5ATTO633N/: 5′ ATTO TM 633 (NHS Ester)(Integrated DNA Technologies)


/3IRQC1N/: 3′ IRDye QC-1 Quencher (Li-Cor)


/3IAbRQSp/: 3′ Iowa Black RQ (Integrated DNA Technologies)


rU: uracil ribonucleotide


rG: guanine ribonucleotide


*This Table refers to the detection moiety and quencher moiety as their tradenames and their source is identified. However, alternatives, generics, or non-tradename moieties with similar function from other sources can also be used.






A detection moiety can be an infrared fluorophore. A detection moiety can be a fluorophore that emits fluorescence in the range of from 500 nm and 720 nm. A detection moiety can be a fluorophore that emits fluorescence in the range of from 500 nm and 720 nm. In some cases, the detection moiety emits fluorescence at a wavelength of 700 nm or higher. In other cases, the detection moiety emits fluorescence at about 660 nm or about 670 nm. In some cases, the detection moiety emits fluorescence at in the range of from 500 to 520, 500 to 540, 500 to 590, 590 to 600, 600 to 610, 610 to 620, 620 to 630, 630 to 640, 640 to 650, 650 to 660, 660 to 670, 670 to 680, 6890 to 690, 690 to 700, 700 to 710, 710 to 720, or 720 to 730 nm. A detection moiety can be a fluorophore that emits a fluorescence in the same range as 6-Fluorescein, IRDye 700, TYE 665, Alex Fluor, or ATTO TM 633 (NHS Ester). A detection moiety can be fluorescein amidite, 6-Fluorescein, IRDye 700, TYE 665, Alex Fluor 594, or ATTO TM 633 (NHS Ester). A detection moiety can be a fluorophore that emits a fluorescence in the same range as 6-Fluorescein (Integrated DNA Technologies), IRDye 700 (Integrated DNA Technologies), TYE 665 (Integrated DNA Technologies), Alex Fluor 594 (Integrated DNA Technologies), or ATTO TM 633 (NHS Ester) (Integrated DNA Technologies). A detection moiety can be fluorescein amidite, 6-Fluorescein (Integrated DNA Technologies), IRDye 700 (Integrated DNA Technologies), TYE 665 (Integrated DNA Technologies), Alex Fluor 594 (Integrated DNA Technologies), or ATTO TM 633 (NHS Ester) (Integrated DNA Technologies). Any of the detection moieties described herein can be from any commercially available source, can be an alternative with a similar function, a generic, or a non-tradename of the detection moieties listed.


A detection moiety can be chosen for use based on the type of sample to be tested. For example, a detection moiety that is an infrared fluorophore is used with a urine sample. As another example, SEQ ID NO: 1 with a fluorophore that emits around 520 nm is used for testing in non-urine samples, and SEQ ID NO: 8 with a fluorophore that emits a fluorescence around 700 nm is used for testing in urine samples.


A quenching moiety can be chosen based on its ability to quench the detection moiety. A quenching moiety can be a non-fluorescent fluorescence quencher. A quenching moiety can quench a detection moiety that emits fluorescence in the range of from 500 nm and 720 nm. A quenching moiety can quench a detection moiety that emits fluorescence in the range of from 500 nm and 720 nm. In some cases, the quenching moiety quenches a detection moiety that emits fluorescence at a wavelength of 700 nm or higher. In other cases, the quenching moiety quenches a detection moiety that emits fluorescence at about 660 nm or about 670 nm. In some cases, the quenching moiety quenches a detection moiety emits fluorescence at in the range of from 500 to 520, 500 to 540, 500 to 590, 590 to 600, 600 to 610, 610 to 620, 620 to 630, 630 to 640, 640 to 650, 650 to 660, 660 to 670, 670 to 680, 6890 to 690, 690 to 700, 700 to 710, 710 to 720, or 720 to 730 nm. A quenching moiety can quench fluorescein amidite, 6-Fluorescein, IRDye 700, TYE 665, Alex Fluor 594, or ATTO TM 633 (NHS Ester). A quenching moiety can be Iowa Black RQ, Iowa Black FQ or IRDye QC-1 Quencher. A quenching moiety can quench fluorescein amidite, 6-Fluorescein (Integrated DNA Technologies), IRDye 700 (Integrated DNA Technologies), TYE 665 (Integrated DNA Technologies), Alex Fluor 594 (Integrated DNA Technologies), or ATTO TM 633 (NHS Ester) (Integrated DNA Technologies). A quenching moiety can be Iowa Black RQ (Integrated DNA Technologies), Iowa Black FQ (Integrated DNA Technologies) or IRDye QC-1 Quencher (LiCor). Any of the quenching moieties described herein can be from any commercially available source, can be an alternative with a similar function, a generic, or a non-tradename of the quenching moieties listed.


The generation of the detectable signal from the release of the detection moiety indicates that cleavage by the programmable nuclease has occurred and that the sample contains the target nucleic acid. In some cases, the detection moiety comprises a fluorescent dye. Sometimes the detection moiety comprises a fluorescence resonance energy transfer (FRET) pair. In some cases, the detection moiety comprises an infrared (IR) dye. In some cases, the detection moiety comprises an ultraviolet (UV) dye. Alternatively, or in combination, the detection moiety comprises a polypeptide. Sometimes the detection moiety comprises a biotin. Sometimes the detection moiety comprises at least one of avidin or streptavidin. In some instances, the detection moiety comprises a polysaccharide, a polymer, or a nanoparticle. In some instances, the detection moiety comprises a gold nanoparticle or a latex nanoparticle.


Alternatively, or in combination, in some embodiments, detecting the presence or absence of a target nucleic acid of interest involves measuring a signal emitted from a conjugate bound to a detection moiety present in a reporter, after cleavage of the reporter by an activated programmable nuclease. The conjugates may comprise a nanoparticle, a gold nanoparticle, a latex nanoparticle, a quantum dot, a chemiluminescent nanoparticle, a carbon nanoparticle, a selenium nanoparticle, a fluorescent nanoparticle, a liposome, or a dendrimer. The surface of the conjugate may be coated by a conjugate binding molecule that binds to the detection moiety or another affinity molecule of the cleaved detector molecule as described herein. Thus, the detecting steps disclosed herein involve indirectly (e.g., via a reporter) measuring the presence of a target nucleic acid, quantifying how much of the target nucleic acid is present, or, measuring a signal indicating that the target nucleic acid is absent in a sample. In some embodiments, a signal is generated upon cleavage of the reporter by the programmable nuclease. In other embodiments, the signal changes upon cleavage of the reporter by the programmable nuclease. In other embodiments, a signal may be present in the absence of reporter cleavage and disappear upon cleavage of the target nucleic acid by the programmable nuclease. For example, a signal may be produced in a microfluidic device or lateral flow device after contacting a sample with a composition comprising a programmable nuclease.


A detection moiety can be any moiety capable of generating a calorimetric, potentiometric, amperometric, optical (e.g., fluorescent, colorimetric, etc.), or piezo-electric signal. A reporter, sometimes, is protein-nucleic acid that is capable of generating a calorimetric, potentiometric, amperometric, optical (e.g., fluorescent, colorimetric, etc.), or piezo-electric signal upon cleavage of the nucleic acid. Often a calorimetric signal is heat produced after cleavage of the reporters. Sometimes, a calorimetric signal is heat absorbed after cleavage of the reporters. A potentiometric signal, for example, is electrical potential produced after cleavage of the reporters. An amperometric signal can be movement of electrons produced after the cleavage of a reporter. Often, the signal is an optical signal, such as a colorimetric signal or a fluorescence signal. An optical signal is, for example, a light output produced after the cleavage of the reporters. Sometimes, an optical signal is a change in light absorbance between before and after the cleavage of reporters. Often, a piezo-electric signal is a change in mass between before and after the cleavage of the reporter.


Often, the protein-nucleic acid is an enzyme-nucleic acid. The enzyme may be sterically hindered when present as in the enzyme-nucleic acid, but then functional upon cleavage from the nucleic acid. Often, the enzyme is an enzyme that produces a reaction with a substrate. An enzyme can be invertase. Often, the substrate of invertase is sucrose and DNS reagent.


Sometimes the protein-nucleic acid is a substrate-nucleic acid. Often the substrate is a substrate that produces a reaction with an enzyme.


A protein-nucleic acid may be attached to a solid support. The solid support, for example, may be a surface. A surface can be an electrode. Sometimes the solid support is a bead. Often the bead is a magnetic bead. Upon cleavage, the protein is liberated from the solid and interacts with other mixtures. For example, the protein is an enzyme, and upon cleavage of the nucleic acid of the enzyme-nucleic acid, the enzyme flows through a chamber into a mixture comprising the substrate. When the enzyme meets the enzyme substrate, a reaction occurs, such as a colorimetric reaction, which is then detected. As another example, the protein is an enzyme substrate, and upon cleavage of the nucleic acid of the enzyme substrate-nucleic acid, the enzyme flows through a chamber into a mixture comprising the enzyme. When the enzyme substrate meets the enzyme, a reaction occurs, such as a calorimetric reaction, which is then detected.


In some embodiments, the reporter comprises a nucleic acid conjugated to an affinity molecule and the affinity molecule conjugated to the fluorophore (e.g., nucleic acid—affinity molecule—fluorophore) or the nucleic acid conjugated to the fluorophore and the fluorophore conjugated to the affinity molecule (e.g., nucleic acid—fluorophore—affinity molecule). In some embodiments, a linker conjugates the nucleic acid to the affinity molecule. In some embodiments, a linker conjugates the affinity molecule to the fluorophore. In some embodiments, a linker conjugates the nucleic acid to the fluorophore. A linker can be any suitable linker known in the art. In some embodiments, the nucleic acid of the reporter can be directly conjugated to the affinity molecule and the affinity molecule can be directly conjugated to the fluorophore or the nucleic acid can be directly conjugated to the fluorophore and the fluorophore can be directly conjugated to the affinity molecule. In this context, “directly conjugated” indicated that no intervening molecules, polypeptides, proteins, or other moieties are present between the two moieties directly conjugated to each other. For example, if a reporter comprises a nucleic acid directly conjugated to an affinity molecule and an affinity molecule directly conjugated to a fluorophore—no intervening moiety is present between the nucleic acid and the affinity molecule and no intervening moiety is present between the affinity molecule and the fluorophore. The affinity molecule can be biotin, avidin, streptavidin, or any similar molecule.


In some cases, the reporter comprises a substrate-nucleic acid. The substrate may be sequestered from its cognate enzyme when present as in the substrate-nucleic acid, but then is released from the nucleic acid upon cleavage, wherein the released substrate can contact the cognate enzyme to produce a detectable signal. Often, the substrate is sucrose and the cognate enzyme is invertase, and a DNS reagent can be used to monitor invertase activity.


A major advantage of the devices and methods disclosed herein is the design of excess reporters to total nucleic acids in an unamplified or an amplified sample, not including the nucleic acid of the reporter. Total nucleic acids can include the target nucleic acids and non-target nucleic acids, not including the nucleic acid of the reporter. The non-target nucleic acids can be from the original sample, either lysed or unlysed. The non-target nucleic acids can also be byproducts of amplification. Thus, the non-target nucleic acids can include both non-target nucleic acids from the original sample, lysed or unlysed, and from an amplified sample. The presence of a large amount of non-target nucleic acids, an activated programmable nuclease may be inhibited in its ability to bind and cleave the reporter sequences. This is because the activated programmable nucleases collaterally cleaves any nucleic acids. If total nucleic acids are present in large amounts, they may outcompete reporters for the programmable nucleases. The devices and methods disclosed herein are designed to have an excess of reporter to total nucleic acids, such that the detectable signals from cleavage reactions (e.g., DETECTR reactions) are particularly superior. In some embodiments, the reporter can be present in at least 1.5 fold, at least 2 fold, at least 3 fold, at least 4 fold, at least 5 fold, at least 6 fold, at least 7 fold, at least 8 fold, at least 9 fold, at least 10 fold, at least 11 fold, at least 12 fold, at least 13 fold, at least 14 fold, at least 15 fold, at least 16 fold, at least 17 fold, at least 18 fold, at least 19 fold, at least 20 fold, at least 30 fold, at least 40 fold, at least 50 fold, at least 60 fold, at least 70 fold, at least 80 fold, at least 90 fold, at least 100 fold, from 1.5 fold to 100 fold, from 2 fold to 10 fold, from 10 fold to 20 fold, from 20 fold to 30 fold, from 30 fold to 40 fold, from 40 fold to 50 fold, from 50 fold to 60 fold, from 60 fold to 70 fold, from 70 fold to 80 fold, from 80 fold to 90 fold, from 90 fold to 100 fold, from 1.5 fold to 10 fold, from 1.5 fold to 20 fold, from 10 fold to 40 fold, from 20 fold to 60 fold, or from 10 fold to 80 fold excess of total nucleic acids.


A second significant advantage of the devices and methods disclosed herein is the design of an excess volume comprising the guide nucleic acid, the programmable nuclease, and the reporter, which contacts a smaller volume comprising the sample with the target nucleic acid of interest. The smaller volume comprising the sample can be unlysed sample, lysed sample, or lysed sample which has undergone any combination of reverse transcription, amplification, and in vitro transcription. The presence of various reagents in a crude, non-lysed sample, a lysed sample, or a lysed and amplified sample, such as buffer, magnesium sulfate, salts, the pH, a reducing agent, primers, dNTPs, NTPs, cellular lysates, non-target nucleic acids, primers, or other components, can inhibit the ability of the programmable nuclease to find and cleave the nucleic acid of the reporter. This may be due to nucleic acids that are not the reporter, which outcompete the nucleic acid of the reporter, for the programmable nuclease. Alternatively, various reagents in the sample may simply inhibit the activity of the programmable nuclease. Thus, the devices and methods provided herein for contacting an excess volume comprising the guide nucleic acid, the programmable nuclease, and the reporter to a smaller volume comprising the sample with the target nucleic acid of interest provides for superior detection of the target nucleic acid by ensuring that the programmable nuclease is able to find and cleaves the nucleic acid of the reporter. In some embodiments, the volume comprising the guide nucleic acid, the programmable nuclease, and the reporter (can be referred to as “a second volume”) is 4-fold greater than a volume comprising the sample (can be referred to as “a first volume”). In some embodiments, the volume comprising the guide nucleic acid, the programmable nuclease, and the reporter (can be referred to as “a second volume”) is at least 1.5 fold, at least 2 fold, at least 3 fold, at least 4 fold, at least 5 fold, at least 6 fold, at least 7 fold, at least 8 fold, at least 9 fold, at least 10 fold, at least 11 fold, at least 12 fold, at least 13 fold, at least 14 fold, at least 15 fold, at least 16 fold, at least 17 fold, at least 18 fold, at least 19 fold, at least 20 fold, at least 30 fold, at least 40 fold, at least 50 fold, at least 60 fold, at least 70 fold, at least 80 fold, at least 90 fold, at least 100 fold, from 1.5 fold to 100 fold, from 2 fold to 10 fold, from 10 fold to 20 fold, from 20 fold to 30 fold, from 30 fold to 40 fold, from 40 fold to 50 fold, from 50 fold to 60 fold, from 60 fold to 70 fold, from 70 fold to 80 fold, from 80 fold to 90 fold, from 90 fold to 100 fold, from 1.5 fold to 10 fold, from 1.5 fold to 20 fold, from 10 fold to 40 fold, from 20 fold to 60 fold, or from 10 fold to 80 fold greater than a volume comprising the sample (can be referred to as “a first volume”). In some embodiments, the volume comprising the sample is at least 0.5 μl, at least 1 μl, at least at least 1 μL, at least 2 μL, at least 3 μL, at least 4 μL, at least 5 μL, at least 6 μL, at least 7 μL, at least 8 μL, at least 9 μL, at least 10 μL, at least 11 μL, at least 12 μL, at least 13 μL, at least 14 μL, at least 15 μL, at least 16 μL, at least 17 μL, at least 18 μL, at least 19 μL, at least 20 μL, at least 25 μL, at least 30 μL, at least 35 μL, at least 40 μL, at least 45 μL, at least 50 μL, at least 55 μL, at least 60 μL, at least 65 μL, at least 70 μL, at least 75 μL, at least 80 μL, at least 85 μL, at least 90 μL, at least 95 μL, at least 100 μL, from 0.5 μL to 5 μl μL, from 5 μL to 10 μL, from 10 μL to 15 μL, from 15 μL to 20 μL, from 20 μL to 25 μL, from 25 μL to 30 μL, from 30 μL to 35 μL, from 35 μL to 40 μL, from 40 μL to 45 μL, from 45 μL to 50 μL, from 10 μL to 20 μL, from 5 μL to 20 μL, from 1 μL to 40 μL, from 2 μL to 10 μL, or from 1 μL to 10 μL. In some embodiments, the volume comprising the programmable nuclease, the guide nucleic acid, and the reporter is at least 10 μL, at least 11 μL, at least 12 μL, at least 13 μL, at least 14 μL, at least 15 μL, at least 16 μL, at least 17 μL, at least 18 μL, at least 19 μL, at least 20 μL, at least 21 μL, at least 22 μL, at least 23 μL, at least 24 μL, at least 25 μL, at least 26 μL, at least 27 μL, at least 28 μL, at least 29 μL, at least 30 μL, at least 40 μL, at least 50 μL, at least 60 μL, at least 70 μL, at least 80 μL, at least 90 μL, at least 100 μL, at least 150 μL, at least 200 μL, at least 250 μL, at least 300 μL, at least 350 μL, at least 400 μL, at least 450 μL, at least 500 μL, from 10 μL to 15 μl μL, from 15 μL to 20 μL, from 20 μL to 25 μL, from 25 μL to 30 μL, from 30 μL to 35 μL, from 35 μL to 40 μL, from 40 μL to 45 μL, from 45 μL to 50 μL, from 50 μL to 55 μL, from 55 μL to 60 μL, from 60 μL to 65 μL, from 65 μL to 70 μL, from 70 μL to 75 μL, from 75 μL to 80 μL, from 80 μL to 85 μL, from 85 μL to 90 μL, from 90 μL to 95 μL, from 95 μL to 100 μL, from 100 μL to 150 μL, from 150 μL to 200 μL, from 200 μL to 250 μL, from 250 μL to 300 μL, from 300 μL to 350 μL, from 350 μL to 400 μL, from 400 μL to 450 μL, from 450 μL to 500 μL, from 10 μL to 20 μL, from 10 μL to 30 μL, from 25 μL to 35 μL, from 10 μL to 40 μL, from 20 μL to 50 μL, from 18 μL to 28 μL, or from 17 μL to 22 μL.


A reporter may be a hybrid nucleic acid reporter. A hybrid nucleic acid reporter comprises a nucleic acid with at least one deoxyribonucleotide and at least one ribonucleotide. In some embodiments, the nucleic acid of the hybrid nucleic acid reporter can be of any length and can have any mixture of DNAs and RNAs. For example, in some cases, longer stretches of DNA can be interrupted by a few ribonucleotides. Alternatively, longer stretches of RNA can be interrupted by a few deoxyribonucleotides. Alternatively, every other base in the nucleic acid may alternate between ribonucleotides and deoxyribonucleotides. A major advantage of the hybrid nucleic acid reporter is increased stability as compared to a pure RNA nucleic acid reporter. For example, a hybrid nucleic acid reporter can be more stable in solution, lyophilized, or vitrified as compared to a pure DNA or pure RNA reporter.


Additionally, target nucleic acid can be amplified before binding to the crRNA of the CRISPR enzyme. This amplification can be PCR amplification or isothermal amplification. This nucleic acid amplification of the sample can improve at least one of sensitivity, specificity, or accuracy of the detection the target RNA. The reagents for nucleic acid amplification can comprise a recombinase, an oligonucleotide primer, a single-stranded DNA binding (SSB) protein, and a polymerase. The nucleic acid amplification can be transcription mediated amplification (TMA). Nucleic acid amplification can be helicase dependent amplification (HDA) or circular helicase dependent amplification (cHDA). In additional cases, nucleic acid amplification is strand displacement amplification (SDA). The nucleic acid amplification can be recombinase polymerase amplification (RPA). The nucleic acid amplification can be at least one of loop mediated amplification (LAMP) or the exponential amplification reaction (EXPAR). Nucleic acid amplification is, in some cases, by rolling circle amplification (RCA), ligase chain reaction (LCR), simple method amplifying RNA targets (SMART), single primer isothermal amplification (SPIA), multiple displacement amplification (MDA), nucleic acid sequence based amplification (NASBA), hinge-initiated primer-dependent amplification of nucleic acids (HIP), nicking enzyme amplification reaction (NEAR), or improved multiple displacement amplification (IMDA). The nucleic acid amplification can be performed for no greater than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 40, 50, or 60 minutes. Sometimes, the nucleic acid amplification reaction is performed at a temperature of around 20-45° C. The nucleic acid amplification reaction can be performed at a temperature no greater than 20° C., 25° C., 30° C., 35° C., 37° C., 40° C., 45° C. The nucleic acid amplification reaction can be performed at a temperature of at least 20° C., 25° C., 30° ° C., 35° C., 37° C., 40° ° C., or 45° C.


In some cases, an amplification method may comprise a set of multiple distinct primers targeting a single amplicon (e.g., a gene, such as the SARS-Cov-2 E and N genes). The set of primers may all target sequences upstream of a gene of interest, and may comprise overlapping, partially overlapping, or entirely distinct sequences. The use of multiple primers can increase the yield from amplification reactions. In some cases, the use of multiple distinct primers can increase the likelihood that damaged nucleic acids are amplified in a reaction. The use of multiple distinct primers can also facilitate amplification (and therefore detection) of mutant nucleic acids. In some cases, a sample may comprise a mutant strain of SARS-COV-2 that would not be detectable if a single primer were used for amplification. A primer may be designed for thermal cycling amplification of the target nucleic acid. A primer may be designed for isothermal amplification of the target nucleic acid.


In some embodiments, an amplification method may utilize an amplification activator. A set of amplification reagents may require addition of an amplification activator for activity. In such cases, an amplification activator may enable temporal control over the start of an amplification reaction. In some embodiments, the amplification activator comprises a magnesium salt, deoxyribonucleoside triphosphates (dNTPs), nucleoside triphosphates (NTPs), adenosine triphosphate (ATP), or a combination thereof. In some embodiments, the magnesium salt is magnesium sulfate, magnesium chloride, magnesium acetate, magnesium phosphate, magnesium iodide, magnesium fluoride, magnesium bromide, or a combination thereof.


Disclosed herein are methods of assaying for a target nucleic acid as described herein wherein a signal is detected. For example, a method of assaying for a target nucleic acid in a sample comprises contacting the sample to a complex comprising a guide nucleic acid comprising a segment that is reverse complementary to a segment of the target nucleic acid and a programmable nuclease that exhibits sequence independent cleavage upon forming a complex comprising the segment of the guide nucleic acid binding to the segment of the target nucleic acid; and assaying for a signal indicating cleavage of at least some protein-nucleic acids of a population of protein-nucleic acids, wherein the signal indicates a presence of the target nucleic acid in the sample and wherein absence of the signal indicates an absence of the target nucleic acid in the sample. As another example, a method of assaying for a target nucleic acid in a sample, for example, comprises: a) contacting the sample to a complex comprising a guide nucleic acid comprising a segment that is reverse complementary to a segment of the target nucleic acid (e.g., a nucleic acid from a coronavirus such as SARS-COV-2) and a programmable nuclease that exhibits sequence independent cleavage upon forming a complex comprising the segment of the guide nucleic acid binding to the segment of the target nucleic acid; b) contacting the complex to a substrate; c) contacting the substrate to a reagent that differentially reacts with a cleaved substrate; and d) assaying for a signal indicating cleavage of the substrate, wherein the signal indicates a presence of the target nucleic acid in the sample and wherein absence of the signal indicates an absence of the target nucleic acid in the sample. Often, the substrate is an enzyme-nucleic acid. Sometimes, the substrate is an enzyme substrate-nucleic acid.


A programmable nuclease can comprise a programmable nuclease capable of being activated when complexed with a guide nucleic acid and target nucleic acid (e.g., a nucleic acid from a coronavirus such as SARS-COV-2). The programmable nuclease can become activated after binding of a guide nucleic acid with a target nucleic acid, in which the activated programmable nuclease can cleave the target nucleic acid and can have trans cleavage activity. Trans cleavage activity can be non-specific cleavage of nearby nucleic acids by the activated programmable nuclease, such as trans cleavage of reporters with a detection moiety. Once the reporter is cleaved by the activated programmable nuclease, the detection moiety can be released from the reporter and can generate a signal. The detection moiety capable of generating the detectable signal can be immobilized on a support medium for detection. The signal can be visualized to assess whether a target nucleic acid comprises a modification.


Often, the signal is a colorimetric signal or a signal visible by eye. In some instances, the signal is fluorescent, electrical, chemical, electrochemical, or magnetic. A signal can be a calorimetric, potentiometric, amperometric, optical (e.g., fluorescent, colorimetric, etc.), or piezo-electric signal. In some cases, the detectable signal is a colorimetric signal or a signal visible by eye. In some instances, the detectable signal is fluorescent, electrical, chemical, electrochemical, or magnetic. In some cases, the first detection signal is generated by binding of the detection moiety to the capture molecule in the detection region, where the first detection signal indicates that the sample contained the target nucleic acid. Sometimes the system is capable of detecting more than one type of target nucleic acid, wherein the system comprises more than one type of guide nucleic acid and more than one type of reporter. In some cases, the detectable signal is generated directly by the cleavage event. Alternatively, or in combination, the detectable signal is generated indirectly by the signal event. Sometimes the detectable signal is not a fluorescent signal. In some instances, the detectable signal is a colorimetric or color-based signal. In some cases, the detected target nucleic acid is identified based on its spatial location on the detection region of the support medium. In some cases, the second detectable signal is generated in a spatially distinct location than the first generated signal.


In some cases, the threshold of detection, for a subject method of detecting a single-stranded target nucleic acid in a sample, is less than or equal to 10 nM. The term “threshold of detection” is used herein to describe the minimal amount of target nucleic acid that must be present in a sample in order for detection to occur. For example, when a threshold of detection is 10 nM, then a signal can be detected when a target nucleic acid is present in the sample at a concentration of 10 nM or more. In some cases, the threshold of detection is less than or equal to 5 nM, 1 nM, 0.5 nM, 0.1 nM, 0.05 nM, 0.01 nM, 0.005 nM, 0.001 nM, 0.0005 nM, 0.0001 nM, 0.00005 nM, 0.00001 nM, 10 pM, 1 pM, 500 fM, 250 fM, 100 fM, 50 fM, 10 fM, 5 fM, 1 fM, 500 attomole (aM), 100 aM, 50 aM, 10 aM, or 1 aM. In some cases, the threshold of detection is in a range of from 1 aM to 1 nM, 1 aM to 500 pM, 1 aM to 200 pM, 1 aM to 100 pM, 1 aM to 10 pM, 1 aM to 1 pM, 1 aM to 500 fM, 1 aM to 100 fM, 1 aM to 1 fM, 1 aM to 500 aM, 1 aM to 100 aM, 1 aM to 50 aM, 1 aM to 10 aM, 10 aM to 1 nM, 10 aM to 500 pM, 10 aM to 200 pM, 10 aM to 100 pM, 10 aM to 10 pM, 10 aM to 1 pM, 10 aM to 500 fM, 10 aM to 100 fM, 10 aM to 1 fM, 10 aM to 500 aM, 10 aM to 100 aM, 10 aM to 50 aM, 100 aM to 1 nM, 100 aM to 500 pM, 100 aM to 200 pM, 100 aM to 100 pM, 100 aM to 10 pM, 100 aM to 1 pM, 100 aM to 500 fM, 100 aM to 100 fM, 100 aM to 1 fM, 100 aM to 500 aM, 500 aM to 1 nM, 500 aM to 500 pM, 500 aM to 200 pM, 500 aM to 100 pM, 500 aM to 10 pM, 500 aM to 1 pM, 500 aM to 500 fM, 500 aM to 100 fM, 500 aM to 1 fM, 1 fM to 1 nM, 1 fM to 500 pM, 1 fM to 200 pM, 1 fM to 100 pM, 1 fM to 10 pM, 1 fM to 1 pM, 10 fM to 1 nM, 10 fM to 500 pM, 10 fM to 200 pM, 10 fM to 100 pM, 10 fM to 10 pM, 10 fM to 1 pM, 500 fM to 1 nM, 500 fM to 500 pM, 500 fM to 200 pM, 500 fM to 100 pM, 500 fM to 10 pM, 500 fM to 1 pM, 800 fM to 1 nM, 800 fM to 500 pM, 800 fM to 200 pM, 800 fM to 100 pM, 800 fM to 10 pM, 800 fM to 1 pM, from 1 pM to 1 nM, 1 pM to 500 pM, 1 pM to 200 pM, 1 pM to 100 pM, or 1 pM to 10 pM. In some cases, the threshold of detection in a range of from 800 fM to 100 pM, 1 pM to 10 pM, 10 fM to 500 fM, 10 fM to 50 fM, 50 fM to 100 fM, 100 fM to 250 fM, or 250 fM to 500 fM. In some cases, the minimum concentration at which a single-stranded target nucleic acid is detected in a sample is in a range of from 1 aM to 1 nM, 10 aM to 1 nM, 100 aM to 1 nM, 500 aM to 1 nM, 1 fM to 1 nM, 1 fM to 500 pM, 1 fM to 200 pM, 1 fM to 100 pM, 1 fM to 10 pM, 1 fM to 1 pM, 10 fM to 1 nM, 10 fM to 500 pM, 10 fM to 200 pM, 10 fM to 100 pM, 10 fM to 10 pM, 10 fM to 1 pM, 500 fM to 1 nM, 500 fM to 500 pM, 500 fM to 200 pM, 500 fM to 100 pM, 500 fM to 10 pM, 500 fM to 1 pM, 800 fM to 1 nM, 800 fM to 500 pM, 800 fM to 200 pM, 800 fM to 100 pM, 800 fM to 10 pM, 800 fM to 1 pM, 1 pM to 1 nM, 1 pM to 500 pM, from 1 pM to 200 pM, 1 pM to 100 pM, or 1 pM to 10 pM. In some cases, the minimum concentration at which a single-stranded target nucleic acid can be detected in a sample is in a range of from 1 aM to 100 pM. In some cases, the minimum concentration at which a single-stranded target nucleic acid can be detected in a sample is in a range of from 1 fM to 100 pM. In some cases, the minimum concentration at which a single-stranded target nucleic acid can be detected in a sample is in a range of from 10 fM to 100 pM. In some cases, the minimum concentration at which a single-stranded target nucleic acid can be detected in a sample is in a range of from 800 fM to 100 pM. In some cases, the minimum concentration at which a single-stranded target nucleic acid can be detected in a sample is in a range of from 1 pM to 10 pM. In some cases, the devices, systems, fluidic devices, kits, and methods described herein detect a target single-stranded nucleic acid in a sample comprising a plurality of nucleic acids such as a plurality of non-target nucleic acids, where the target single-stranded nucleic acid is present at a concentration as low as 1 aM, 10 aM, 100 aM, 500 aM, 1 fM, 10 fM, 500 fM, 800 fM, 1 pM, 10 pM, 100 pM, or 1 pM.


In some cases, the devices, systems, fluidic devices, kits, and methods described herein detect a target single-stranded nucleic acid (e.g., a nucleic acid from a coronavirus such as SARS-COV-2) in a sample where the sample is contacted with the reagents for a predetermined length of time sufficient for the trans cleavage to occur or cleavage reaction to reach completion. In some cases, the devices, systems, fluidic devices, kits, and methods described herein detect a target single-stranded nucleic acid in a sample where the sample is contacted with the reagents for no greater than 60 minutes. Sometimes the sample is contacted with the reagents for no greater than 120 minutes, 110 minutes, 100 minutes, 90 minutes, 80 minutes, 70 minutes, 60 minutes, 55 minutes, 50 minutes, 45 minutes, 40 minutes, 35 minutes, 30 minutes, 25 minutes, 20 minutes, 15 minutes, 10 minutes, 5 minutes, 4 minutes, 3 minutes, 2 minutes, or 1 minute. Sometimes the sample is contacted with the reagents for at least 120 minutes, 110 minutes, 100 minutes, 90 minutes, 80 minutes, 70 minutes, 60 minutes, 55 minutes, 50 minutes, 45 minutes, 40 minutes, 35 minutes, 30 minutes, 25 minutes, 20 minutes, 15 minutes, 10 minutes, or 5 minutes. In some cases, the devices, systems, fluidic devices, kits, and methods described herein can detect a target nucleic acid in a sample in less than 10 hours, less than 9 hours, less than 8 hours, less than 7 hours, less than 6 hours, less than 5 hours, less than 4 hours, less than 3 hours, less than 2 hours, less than 1 hour, less than 50 minutes, less than 45 minutes, less than 40 minutes, less than 35 minutes, less than 30 minutes, less than 25 minutes, less than 20 minutes, less than 15 minutes, less than 10 minutes, less than 9 minutes, less than 8 minutes, less than 7 minutes, less than 6 minutes, or less than 5 minutes.


When a guide nucleic acid binds to a target nucleic acid (e.g., a nucleic acid from a coronavirus such as SARS-COV-2), the programmable nuclease's trans cleavage activity can be initiated, and reporters can be cleaved, resulting in the detection of fluorescence. Some methods as described herein can a method of assaying for a target nucleic acid in a sample comprises contacting the sample to a complex comprising a guide nucleic acid comprising a segment that is reverse complementary to a segment of the target nucleic acid and a programmable nuclease that exhibits sequence independent cleavage upon forming a complex comprising the segment of the guide nucleic acid binding to the segment of the target nucleic acid; and assaying for a signal indicating cleavage of at least some protein-nucleic acids of a population of protein-nucleic acids, wherein the signal indicates a presence of the target nucleic acid in the sample and wherein absence of the signal indicates an absence of the target nucleic acid in the sample. The cleaving of the reporter using the programmable nuclease may cleave with an efficiency of 50% as measured by a change in a signal that is calorimetric, potentiometric, amperometric, optical (e.g., fluorescent, colorimetric, etc.), or piezo-electric, as non-limiting examples. Some methods as described herein can be a method of detecting a target nucleic acid in a sample comprising contacting the sample comprising the target nucleic acid with a guide nucleic acid targeting a target nucleic acid segment, a programmable nuclease capable of being activated when complexed with the guide nucleic acid and the target nucleic acid segment, a single-stranded reporter comprising a detection moiety, wherein the reporter is capable of being cleaved by the activated programmable nuclease, thereby generating a first detectable signal, cleaving the single-stranded reporter using the programmable nuclease that cleaves as measured by a change in color, and measuring the first detectable signal on the support medium. The cleaving of the single-stranded reporter using the programmable nuclease may cleave with an efficiency of 50% as measured by a change in color. In some cases, the cleavage efficiency is at least 40%, 50%, 60%, 70%, 80%, 90%, or 95% as measured by a change in color. The change in color may be a detectable colorimetric signal or a signal visible by eye. The change in color may be measured as a first detectable signal. The first detectable signal can be detectable within 5 minutes of contacting the sample comprising the target nucleic acid with a guide nucleic acid targeting a target nucleic acid segment, a programmable nuclease capable of being activated when complexed with the guide nucleic acid and the target nucleic acid segment, and a single-stranded reporter comprising a detection moiety, wherein the reporter is capable of being cleaved by the activated nuclease. The first detectable signal can be detectable within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 110, or 120 minutes of contacting the sample.


In some cases, the methods, reagents, and devices described herein detect a target nucleic acid with a programmable nuclease and a single-stranded reporter in a sample where the sample is contacted with the reagents for a predetermined length of time sufficient for trans cleavage of the single-stranded reporter. For example, a programmable nuclease is LbuCas13a that detects a target nucleic acid and a single-stranded reporter comprises two adjacent uracil nucleotides with a green detectable moiety that is detected upon cleavage. As another example, a programmable nuclease is LbaCas13a that detects a target nucleic acid and a single-stranded reporter comprises two adjacent adenine nucleotides with a red detectable moiety that is detected upon cleavage. The target nucleic acid may be a single-stranded nucleic acid (e.g., a single-stranded DNA (ssDNA) or a single-stranded RNA), or the target nucleic acid may be a double-stranded nucleic acid (e.g., a double-stranded DNA (dsDNA) or a double-stranded RNA).


Buffers for Detection of Nucleic Acids

The reagents described herein can also include buffers, which are compatible with the devices, systems, fluidic devices, kits, and methods disclosed herein. These buffers are compatible with the other reagents, samples, and support mediums as described herein for detection of an ailment, such as a disease, including those caused by viruses such as influenza. The methods described herein can also include the use of buffers, which are compatible with the methods disclosed herein.


A buffer may be configured to support multiple reactions or processes. A buffer may be configured to enable fast reaction rates for multiple types of reactions. For example, a buffer may enable a reaction to achieve a rate within 25%, 50%, 75%, or 90% of the fastest reported rate for that reaction at a specified temperature. The specified temperature may be between 0 and 10° C., 10 and 20° C., 20 and 30° C., 30 and 40° C., 40 and 50° C., 50 and 60° C., 60 and 70° C., 70 and 80° C., 80 and 90° C., or higher than 90° C. A buffer may enable amplification and DETECTR reactions to each reach completion (e.g., consume at least 80% of a limiting reagent) in less than two hours, less than 1 hour, less than 45 minutes, less than 30 minutes, less than 20 minutes, or less than 15 minutes. A buffer may enable fast reaction rates for 2, 3, 4, or 5 or more types of reactions, wherein each reaction comprises one or more targets. For example, a buffer may enable fast reaction rates for amplification and DETECTR reactions, and may comprise amplification and DETECTR reagents each targeting 100 nucleic acid sequences.


Time to completion may be measured by the consumption of a reagent. For example, a single-buffer comprising amplification and DETECTR reagents may consume (e.g., subject to transcollateral cleavage) at least 50 nM of reporters within 3 hours, 2 hours, 1.5 hours, 1 hour, 45 minutes, 30 minutes, 20 minutes, or 15 minutes of addition of 10000 copies, 5000 copies, 2000 copies, 1000 copies, 500 copies, 300 copies, 200 copies, 100 copies, 50 copies, or 10 copies of a target nucleic acid. A single-buffer comprising amplification and DETECTR reagents may consume at least 20 nM of reporters within 3 hours, 2 hours, 1.5 hours, 1 hour, 45 minutes, 30 minutes, 20 minutes, or 15 minutes of addition of 10000 copies, 5000 copies, 2000 copies, 1000 copies, 500 copies, 300 copies, 200 copies, 100 copies, 50 copies, or 10 copies of a target nucleic acid. A single-buffer comprising amplification and DETECTR reagents may consume at least 10 nM of reporters within 3 hours, 2 hours, 1.5 hours, 1 hour, 45 minutes, 30 minutes, 20 minutes, or 15 minutes of addition of 10000 copies, 5000 copies, 2000 copies, 1000 copies, 500 copies, 300 copies, 200 copies, 100 copies, 50 copies, or 10 copies of a target nucleic acid. A single-buffer comprising amplification and DETECTR reagents may consume at least 5 nM of reporters within 3 hours, 2 hours, 1.5 hours, 1 hour, 45 minutes, 30 minutes, 20 minutes, or 15 minutes of addition of 10000 copies, 5000 copies, 2000 copies, 1000 copies, 500 copies, 300 copies, 200 copies, 100 copies, 50 copies, or 10 copies of a target nucleic acid. A single-buffer comprising amplification and DETECTR reagents may consume at least 1 nM of reporters within 3 hours, 2 hours, 1.5 hours, 1 hour, 45 minutes, 30 minutes, 20 minutes, or 15 minutes of addition of 10000 copies, 5000 copies, 2000 copies, 1000 copies, 500 copies, 300 copies, 200 copies, 100 copies, 50 copies, or 10 copies of a target nucleic acid. A single-buffer comprising amplification and DETECTR reagents may consume at least 500 pM of reporters within 3 hours, 2 hours, 1.5 hours, 1 hour, 45 minutes, 30 minutes, 20 minutes, or 15 minutes of addition of 10000 copies, 5000 copies, 2000 copies, 1000 copies, 500 copies, 300 copies, 200 copies, 100 copies, 50 copies, or 10 copies of a target nucleic acid.


A buffer may also be configured to maximize the stability of a reagent (e.g., a programmable nuclease or a target nucleic acid) or reaction species (e.g., a DNA molecule produced by reverse transcription of a target RNA molecule). For example, a buffer may stabilize RNA so that its average half-life is 1.5 hours, 1 hour, 30 minutes, 20 minutes, 10 minutes, 5 minutes, 3 minutes, 2 minutes, or 1 minute at room temperature. A buffer may stabilize a reagent or enzyme for long-term storage. A buffer of the present disclosure may enable stable, long-term storage for a set of reagents, such as a composition comprising amplification and DETECTR reagents. A central challenge to the design of such a buffer stems from different proteins having different optimal conditions for stability. Optimal conditions for two proteins may comprise different osmolarities, viscosities, dielectric constants, pH, lipid content, detergent content, proportions of non-aqueous solvents (e.g., methanol, ethanol, acetone, acetonitrile, or a halomethane such as trichloromethane), or densities. The present disclosure provides buffers that are optimized to simultaneously stabilize a plurality of different proteins, such as a polymerase and a programmable nuclease (e.g., CRISPR-Cas enzyme). Enzymes for amplification and CRISPR-Cas reactions may have half-lives of at least three months, two months, one month, two weeks, one week, five days, 3 days, 2 days, 1 day, 12 hours, 8 hours, 6 hours, 4 hours, or 3 hours, 2 hours, or 1 hours at room temperature within a buffer of the present disclosure. Enzymes for amplification and CRISPR-Cas reactions may have half-lives that are at least 100 times, 50 times, 30 times, 20 times, 10 times, 5 times, or 2 times longer in a buffer of the present disclosure than in 50 mM pH 7.0 HEPES buffer, Escherichia coli cell lysate, or Pichia pastoris cell lysate. Enzymes for amplification and CRISPR-Cas reactions contained within buffers of the present disclosure may be stable through multiple freeze-thaw cycles (e.g., less than 10%, 5%, 2%, 1% or 0.5% activity through an individual freeze-thaw cycle), therein enabling compositions and kits of the present disclosure to be stored and shipped at temperatures below 0° C.


A buffer may comprise biological buffer, such as TRIS, DIPSO, HEPES, PIPES, maleic acid, citric acid, malic acid, formic acid, lactic acid, succinic acid, acetic acid, pivalic acid, pyridine, piperazine, picolinic acid, L-histidine, MES, bis-tris, bis-trispropane, MOPS, ADA, ACES, MOPSO, imidazole, MOPS, BES, TES, HEPES, DIPSO, TAPSO, TEA (triethanolamine)), N-ethylmorpholine, POPSO, EPPS, HEPPS, HEPPSO, Tris, Tricine, glycylglycine, bicine, TAPS, morpholine, N-methyldiethanolamine, AMPD (2-amino-2-methyl-1,3-propanediol)), Diethanolamine AMPSO, CHES, glycine, CAPSO, ethanolamine, AMP (2-amino-2-methyl-1-propanol), piperazine, CAPS, 1,3-diaminopropane, CABS, or piperidine. A buffer may comprise an inorganic buffer, such as phosphoric acid, carbonate, bicarbonate (e.g., sodium bicarbonate), or ammonia. A buffer may comprise a pH of about 3.5, about 3.6, about 3.7, about 3.8, about 3.9, about 4, about 4.1, about 4.2, about 4.3, about 4.4, about 4.5, about 4.6, about 4.7, about 4.8, about 4.9, about 5, about 5.1, about 5.2, about 5.3, about 5.4, about 5.5, about 6, about 6.5, about 7, about 7.5, about 8, about 8.5, or about 9. In some embodiments, a buffer may comprise a pH of from 3.5 to 4.5, from 4 to 5, from 4.5 to 5.5, from 3.5 to 4, from 4 to 4.5, from 4.5 to 5, from 5 to 5.5, from 5 to 6, from 6 to 7, from 7 to 8, from 8 to 9, or from 9 to 10.


A buffer may comprise a calcium or magnesium concentration of about 0 mM, about 2 mM, about 4 mM, about 5 mM, about 6 mM, about 8 mM, about 10 mM, about 12 mM, about 13 mM, about 14 mM, about 15 mM, about 20 mM, about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM, about 50 mM, about 55 mM, or about 60 mM of (e.g., MgSO4, MgCl2, Mg(OAc)2, or Ca(NO3)2). A buffer may comprise a mixture of magnesium and/or calcium salts. Magnesium and/or calcium may be added to a sample to activate amplification reagents.


A buffer may comprise a reducing agent (e.g., NAC, DTT, BME, or TCEP) at a concentration of about 1 mM, about 2 mM, about 3 mM, about 4 mM, about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 10 mM, about 12 mM, about 15 mM, about 20 mM, about 25 mM, about 30 mM, about 40 mM, about 50 mM, about 60 mM, about 7 mM, about 80 mM, about 90 mM, about 100 mM, or about 120 mM. A buffer may comprise a reducing agent (e.g., NAC, DTT, BME, or TCEP) at a concentration of from 1 mM to 5 mM, from 5 mM to 10 mM, from 10 mM to 15 mM, from 15 mM to 20 mM, from 20 mM to 25 mM, from 25 mM to 30 mM, from 30 mM to 40 mM, from 40 mM to 50 mM, from 50 mM to 60 mM, from 60 mM to 70 mM, from 70 mM to 80 mM, or from 80 mM to 90 mM, from 90 mM to 100 mM, or from 100 mM to 120 mM. A buffer may comprise a chelator (e.g., EDTA or EGTA) at a concentration of about 0.1 mM, about 0.2 mM, about 0.3 mM, about 0.4 mM, about 0.5 mM, about 0.6 mM, about 0.7 mM, about 0.8 mM, about 0.9 mM, about 1 mM, about 2 mM, about 3 mM, about 4 mM, about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 10 mM, about 12 mM, about 15 mM, about 20 mM, about 25 mM, or about 30 mM. A buffer may comprise a chelator (e.g., EDTA or EGTA) at a concentration of from 0.1 mM to 0.5 mM, from 0.25 mM to 0.5 mM, from 0.4 mM to 0.6 mM, from 0.5 mM to 1 mM, from 1 mM to 5 mM, from 5 mM to 10 mM, from 10 mM to 15 mM, from 15 mM to 20 mM, from 20 mM to 25 mM, or from 25 mM to 30 mM.


A buffer may comprise a salt (e.g., ammonium acetate ((NH4)2OAc), magnesium acetate (Mg(OAc)2), manganese acetate (MnOAc), potassium acetate (K2OAc), sodium acetate (Na2OAc), ammonium chloride (NH4Cl), potassium chloride (KCl), magnesium chloride (MgCl2), manganese chloride (MnCl2), sodium chloride (NaCl), ammonium sulfate ((NH4)2SO4), magnesium sulfate (MgSO4), manganese sulfate (MnSO4), potassium sulfate (K2SO4), or sodium sulfate (Na2SO4)) at a concentration of about 1 mM, about 5 mM, about 10 mM, about 15 mM, about 20 mM, about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM, about 50 mM, about 55 mM, about 60 mM, about 70 mM, about 80 mM, about 90 mM, or about 100 mM. A buffer may comprise a salt (e.g., (NH4)2OAc, Mg(OAc)2, MnOAc, K2OAc, Na2OAc, NH4Cl, KCl, MgCl2, MnCl2, NaCl, (NH4)2SO4, MgSO4, MnSO4, K2SO4, or Na2SO4) at a concentration of from 1 mM to 5 mM, from 1 mM to 10 mM, from 5 mM to 10 mM, from 10 mM to 15 mM, from 15 mM to 20 mM, from 20 mM to 25 mM, from 25 mM to 30 mM, from 30 mM to 35 mM, from 35 mM to 40 mM, from 40 mM to 45 mM, from 45 mM to 50 mM, from 50 mM to 55 mM, from 55 mM to 60 mM, from 60 mM to 70 mM, from 70 mM to 80 mM, from 80 mM to 90 mM, or from 90 mM to 100 mM.


A buffer may comprise a detergent (e.g., deoxycholate, NP-40 (Ipgal), Triton X-100, or Tween 20) at a concentration of about 0.01%, about 0.05%, about 0.10%, about 0.15%, about 0.20%, about 0.25%, about 0.30%, about 0.35%, about 0.40%, about 0.45%, about 0.50%, about 0.55%, about 0.60%, about 0.65%, about 0.70%, about 0.75%, about 0.80%, about 0.85%, about 0.90%, about 0.95%, about 1.00%, about 1.10%, about 1.20%, about 1.30%, about 1.40%, about 1.50%, about 2.00%, about 2.50%, about 3.00%, about 3.50%, about 4.00%, about 4.50%, or about 5.00%. A buffer may comprise a detergent (e.g., deoxycholate, NP-40 (Ipgal), Triton X-100, or Tween 20) at a concentration of from 0.01% to 0.10%, from 0.05% to 0.15%, from 0.10% to 0.20%, from 0.15% to 0.25%, from 0.20% to 0.30%, from 0.25% to 0.35%, from 0.30% to 0.40%, from 0.35% to 0.45%, from 0.40% to 0.50%, from 0.45% to 0.55%, from 0.50% to 0.60%, from 0.55% to 0.65%, from 0.60% to 0.70%, from 0.65% to 0.75%, from 0.70% to 0.80%, from 0.75% to 0.85%, from 0.80% to 0.90%, from 0.85% to 0.95%, from 0.90% to 1.00%, from 0.95% to 1.10%, from 1.00% to 1.20%, from 1.10% to 1.30%, from 1.20% to 1.40%, from 1.30% to 1.50%, from 1.40% to 1.60%, from 1.50% to 2.00%, from 2.00% to 2.50%, from 2.50% to 3.00%, from 3.00% to 3.50%, from 3.50% to 4.00%, from 4.00% to 4.50%, or from 4.50% to 5.00%.


For example, a buffer comprises 20 mM HEPES pH 7.5, 2.2 mM KOAc, 5.6 mM Mg(OAc)2, 1.1% glycerol, and 0.00018% (volume by volume) Triton-X-100. In some instances the buffer comprises from 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 10 to 20, 10 to 30, 10 to 40, 10 to 50, 10 to 75, 10 to 100, 15 to 20, 15 to 25, 15 to 30, 15 to 40, 15 to 50, 15 to 75, 15 to 100, 20 to 25, 20 to 30, 20 to 40, 20 to 50, 20 to 75, 20 to 100, 30 to 35, 30 to 40, 30 to 50, 30 to 75, 30 to 100, 40 to 45, 40 to 50, 40 to 60, 40 to 75, 40 to 100, 50 to 55, 50 to 60, 50 to 75 or 50 to 100 mM HEPES. The HEPES can be pH 6, pH 6.1, pH 6.2, pH 6.3, pH 6.4, pH 6.5, pH 6.6, pH 6.7, pH 6.8, pH 6.9, pH 7.0, pH 7.1, pH 7.2, pH 7.3, pH 7.4, pH 7.5, pH 7.6, pH 7.7, pH 7.8, pH 7.9, or pH 8.0. The buffer can comprise to 0 to 20, 0 to 18, 0 to 16, 0 to 15, 0 to 14, 0 to 12, 0 to 10, 0 to 7.5, 0 to 5, 0 to 2.5, 0 to 2, 0 to 1.5, 0 to 1, 0.5 to 2, 0.5 to 3, 0.5 to 4, 0.5 to 5, 0.5 to 6, 0.5 to 8, 0.5 to 10, 0.5 to 15, 0.5 to 20, 1 to 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 8, 1 to 10, 1 to 20, 2 to 2.5, 2 to 3, 2 to 4, 2 to 5, 2 to 10, 2 to 20, 2.5 to 3, 2.5 to 5, or 2.2 mM KOAc. In other instances the buffer comprises 0 to 20, 0 to 18, 0 to 16, 0 to 15, 0 to 14, 0 to 12, 0 to 10, 0 to 7.5, 0 to 5, 0 to 2.5, 0 to 2, 0 to 1.5, 0 to 1, 0.5 to 2, 0.5 to 3, 0.5 to 4, 0.5 to 5, 0.5 to 6, 0.5 to 8, 0.5 to 10, 0.5 to 15, 0.5 to 20, 1 to 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 8, 1 to 10, 1 to 20, 2 to 2.5, 2 to 3, 2 to 4, 2 to 5, 2 to 10, 2 to 20, 2.5 to 3, 2.5 to 5, 2.5 to 6, 2.5 to 8, 2.5 to 10, 2.5 to 20, 3 to 4, 3 to 5, 3 to 6, 3 to 8, 3 to 10, 3 to 15, 3 to 20, 4 to 5, 4 to 6, 4 to 8, 4 to 10, 4 to 15, 4 to 20, 5 to 6, 5 to 8, 5 to 10, 5 to 15, 5 to 20, or 5.6 mM Mg(OAc)2. The buffer can comprise 0 to 25, 0 to 20, 0 to 10, 0 to 5, 0 to 4, 0 to 3, 0 to 2, 0 to 1.5, 0.5 to 1, 0.5 to 1.5, 0.5 to 2, 0.5 to 2.5, 0.5 to 3, 1 to 2, 1 to 3, 5 to 10, 5 to 15, 5 to 20, 5 to 25, 5 to 30%, or 1.1% glycerol. The buffer can comprise 0 to 0.0001%, 0 to 0.0002%, 0 to 0.0003%, 0 to 0.0004%, 0 to 0.0005%, 0 to 0.0006%, 0 to 0.0007%, 0 to 0.0008%, 0 to 0.0009%, 0 to 0.001%, 0.0001% to 0.0002%, 0.0001% to 0.0003%, 0.0001% to 0.0004%, 0.0001% to 0.0005%, 0.0001% to 0.0006%, 0.0001% to 0.0007%, 0.0001% to 0.0008%, 0.0001% to 0.0009%, 0.0001% to 0.001%, 0.0002% to 0.0003%, 0.0002% to 0.0004%, 0.0002% to 0.0005%, 0.0002% to 0.0006%, 0.0002% to 0.0007%, 0.0002% to 0.0008%, 0.0002% to 0.0009%, 0.0002% to 0.001%, or 0.00018% Triton-X-100.\


As another example, a buffer comprises 26.49 mM Tris HCl pH 8.8; 13.25 mM (NH4)2SO4, 66.23 mM KCl, 2.65 mM MgSO4, and 0.13% (volume by volume) Tween 20. In some instances the buffer comprises 0 to 200, 0 to 150, 0 to 100, 0 to 75, 0 to 50, 0 to 40, 0 to 30, 0 to 25, 0 to 20, 0 to 15, 0 to 10, 0 to 5, 0 to 2.5, 2.5 to 200, 2.5 to 150, 2.5 to 100, 2.5 to 75, 2.5 to 50, 2.5 to 40, 2.5 to 30, 2.5 to 25, 2.5 to 20, 2.5 to 15, 2.5 to 10, 2.5 to 5, 5 to 200, 5 to 150, 5 to 100, 5 to 75, 5 to 50, 5 to 40, 5 to 30, 5 to 25, 5 to 20, 5 to 15, 5 to 10, 10 to 200, 10 to 150, 10 to 100, 10 to 75, 10 to 50, 10 to 40, 10 to 30, 10 to 25, 10 to 20, 10 to 15, 15 to 200, 15 to 150, 15 to 100, 15 to 75, 15 to 50, 15 to 40, 15 to 30, 15 to 25, 15 to 20, 20 to 200, 20 to 150, 20 to 100, 20 to 75, 20 to 50, 20 to 40, 20 to 30, 20 to 25, 25 to 200, 25 to 150, 25 to 100, 25 to 75, 25 to 50, 25 to 40, 25 to 30, or 26.49 mM Tris HCl. The Tris HCl can be pH 7.6, pH 7.7, pH 7.8, pH 7.9, pH 8.0, pH 8.1, pH 8.2, pH 8.3, pH 8.4, pH 8.5, pH 8.6, pH 8.7, pH 8.8, pH 8.9, pH 9.0, pH 9.1, pH 9.2, pH 9.3, pH 9.4, pH 9.5, pH 9.6, pH 9.7, or pH 9.8. The buffer can comprise 0 to 200, 0 to 150, 0 to 100, 0 to 75, 0 to 50, 0 to 40, 0 to 30, 0 to 25, 0 to 20, 0 to 15, 0 to 10, 0 to 5, 0 to 2.5, 2.5 to 200, 2.5 to 150, 2.5 to 100, 2.5 to 75, 2.5 to 50, 2.5 to 40, 2.5 to 30, 2.5 to 25, 2.5 to 20, 2.5 to 15, 2.5 to 10, 2.5 to 5, 5 to 200, 5 to 150, 5 to 100, 5 to 75, 5 to 50, 5 to 40, 5 to 30, 5 to 25, 5 to 20, 5 to 15, 5 to 10, 10 to 200, 10 to 150, 10 to 100, 10 to 75, 10 to 50, 10 to 40, 10 to 30, 10 to 25, 10 to 20, 10 to 15, 15 to 200, 15 to 150, 15 to 100, 15 to 75, 15 to 50, 15 to 40, 15 to 30, 15 to 25, 15 to 20, 20 to 200, 20 to 150, 20 to 100, 20 to 75, 20 to 50, 20 to 40, 20 to 30, 20 to 25, 25 to 200, 25 to 150, 25 to 100, 25 to 75, 25 to 50, 25 to 40, 25 to 30, or 13.25 mM (NH4)2SO4. In some instances, the buffer comprises between 0 to 500, 0 to 400, 0 to 300, 0 to 200, 0 to 100, 0 to 75, 0 to 50, 0 to 40, 0 to 30, 0 to 20, 0 to 10, 5 to 500, 5 to 400, 5 to 300, 5 to 200, 5 to 100, 5 to 75, 5 to 50, 5 to 40, 5 to 30, 5 to 20, 5 to 10, 10 to 500, 10 to 400, 10 to 300, 10 to 200, 10 to 100, 10 to 75, 10 to 50, 10 to 40, 10 to 30, 10 to 20, 20 to 500, 20 to 400, 20 to 300, 20 to 200, 20 to 100, 20 to 75, 20 to 50, 20 to 40, 20 to 30, 30 to 500, 30 to 400, 30 to 300, 30 to 200, 30 to 100, 30 to 75, 30 to 50, 30 to 40, 40 to 500, 40 to 400, 40 to 300, 40 to 200, 40 to 100, 40 to 75, 40 to 50, 50 to 500, 50 to 400, 50 to 300, 50 to 200, 50 to 100, 50 to 75, 60 to 500, 60 to 400, 60 to 300, 60 to 200, 60 to 100, 60 to 75, 75 to 500, 75 to 400, 75 to 300, 75 to 200, 75 to 100, or 66.23 mM KCl. In some instances, the buffer comprises between 0 to 20, 0 to 18, 0 to 16, 0 to 15, 0 to 14, 0 to 12, 0 to 10, 0 to 7.5, 0 to 5, 0 to 2.5, 0 to 2, 0 to 1.5, 0 to 1, 0.5 to 2, 0.5 to 3, 0.5 to 4, 0.5 to 5, 0.5 to 6, 0.5 to 8, 0.5 to 10, 0.5 to 15, 0.5 to 20, 1 to 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 8, 1 to 10, 1 to 20, 2 to 2.5, 2 to 3, 2 to 4, 2 to 5, 2 to 10, 2 to 20, 2.5 to 3, 2.5 to 5, or 2.65 mM MgSO4. The buffer may comprise between 0 and 2%, 0 and 1%, 0 and 0.9%, 0 and 0.8%, 0 and 0.7%, 0 and 0.6%, 0 and 0.5%, 0 and 0.4%, 0 and 0.3%, 0 and 0.2%, 0 and 0.1%, 0 and 0.05%, 0 and 0.02%, 0.02% and 2%, 0.02% and 1%, 0.02% and 0.9%, 0.02% and 0.8%, 0.02% and 0.7%, 0.02% and 0.6%, 0.02% and 0.5%, 0.02% and 0.4%, 0.02% and 0.3%, 0.02% and 0.2%, 0.02% and 0.1%, 0.02% and 0.05%, 0.05% and 1%, 0.05% and 0.9%, 0.05% and 0.8%, 0.05% and 0.7%, 0.05% and 0.6%, 0.05% and 0.5%, 0.05% and 0.4%, 0.05% and 0.3%, 0.05% and 0.2%, 0.05% and 0.1%, 0.1% and 1%, 0.1% and 0.9%, 0.1% and 0.8%, 0.1% and 0.7%, 0.1% and 0.6%, 0.1% and 0.5%, 0.1% and 0.4%, 0.1% and 0.3%, 0.1% and 0.2%, 0.2% and 1%, 0.2% and 0.9%, 0.2% and 0.8%, 0.2% and 0.7%, 0.2% and 0.6%, 0.2% and 0.5%, 0.2% and 0.4%, 0.2% and 0.3%, 0.3% and 1%, 0.3% and 0.9%, 0.3% and 0.8%, 0.3% and 0.7%, 0.3% and 0.6%, 0.3% and 0.5%, 0.3% and 0.4%, 0.4% and 1%, 0.4% and 0.9%, 0.4% and 0.8%, 0.4% and 0.7%, 0.4% and 0.6%, 0.4% and 0.5%, 0.5% and 1%, 0.5% and 0.9%, 0.5% and 0.8%, 0.5% and 0.7%, 0.5% and 0.6%, or 0.13% Tween 20.


A buffer may be optimized to support multiple reactions. A challenge associated with nucleic acid detection is that many amplification and nucleic acid detection reactions require specific conditions, such as buffer type, ionic strength, viscosity, pH, osmolarity, and dielectric constant. In many cases, these conditions are not cross compatible between separate reactions (e.g., an amplification reaction and a DETECTR reaction). An aspect of the present disclosure provides buffer compositions that are suitable for supporting amplification and CRISPR-Cas enzyme (e.g., DETECTR) reactions. Such buffers may also support additional types of reactions, such as viral lysis, cellular lysis, reverse transcription, transcription, and demethylation. In some instances, a buffer may support fast amplification, CRISPR, and viral lysis reactions. In particular instances, a buffer may support fast RT-LAMP, transcollateral cleavage (e.g., DETECTR), and viral lysis reactions.


In many cases, the buffer composition allows the amplification and CRISPR reactions to approach completion at the same rate or close to its optimal rate (e.g., in a buffer optimized solely for one type of reaction). For example, a buffer may enable an amplification reaction to proceed at least at 90% of its optimal rate and a DETECTR reaction to proceed at least at 85% of its optimal rate. A buffer may enable an amplification reaction to proceed at least at 85% of its optimal rate and a DETECTR reaction to proceed at least at 85% of its optimal rate. A buffer may enable an amplification reaction to proceed at least at 80% of its optimal rate and a DETECTR reaction to proceed at least at 90% of its optimal rate. A buffer may enable an amplification reaction and a DETECTR reaction to each proceed at least at 45% of their optimal rates. A buffer may enable an amplification reaction and a DETECTR reaction to each proceed at least at 50% of their optimal rates. A buffer may enable an amplification reaction and a DETECTR reaction to each proceed at least at 60% of their optimal rates. A buffer may enable an amplification reaction and a DETECTR reaction to each proceed at least at ⅔ of their optimal rates. A buffer may enable an amplification reaction and a DETECTR reaction to each proceed at least at 75% of their optimal rates. A buffer may enable an amplification reaction and a DETECTR reaction to each proceed at least at 80% of their optimal rates. A buffer may enable an amplification reaction and a DETECTR reaction to each proceed at least at 85% of their optimal rates. A buffer may enable an amplification reaction and a DETECTR reaction to each proceed at least at 90% of their optimal rates.


A buffer may enable an amplification reaction and a DETECTR reaction to each reach completion within 2 hours. A buffer may enable an amplification reaction and a DETECTR reaction to each reach completion within 1.5 hours. A buffer may enable an amplification reaction and a DETECTR reaction to each reach completion within 1.2 hours. A buffer may enable an amplification reaction and a DETECTR reaction to each reach completion within 1 hour. A buffer may enable an amplification reaction and a DETECTR reaction to each reach completion within 45 minutes. A buffer may enable an amplification reaction and a DETECTR reaction to each reach completion within 30 minutes. A buffer may enable an amplification reaction and a DETECTR reaction to each reach completion within 20 minutes. A buffer may enable an amplification reaction and a DETECTR reaction to each reach completion within 15 minutes. A buffer may enable an amplification reaction and a DETECTR reaction to each reach completion within 10 minutes. As used herein, the term ‘reach completion’ can refer to a reaction consuming at least about 80%, at least about 90%, at least about 95%, at least about 99%, or 100% of a limiting reagent (e.g., a reporter, a primer, a dNTP or rNTP, or an amplicon). The term ‘reach completion’ can also refer to a point in time where the rate of product production has diminished from its maximum rate to less than 5%, 2%, 1%, or 0.5% its maximum rate.


A buffer may comprise an amplification activator. A set of amplification reagents may require an inorganic, molecular, or biological species to initiate an amplification reaction. A buffer many comprise an amplification activator. Conversely, an amplification activator may be provided separately (e.g., in a separate vial in a kit) and added to a reaction mixture at a desired timepoint. An amplification activator may comprise a calcium or magnesium salt (e.g., CaOAc or Mg(OAc)2).


In some cases, a buffer may comprise a catalytic reagent for signal improvement or enhancement. In some cases, the catalytic reagent may enhance signal generation via hydrolysis of inorganic pyrophosphates. In some cases, the catalytic reagent may enhance signal generation via enhancement of DNA replication. In some cases, the catalytic reagent may enhance signal amplification via revival of Mg+2 ions in the buffer solution which may otherwise be taken up by the phosphates produced from usage of dNTPs during the LAMP reaction. In some cases, the catalytic reagent may enhance signal generation by reviving the concentration of Mg+2 ions in the buffer thereby enhancing the function of the Cas nuclease effector enzyme. In some cases, the catalytic reagent for signal improvement may be an enzyme. In some cases, the catalytic reagent for signal improvement may be a Thermostable Inorganic Pyrophosphatase (TIPP).


Any of the systems, methods, or devices described herein may comprise thermostable inorganic pyrophosphatase (TIPP). In some embodiments, a composition for an assay may comprise TIPP. In some embodiments, the composition may comprise about 0.5 enzyme unit (U) per 10 μL of solution. In some embodiments, the composition may comprise at least about 0.1 U per 10 μL of solution. In some embodiments, the composition may comprise at most about 2 U per 10 μL of solution. In some embodiments, the composition may comprise at least about 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 U per 10 μL of solution. In some embodiments, the composition may comprise at most about 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 U per 10 μL of solution.


In some embodiments, TIPP may improve the signal to noise ratio of a programmable nuclease-based detection reaction. In some embodiments, TIPP may improve overall signal (e.g., fluorescence of a cleaved reporter as shown in FIG. 65 or intensity of a lateral flow assay strip detection location as shown in FIGS. 66A-66B). TIPP may improve signal by a factor, wherein the signal is indicative of the presence of a target nucleic acid. In some embodiments, the factor may be at least about 1.1. In some embodiments, the factor may be at least about 2. In some embodiments, the factor may be at least about 3. In some embodiments, the factor may be at least about 4. In some embodiments, the factor may be at least about 5. In some embodiments, the factor may be at least about 6. In some embodiments, the factor may be at least about 7. In some embodiments, the factor may be at least about 8. In some embodiments, the factor may be at least about 9. In some embodiments, the factor may be at least about 10.


In some cases, the TIPP enzyme may be present in the HotPot reaction mix at a concentration of 0.125 Units, 0.5 Units, 0.25 Units, 1.0 Units, 2.0 Units, 2.5 Units, or 4 Units per discrete reaction volume. In some cases, the buffer may comprise the TIPP signal enhancement reagent. Conversely, the TIPP signal enhancement reagent may be provided separately (e.g., in a separate vial in a kit) and added to a reaction mixture at a desired timepoint.


A buffer or reaction volume (e.g., a solution comprising reagents, such as a buffer comprising amplification and DETECTR reagents) may comprise varying amounts of total dissolved solids. A buffer may comprise (excluding reagents) at least 50 mM of total dissolved solids. A buffer may comprise (excluding reagents) at least 80 mM of total dissolved solids. A buffer may comprise (excluding reagents) at least 100 mM of total dissolved solids. A buffer may comprise (excluding reagents) at least 120 mM of total dissolved solids. A buffer may comprise (excluding reagents) at least 150 mM of total dissolved solids. A buffer may comprise (excluding reagents) at least 200 mM of total dissolved solids. A buffer may comprise (excluding reagents) at least 250 mM of total dissolved solids. A buffer may comprise (excluding reagents) at least 300 mM of total dissolved solids. A buffer may comprise (excluding reagents) at least 400 mM of total dissolved solids. A buffer may comprise (excluding reagents) at least 500 mM of total dissolved solids. A buffer may comprise (excluding reagents) at least 600 mM of total dissolved solids. A buffer may comprise (excluding reagents) at least 800 mM of total dissolved solids.


A buffer may comprise a viscosity of at least 1.5 centipoise (cP), at least 2 cP, at least 2.5 cP, at least 3 cP, at least 3.5 cP, at least 4 cP, at least 5 cP, at least 6 cP, at least 8 cP, at least 10 cP, at least 12 cP, at least 15 cP, at least 18 cP, at least 24 cP, at least 30 cP, at least 40 cP, or at least 50 cP at room temperature. High viscosities may stabilize enzymes and other reagents for amplification and programmable nuclease enzyme (e.g., CRISPR-Cas enzyme) reactions. High viscosities may also favor faster reaction rates for certain enzymes.


For example, a buffer comprises 20 mM HEPES pH 6.8, 50 mM KCl, 5 mM MgCl2, and 5% glycerol. In some instances the buffer comprises from 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 10 to 20, 10 to 30, 10 to 40, 10 to 50, 15 to 20, 15 to 25, 15 to 30, 15 to 4, 15 to 50, 20 to 25, 20 to 30, 20 to 40, or 20 to 50 mM HEPES pH 6.8. The buffer can comprise to 0 to 500, 0 to 400, 0 to 300, 0 to 250, 0 to 200, 0 to 150, 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 5 to 150, 5 to 200, 5 to 250, 5 to 300, 5 to 400, 5 to 500, 25 to 50, 25 to 75, 25 to 100, 50 to 100, 50 150, 50 to 200, 50 to 250, 50 to 300, 100 to 200, 100 to 250, 100 to 300, or 150 to 250 mM KCl. In other instances the buffer comprises 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 10 to 20, 10 to 30, 10 to 40, 10 to 50, 15 to 20, 15 to 25, 15 to 30, 15 to 4, 15 to 50, 20 to 25, 20 to 30, 20 to 40, or 20 to 50 mM MgCl2. The buffer can comprise 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, 5 to 30% glycerol.


As another example, a buffer comprises 100 mM Imidazole pH 7.5; 250 mM KCl, 25 mM MgCl2, 50 ug/mL BSA, 0.05% Igepal Ca-630, and 25% Glycerol. In some instances the buffer comprises 0 to 500, 0 to 400, 0 to 300, 0 to 250, 0 to 200, 0 to 150, 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 5 to 150, 5 to 200, 5 to 250, 5 to 300, 5 to 400, 5 to 500, 25 to 50, 25 to 75, 25 to 100, 50 to 100, 50 150, 50 to 200, 50 to 250, 50 to 300, 100 to 200, 100 to 250, 100 to 300, or 150 to 250 mM Imidazole pH 7.5. The buffer can comprise to 0 to 500, 0 to 400, 0 to 300, 0 to 250, 0 to 200, 0 to 150, 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 5 to 150, 5 to 200, 5 to 250, 5 to 300, 5 to 400, 5 to 500, 25 to 50, 25 to 75, 25 to 100, 50 to 100, 50 150, 50 to 200, 50 to 250, 50 to 300, 100 to 200, 100 to 250, 100 to 300, or 150 to 250 mM KCl. In other instances the buffer comprises 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 10 to 20, 10 to 30, 10 to 40, 10 to 50, 15 to 20, 15 to 25, 15 to 30, 15 to 4, 15 to 50, 20 to 25, 20 to 30, 20 to 40, or 20 to 50 mM MgCl2. The buffer, in some instances, comprises 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 50, 5 to 75, 5 to 100, 10 to 20, 10 to 50, 10 to 75, 10 to 100, 25 to 50, 25 to 75 25 to 100, 50 to 75, or 50 to 100 ug/mL BSA. In some instances, the buffer comprises 0 to 1, 0 to 0.5, 0 to 0.25, 0 to 0.01, 0 to 0.05, 0 to 0.025, 0 to 0.01, 0.01 to 0.025, 0.01 to 0.05, 0.01 to 0.1, 0.01 to 0.25, 0.01, to 0.5, 0.01 to 1, 0.025 to 0.05, 0.025 to 0.1, 0.025, to 0.5, 0.025 to 1, 0.05 to 0.1, 0.05 to 0.25, 0.05 to 0.5, 0.05 to 0.75, 0.05 to 1, 0.1 to 0.25, 0.1 to 0.5, or 0.1 to 1% Igepal Ca-630. The buffer can comprise 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, 5 to 30% glycerol.


In some embodiments, a buffer comprises a pH of between 8 and 8.8, 20 mM Tris-HCl, 2 to 10 mM (NH4)2SO4, 2 to 50 mM KCl, 2 to 6.5 mM MgSO4, and 0.1% Tween20. In some embodiments, a buffer comprises a pH of 7.5, 20 mM HEPES, 20 mM KOAc, 5 mM MG(OAc)2, 1% glycerol and 0.00016% Triton X-100. In some embodiments, a buffer comprises a pH of between 7.5 and 8.25 phosphate buffer, between 0 and 2 mM (NH4)2SO4, 2 to 30 mM KCl, 5 mM Mg(OAc)2, 1% glycerol, and either 0.1% Tween 20 or 0.00016% Triton x-100. In some embodiments, a buffer comprises a pH of between 7.5 and 8.25, 0 to 2 mM (NH4)2SO44, 15 to 30 mM KOAc, 2 to 5 mM Mg(OAc)2, 1% glycerol and 0.1% Tween 20. In some embodiments, a buffer comprises a pH of 8, 10 mM phosphate, 0 to 2 mM (NH4)2SO4, 20 mM KOAc, 5 mM Mg(OAc)2, 1% glycerol, and 0.1% Tween 20.


A buffer of the present disclosure may comprise a viral lysis buffer. A viral lysis buffer may lyse a coronavirus capsid in a viral sample (e.g., a sample collected from an individual suspected of having a coronavirus infection), releasing a viral genome. The viral lysis buffer may be compatible with amplification (e.g., RT-LAMP amplification) of a target region of the viral genome. The viral lysis buffer may be compatible with detection (e.g., a DETECTR reaction disclosed herein). A sample may be prepared in a one-step sample preparation method comprising suspending the sample in a viral lysis buffer compatible with amplification, detection (e.g., a DETECTR reaction), or both. A viral lysis buffer compatible with amplification (e.g., RT-LAMP amplification), detection (e.g., DETECTR), or both, may comprise a buffer (e.g., Tris-HCl, phosphate, or HEPES), a reducing agent (e.g., N-Acetyl Cysteine (NAC), Dithiothreitol (DTT), β-mercaptoethanol (BME), or tris(2-carboxyethyl)phosphine (TCEP)), a chelating agent (e.g., EDTA or EGTA), a detergent (e.g., deoxycholate, NP-40 (Ipgal), Triton X-100, or Tween 20), a salt (e.g., ammonium acetate, magnesium acetate, manganese acetate, potassium acetate, sodium acetate, ammonium chloride, potassium chloride, magnesium chloride, manganese chloride, sodium chloride, ammonium sulfate, magnesium sulfate, manganese sulfate, potassium sulfate, or sodium sulfate), or a combination thereof. For example, a viral lysis buffer may comprise a buffer and a reducing agent, or a viral lysis buffer may comprise a buffer and a chelating agent. The viral lysis buffer may be formulated at a low pH. For example, the viral lysis buffer may be formulated at a pH of from about pH 4 to about pH 5. In some embodiments, the viral lysis buffer may be formulated at a pH of from about pH 4 to about pH 9. The viral lysis buffer may further comprise a preservative (e.g., ProClin 150). In some embodiments, the viral lysis buffer may comprise an activator of the amplification reaction. For example, the buffer may comprise primers, dNTPs, or magnesium (e.g., MgSO4, MgCl2 or Mg(OAc)2), or a combination thereof, to activate the amplification reaction. In some embodiments, an activator (e.g., primers, dNTPs, or magnesium) may be added to the buffer following lysis of the coronavirus to initiate the amplification reaction.


A viral lysis buffer may comprise a pH of about 3.5, about 3.6, about 3.7, about 3.8, about 3.9, about 4, about 4.1, about 4.2, about 4.3, about 4.4, about 4.5, about 4.6, about 4.7, about 4.8, about 4.9, about 5, about 5.1, about 5.2, about 5.3, about 5.4, about 5.5, about 6, about 6.5, about 7, about 7.5, about 8, about 8.5, or about 9. In some embodiments, a viral lysis buffer may comprise a pH of from 3.5 to 4.5, from 4 to 5, from 4.5 to 5.5, from 3.5 to 4, from 4 to 4.5, from 4.5 to 5, from 5 to 5.5, from 5 to 6, from 6 to 7, from 7 to 8, or from 8 to 9.


A viral lysis buffer may comprise a magnesium concentration of about 0 mM, about 2 mM, about 4 mM, about 5 mM, about 6 mM, about 8 mM, about 10 mM, about 12 mM, about 13 mM, about 14 mM, about 15 mM, about 20 mM, about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM, about 50 mM, about 55 mM, or about 60 mM of magnesium (e.g., MgSO4, MgCl2 or Mg(OAc)2. A viral lysis buffer may comprise a magnesium concentration of from 0 mM to 5 mM, from 5 mM to 10 mM, from 10 mM to 15 mM, from 15 mM to 20 mM, from 20 mM to 25 mM, from 25 mM to 30 mM, from 30 mM to 40 mM, from 40 mM to 50 mM, or from 50 mM to 60 mM of magnesium (e.g., MgSO4, MgCl2 or Mg(OAc)2). In some embodiments, the magnesium may be added after viral lysis to activate an amplification reaction.


A viral lysis buffer may comprise a reducing agent (e.g., NAC, DTT, BME, or TCEP) at a concentration of about 1 mM, about 2 mM, about 3 mM, about 4 mM, about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 10 mM, about 12 mM, about 15 mM, about 20 mM, about 25 mM, about 30 mM, about 40 mM, about 50 mM, about 60 mM, about 7 mM, about 80 mM, about 90 mM, about 100 mM, or about 120 mM. A viral lysis buffer may comprise a reducing agent (e.g., NAC, DTT, BME, or TCEP) at a concentration of from 1 mM to 5 mM, from 5 mM to 10 mM, from 10 mM to 15 mM, from 15 mM to 20 mM, from 20 mM to 25 mM, from 25 mM to 30 mM, from 30 mM to 40 mM, from 40 mM to 50 mM, from 50 mM to 60 mM, from 60 mM to 70 mM, from 70 mM to 80 mM, or from 80 mM to 90 mM, from 90 mM to 100 mM, or from 100 mM to 120 mM. A viral lysis buffer may comprise a chelator (e.g., EDTA or EGTA) at a concentration of about 0.1 mM, about 0.2 mM, about 0.3 mM, about 0.4 mM, about 0.5 mM, about 0.6 mM, about 0.7 mM, about 0.8 mM, about 0.9 mM, about 1 mM, about 2 mM, about 3 mM, about 4 mM, about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 10 mM, about 12 mM, about 15 mM, about 20 mM, about 25 mM, or about 30 mM. A viral lysis buffer may comprise a chelator (e.g., EDTA or EGTA) at a concentration of from 0.1 mM to 0.5 mM, from 0.25 mM to 0.5 mM, from 0.4 mM to 0.6 mM, from 0.5 mM to 1 mM, from 1 mM to 5 mM, from 5 mM to 10 mM, from 10 mM to 15 mM, from 15 mM to 20 mM, from 20 mM to 25 mM, or from 25 mM to 30 mM.


A viral lysis buffer may comprise a salt (e.g., ammonium acetate ((NH4)2OAc), magnesium acetate (Mg(OAc)2), manganese acetate (MnOAc), potassium acetate (K2OAc), sodium acetate (Na2OAc), ammonium chloride (NH4Cl), potassium chloride (KCl), magnesium chloride (MgCl2), manganese chloride (MnCl2), sodium chloride (NaCl), ammonium sulfate ((NH4)2SO4), magnesium sulfate (MgSO4), manganese sulfate (MnSO4), potassium sulfate (K2SO4), or sodium sulfate (Na2SO4)) at a concentration of about 1 mM, about 5 mM, about 10 mM, about 15 mM, about 20 mM, about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM, about 50 mM, about 55 mM, about 60 mM, about 70 mM, about 80 mM, about 90 mM, or about 100 mM. A viral lysis buffer may comprise a salt (e.g., (NH4)2OAc, Mg(OAc)2, MnOAc, K2OAc, Na2OAc, NH4Cl, KCl, MgCl2, MnCl2, NaCl, (NH4)2SO4, MgSO4, MnSO4, K2SO4, or Na2SO4) at a concentration of from 1 mM to 5 mM, from 1 mM to 10 mM, from 5 mM to 10 mM, from 10 mM to 15 mM, from 15 mM to 20 mM, from 20 mM to 25 mM, from 25 mM to 30 mM, from 30 mM to 35 mM, from 35 mM to 40 mM, from 40 mM to 45 mM, from 45 mM to 50 mM, from 50 mM to 55 mM, from 55 mM to 60 mM, from 60 mM to 70 mM, from 70 mM to 80 mM, from 80 mM to 90 mM, or from 90 mM to 100 mM.


A viral lysis buffer may comprise a detergent (e.g., deoxycholate, NP-40 (Ipgal), Triton X-100, or Tween 20) at a concentration of about 0.01%, about 0.05%, about 0.10%, about 0.15%, about 0.20%, about 0.25%, about 0.30%, about 0.35%, about 0.40%, about 0.45%, about 0.50%, about 0.55%, about 0.60%, about 0.65%, about 0.70%, about 0.75%, about 0.80%, about 0.85%, about 0.90%, about 0.95%, about 1.00%, about 1.10%, about 1.20%, about 1.30%, about 1.40%, about 1.50%, about 2.00%, about 2.50%, about 3.00%, about 3.50%, about 4.00%, about 4.50%, or about 5.00%. A viral lysis buffer may comprise a detergent (e.g., deoxycholate, NP-40 (Ipgal), Triton X-100, or Tween 20) at a concentration of from 0.01% to 0.10%, from 0.05% to 0.15%, from 0.10% to 0.20%, from 0.15% to 0.25%, from 0.20% to 0.30%, from 0.25% to 0.35%, from 0.30% to 0.40%, from 0.35% to 0.45%, from 0.40% to 0.50%, from 0.45% to 0.55%, from 0.50% to 0.60%, from 0.55% to 0.65%, from 0.60% to 0.70%, from 0.65% to 0.75%, from 0.70% to 0.80%, from 0.75% to 0.85%, from 0.80% to 0.90%, from 0.85% to 0.95%, from 0.90% to 1.00%, from 0.95% to 1.10%, from 1.00% to 1.20%, from 1.10% to 1.30%, from 1.20% to 1.40%, from 1.30% to 1.50%, from 1.40% to 1.60%, from 1.50% to 2.00%, from 2.00% to 2.50%, from 2.50% to 3.00%, from 3.00% to 3.50%, from 3.50% to 4.00%, from 4.00% to 4.50%, or from 4.50% to 5.00%.


A lysis reaction may be performed at a range of temperatures. In some embodiments, a lysis reaction may be performed at about room temperature. In some embodiments, a lysis reaction may be performed at about 95° C. In some embodiments, a lysis reaction may be performed at from 1° ° C. to 10° C., from 4° ° C. to 8° C., from 10° ° C. to 20° C., from 15° ° C. to 25° C., from 15° C. to 20° C., from 18° C. to 25° C., from 18° C. to 95° C., from 20° C. to 37° C., from 25° C. to 40° C., from 35° C. to 45° C., from 40° ° C. to 60° C., from 50° C. to 70° C., from 60° ° C. to 80° C., from 70° C. to 90° C., from 80° C. to 95° C., or from 90° C. to 99° C. In some embodiments, a lysis reaction may be performed for about 5 minutes, about 15 minutes, or about 30 minutes. In some embodiments, a lysis reaction may be performed for from 2 minutes to 5 minutes, from 3 minutes to 8 minutes, from 5 minutes to 15 minutes, from 10 minutes to 20 minutes, from 15 minutes to 25 minutes, from 20 minutes to 30 minutes, from 25 minutes to 35 minutes, from 30 minutes to 40 minutes, from 35 minutes to 45 minutes, from 40 minutes to 50 minutes, from 45 minutes to 55 minutes, from 50 minutes to 60 minutes, from 55 minutes to 65 minutes, from 60 minutes to 70 minutes, from 65 minutes to 75 minutes, from 70 minutes to 80 minutes, from 75 minutes to 85 minutes, or from 80 minutes to 90 minutes.


The reagents described herein can also include buffers, which are compatible with the devices, systems, fluidic devices, kits, and methods disclosed herein. The buffers described herein are compatible for use in the devices described herein (e.g., pneumatic valve devices, sliding valve devices, rotating valve devices, and lateral flow devices) and may be used in conjunction with compositions disclosed herein (e.g., programmable nucleases, guide nucleic acids, reagents for in vitro transcription, reagents for amplification, reagents for reverse transcription, reporters, or any combination thereof) to carry out highly efficient, rapid, and accurate reactions for detecting whether the target nucleic acid is in the sample (e.g., DETECTR reactions). These buffers are compatible with the other reagents, samples, and support mediums as described herein for detection of an ailment, such as a disease, cancer, or genetic disorder, or genetic information, such as for phenotyping, genotyping, or determining ancestry. The methods described herein can also include the use of buffers, which are compatible with the methods disclosed herein. For example, a buffer may comprise HEPES, MES, TCEP, EGTA, Tween 20, KCl, MgCl2, glycerol, or any combination thereof. In some instances, a buffer may comprise Tris-HCl pH 8.8, VLB, EGTA, CH3COOH, TCEP, IsoAmp, (NH4)2SO4, KCl, MgSO4, Tween20, KOAc, MgOAc, BSA, TCEP, or any combination thereof. In some instances the buffer may comprise from 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 10 to 20, 10 to 30, 10 to 40, 10 to 50, 15 to 20, 15 to 25, 15 to 30, 15 to 4, 15 to 50, 20 to 25, 20 to 30, 20 to 40, or 20 to 50 mM HEPES pH 6.8. The buffer can comprise to 0 to 500, 0 to 400, 0 to 300, 0 to 250, 0 to 200, 0 to 150, 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 5 to 150, 5 to 200, 5 to 250, 5 to 300, 5 to 400, 5 to 500, 25 to 50, 25 to 75, 25 to 100, 50 to 100, 50 150, 50 to 200, 50 to 250, 50 to 300, 100 to 200, 100 to 250, 100 to 300, or 150 to 250 mM KCl. In other instances the buffer may comprise 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 10 to 20, 10 to 30, 10 to 40, 10 to 50, 15 to 20, 15 to 25, 15 to 30, 15 to 4, 15 to 50, 20 to 25, 20 to 30, 20 to 40, or 20 to 50 mM MgCl2. The buffer can comprise 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, 5 to 30% glycerol. The buffer can comprise from 0% to 30%, from 5% to 30%, from 10% to 30%, from 15% to 30%, from 20% to 30%, from 25% to 30%, from 0% to 25%, from 2% to 25%, from 5% to 25%, from 10% to 25%, from 15% to 25%, from 20% to 25%, from 0% to 20%, from 5% to 20%, from 10% to 20%, from 15% to 20%, from 0% to 15%, from 5% to 15%, from 10% to 15%, from 0% to 10%, from 5% to 10%, or from 0% to 5% glycerol. The buffer can comprise to 0 to 500, 0 to 400, 0 to 300, 0 to 250, 0 to 200, 0 to 150, 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 5 to 150, 5 to 200, 5 to 250, 5 to 300, 5 to 400, 5 to 500, 25 to 50, 25 to 75, 25 to 100, 50 to 100, 50 150, 50 to 200, 50 to 250, 50 to 300, 100 to 200, 100 to 250, 100 to 300, or 150 to 250 mM Tris-HCl pH 8.8. The buffer can comprise to 0 to 500, 0 to 400, 0 to 300, 0 to 250, 0 to 200, 0 to 150, 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 5 to 150, 5 to 200, 5 to 250, 5 to 300, 5 to 400, 5 to 500, 25 to 50, 25 to 75, 25 to 100, 50 to 100, 50 150, 50 to 200, 50 to 250, 50 to 300, 100 to 200, 100 to 250, 100 to 300, or 150 to 250 mM KOAc. The buffer can comprise to 0 to 500, 0 to 400, 0 to 300, 0 to 250, 0 to 200, 0 to 150, 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 5 to 150, 5 to 200, 5 to 250, 5 to 300, 5 to 400, 5 to 500, 25 to 50, 25 to 75, 25 to 100, 50 to 100, 50 150, 50 to 200, 50 to 250, 50 to 300, 100 to 200, 100 to 250, 100 to 300, or 150 to 250 mM MgOAc. In some instances the buffer may comprise from 0 to 100, 0 to 75, 0 to 50, 0 to 25, 0 to 20, 0 to 10, 0 to 5, 5 to 10, 5 to 15, 5 to 20, 5 to 25, to 30, 5 to 40, 5 to 50, 5 to 75, 5 to 100, 10 to 20, 10 to 30, 10 to 40, 10 to 50, 15 to 20, 15 to 25, 15 to 30, 15 to 4, 15 to 50, 20 to 25, 20 to 30, 20 to 40, or 20 to 50 mM EGTA. The buffer can comprise from 0% to 30%, from 5% to 30%, from 10% to 30%, from 15% to 30%, from 20% to 30%, from 25% to 30%, from 0% to 25%, from 2% to 25%, from 5% to 25%, from 10% to 25%, from 15% to 25%, from 20% to 25%, from 0% to 20%, from 5% to 20%, from 10% to 20%, from 15% to 20%, from 0% to 15%, from 5% to 15%, from 10% to 15%, from 0% to 10%, from 5% to 10%, or from 0% to 5% Tween 20.


Compositions Comprising One or More Additives for Improving Assay Signals

In some embodiments, the reagents described herein may include a composition for improving detection signal strength, detection reaction time, detection reaction efficiency, stability, solubility, or the like. In some embodiments, the composition may comprise one or more additives. The one or more additives may, for example, comprise amino acids or derivatives thereof, chaotrpes, chelators, cyclodextrins, inhibitors, ionic liquids, linkers, metals, non detergent sulfobetaines, organic acids, osmolytes, peptides, polyamides, polymers, polyols, polyols and salts, salts, or combinations thereof. In some embodiments, the one or more additives may, for example, comprise one or more of trichloroacetic acid, L-Arginine, L-Glutamic acid, glycine, L-Proline, L-Histidine, beta (β)-Alanine, L-Serine, L-Arginine ethyl ester dihydrochloride, L-Argininamide dihydrochloride, 6-Aminohexanoic acid, Gly-gly peptide, Gly-gly-gly peptide, tryptone, betaine monohydrate, D-(+)-Trehalose dihydrate, Xylitol, D-Sorbitol, sucrose, hydroxyectoine, Trimethylamine N-oxide dihydrate, methyl alpha (α)-D-gluocopyranoside, triethylene glycol, spermine tetrahydrochloride, spermidine, 5-aminovaleric acid, glutaric acid, adipic acid, ethylenediamine dihydrochloride, guanidine hydrochloride, urea, N-methylurea, N-ethylurea, N-methylformamide, hypotauring, TCEP hydrochloride, GSH (L-Glutathione reduced), GSSG (L-Glutathione oxidized), benzaminidine hydrochloride, ethylenediaminetetraacetic acid disodium salt dihydrate, magnesium chloride hexahydrate, calcium chloride dihydrate, cadmium chloride hydrate, cobalt(II) chloride hexahydrate, Non Detergent Sulfobetaine 195 (NDSB-195), NDSB-201, NDSB-211, NDSB-221, NDSB-256, taurine, acetamide, oxalic acid dihydrate, sodium malonate pH 7.0, succinic acid pH 7.0, tacsimate pH 7.0, tetraethylammonium bromide, cholin acetate, 1-Ethyl-3-methylimidazolium acetate, 1-Butyl-3-methylimidazolium chloride, ethylammonium nitrate, ammonium sulfate, ammonium chloride, magnesium sulfate hydrate, potassium thiocynate, gadolinium(III) chloride hexahydrate, cesium chloride, 4-aminobutyric acid (GABA), lithium nitrate, DL-malic acid pH 7.0, lithium citrate tribasic tetrahydrate, ammonium acetate, sodium benzenesulfonate, sodium p-toluenesulfonate, sodium chloride, potassium chloride, sodium phosphate monobasic monohydrate, potassium phosphate dibasic, sodium sulfate decahydrate, lithium chloride, sodium bromide, glycerol, ethylene glycol, polyethylene glycol 200 (PEG-200), PEG 3350, PEG 8000, PEG monomethyl ether 550, PEG monomethyl ether 750, PEG monomethyl ether 1900, formamide, polypropylene glycol P 400, pentaerythritol ethoxylate, 1,2-Propanediol, polyvinylpyrrolidone K 15, 6-O-α-Maltosyl-β-cyclodextrin, (2-Hydroxypropyl)-β-cyclodextrin, α-cyclodextrin, β-cyclodextrin, Methyl-β-cyclodextrin, or any combination thereof.


In some embodiments, the reagents described herein may include a composition for increasing the signal strength of any of the assays described herein. In some embodiments, the composition comprises water and an additive. In some embodiments, the additive may comprise trehalose, xylitol, D-sorbitol, sucrose, and trimethylamine N-oxide dihydrate, or any combination thereof. In some embodiments, the composition further comprises one or more targets, one or more enzymes, one or more reporters, one or more substrates, or any combination thereof as described herein.


In some embodiments, the reagents described herein may include a composition for reducing the time it takes for the signal of any of the assays described herein to saturate. In some embodiments, the composition comprises an additive. In some embodiments, the additive may comprise betaine monohydrate, acetamide, GABA, L-proline, beta-alanine, 6-aminohexanonic acid, urea, methylurea, ethylurea, hypotaurine, NDSB-256, ammonium acetate, or any combination thereof. In some embodiments, the composition further comprises one or more targets, one or more enzymes, one or more reporters, one or more substrates, or any combination thereof as described herein.


In some embodiments, the additive is present at a concentration of at least about 1 nM, at least about 2 nM, at least about 3 nM, at least about 4 nM, at least about 5 nM, at least about 6 nM, at least about 7 nM, at least about 8 nM, at least about 9 nM, at least about 10 nM, at least about 20 nM, at least about 30 nM, at least about 40 nM, at least about 50 nM, at least about 60 nM, at least about 70 nM, at least about 80 nM, at least about 90 nM, at least about 100 nM, at least about 200 nM, at least about 300 nM, at least about 400 nM, at least about 500 nM, at least about 600 nM, at least about 700 nM, at least about 800 nM, at least about 900 nM, at least about 1 μM, at least about 2 μM, at least about 3 μM, at least about 4 μM, at least about 5 μM, at least about 6 M, at least about 7 μM, at least about 8 μM, at least about 9 μM, at least about 10 μM, at least about 20 μM, at least about 30 μM, at least about 40 μM, at least about 50 μM, at least about 60 μM, at least about 70 μM, at least about 80 μM, at least about 90 μM, at least about 100 μM, at least about 200 μM, at least about 300 μM, at least about 400 μM, at least about 500 μM, at least about 600 μM, at least about 700 μM, at least about 800 μM, at least about 900 μM, at least about 1 mM, at least about 2 mM, at least about 3 mM, at least about 4 mM, at least about 5 mM, at least about 6 mM, at least about 7 mM, at least about 8 mM, at least about 9 mM, at least about 10 mM, at least about 20 mM, at least about 30 mM, at least about 40 mM, at least about 50 mM, at least about 60 mM, at least about 70 mM, at least about 80 mM, at least about 90 mM, at least about 100 mM, at least about 200 mM, at least about 300 mM, at least about 400 mM, at least about 500 mM, at least about 600 mM, at least about 700 mM, at least about 800 mM, at least about 900 mM, at least about 1 M, at least about 2 M, at least about 3 M, at least about 4 M, at least about 5 M, at least about 6 M, at least about 7 M, at least about 8 M, at least about 9 M, or at least about 10 M.


In some embodiments, the composition increases the signal strength by at least about a factor of 1.01, at least about a factor of 1.02, at least about a factor of 1.03, at least about a factor of 1.04, at least about a factor of 1.05, at least about a factor of 1.06, at least about a factor of 1.07, at least about a factor of 1.08, at least about a factor of 1.09, at least about a factor of 1.1, at least about a factor of 1.2, at least about a factor of 1.3, at least about a factor of 1.4, at least about a factor of 1.5, at least about a factor of 1.6, at least about a factor of 1.7, at least about a factor of 1.8, at least about a factor of 1.9, at least about a factor of 2, at least about a factor of 3, at least about a factor of 4, at least about a factor of 5, at least about a factor of 6, at least about a factor of 7, at least about a factor of 8, at least about a factor of 9, at least about a factor of 10, at least about a factor of 20, at least about a factor of 30, at least about a factor of 40, at least about a factor of 50, at least about a factor of 60, at least about a factor of 70, at least about a factor of 80, at least about a factor of 90, at least about a factor of 100, at least about a factor of 200, at least about a factor of 300, at least about a factor of 400, at least about a factor of 500, at least about a factor of 600, at least about a factor of 700, at least about a factor of 800, at least about a factor of 900, or at least about a factor of 1000.


In some embodiments, the composition reduces the time it takes for the signal to saturate by at least about a factor of 1.01, at least about a factor of 1.02, at least about a factor of 1.03, at least about a factor of 1.04, at least about a factor of 1.05, at least about a factor of 1.06, at least about a factor of 1.07, at least about a factor of 1.08, at least about a factor of 1.09, at least about a factor of 1.1, at least about a factor of 1.2, at least about a factor of 1.3, at least about a factor of 1.4, at least about a factor of 1.5, at least about a factor of 1.6, at least about a factor of 1.7, at least about a factor of 1.8, at least about a factor of 1.9, at least about a factor of 2, at least about a factor of 3, at least about a factor of 4, at least about a factor of 5, at least about a factor of 6, at least about a factor of 7, at least about a factor of 8, at least about a factor of 9, at least about a factor of 10, at least about a factor of 20, at least about a factor of 30, at least about a factor of 40, at least about a factor of 50, at least about a factor of 60, at least about a factor of 70, at least about a factor of 80, at least about a factor of 90, at least about a factor of 100, at least about a factor of 200, at least about a factor of 300, at least about a factor of 400, at least about a factor of 500, at least about a factor of 600, at least about a factor of 700, at least about a factor of 800, at least about a factor of 900, or at least about a factor of 1000. In some embodiments, the additive is present at a concentration of at least about 1 nM, at least about 2 nM, at least about 3 nM, at least about 4 nM, at least about 5 nM, at least about 6 nM, at least about 7 nM, at least about 8 nM, at least about 9 nM, at least about 10 nM, at least about 20 nM, at least about 30 nM, at least about 40 nM, at least about 50 nM, at least about 60 nM, at least about 70 nM, at least about 80 nM, at least about 90 nM, at least about 100 nM, at least about 200 nM, at least about 300 nM, at least about 400 nM, at least about 500 nM, at least about 600 nM, at least about 700 nM, at least about 800 nM, at least about 900 nM, at least about 1 μM, at least about 2 μM, at least about 3 μM, at least about 4 μM, at least about 5 μM, at least about 6 μM, at least about 7 μM, at least about 8 μM, at least about 9 UM, at least about 10 μM, at least about 20 μM, at least about 30 μM, at least about 40 μM, at least about 50 μM, at least about 60 M, at least about 70 μM, at least about 80 μM, at least about 90 μM, at least about 100 μM, at least about 200 μM, at least about 300 μM, at least about 400 μM, at least about 500 μM, at least about 600 μM, at least about 700 μM, at least about 800 μM, at least about 900 μM, at least about 1 mM, at least about 2 mM, at least about 3 mM, at least about 4 mM, at least about 5 mM, at least about 6 mM, at least about 7 mM, at least about 8 mM, at least about 9 mM, at least about 10 mM, at least about 20 mM, at least about 30 mM, at least about 40 mM, at least about 50 mM, at least about 60 mM, at least about 70 mM, at least about 80 mM, at least about 90 mM, at least about 100 mM, at least about 200 mM, at least about 300 mM, at least about 400 mM, at least about 500 mM, at least about 600 mM, at least about 700 mM, at least about 800 mM, at least about 900 mM, at least about 1 M, at least about 2 M, at least about 3 M, at least about 4 M, at least about 5 M, at least about 6 M, at least about 7 M, at least about 8 M, at least about 9 M, at least about 10 M.


Assay Compositions with Thermostable Inorganic Pyrophosphatase


Any of the systems, methods, or devices described herein may comprise using thermostable inorganic pyrophosphatase (TIPP). In some embodiments, a composition for an assay may comprise TIPP. In some embodiments, the composition may comprise about 0.5 enzyme unit (U) per 10 μL of solution. In some embodiments, the composition may comprise at least about 0.1 U per 10 μL of solution. In some embodiments, the composition may comprise at most about 2 U per 10 μL of solution. In some embodiments, the composition may comprise at least about 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 U per 10 μL of solution. In some embodiments, the composition may comprise at most about 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 U per 10 μL of solution.


In some embodiments, TIPP may improve the signal to noise ratio of a programmable nuclease-based detection reaction. In some embodiments, TIPP may improve overall signal (e.g., fluorescence of a cleaved reporter as shown in FIG. 84 or intensity of a lateral flow assay strip detection location as shown in FIGS. 85A-85B). TIPP may improve signal by a factor, wherein the signal is indicative of the presence of a target nucleic acid. In some embodiments, the factor may be at least about 1.1. In some embodiments, the factor may be at least about 2. In some embodiments, the factor may be at least about 3. In some embodiments, the factor may be at least about 4. In some embodiments, the factor may be at least about 5. In some embodiments, the factor may be at least about 6. In some embodiments, the factor may be at least about 7. In some embodiments, the factor may be at least about 8. In some embodiments, the factor may be at least about 9. In some embodiments, the factor may be at least about 10.


In some cases, the TIPP enzyme may be present in the HotPot reaction mix at a concentration of 0.125 Units, 0.5 Units, 0.25 Units, 1.0 Units, 2.0 Units, 2.5 Units, or 4 Units per discrete reaction volume. In some cases, the buffer may comprise the TIPP signal enhancement reagent. Conversely, the TIPP signal enhancement reagent may be provided separately (e.g., in a separate vial in a kit) and added to a reaction mixture at a desired timepoint.


In some cases, a buffer may comprise a catalytic reagent for signal improvement or enhancement. In some cases, the catalytic reagent may enhance signal generation via hydrolysis of inorganic pyrophosphates. In some cases, the catalytic reagent may enhance signal generation via enhancement of DNA replication. In some cases, the catalytic reagent may enhance signal amplification via revival of Mg+2 ions in the buffer solution which may otherwise be taken up by the phosphates produced from usage of dNTPs during the LAMP reaction. In some cases, the catalytic reagent may enhance signal generation by reviving the concentration of Mg+2 ions in the buffer thereby enhancing the function of the Cas nuclease effector enzyme. In some cases, the catalytic reagent for signal improvement may be an enzyme. In some cases, the catalytic reagent for signal improvement may be a Thermostable Inorganic Pyrophosphatase (TIPP).


In some embodiments, the one or more enzymes may comprise a nuclease. In some embodiments, the one or more enzymes may comprise a programmable nuclease. In some embodiments, the one or more enzymes may comprise a Cas12 enzyme. In some embodiments, the one or more enzymes may comprise a Cas14 enzyme. In some embodiments, the one or more enzymes may comprise a CasPhi enzyme. In some embodiments, the one or more enzymes may comprise a Cas13 enzyme. In some embodiments, the one or more enzymes may comprise HRP. In some embodiments, the one or more enzymes may comprise any one or combination of enzymes presented in this disclosure.


In some embodiments, the one or more reporters may comprise a reporter free in solution. In some embodiments, the one or more reporters may comprise a reporter immobilized on a substrate. In some embodiments, the one or more reporters may comprise biotin. In some embodiments, the one or more reporters may comprise a fluorescent moiety. In some embodiments, the one or more reporters may comprise a nucleic acid tether. In some embodiments, the one or more reporters may comprise a linker. In some embodiments, the one or more reporters may comprise any one or combination of reporters presented in this disclosure.


In some embodiments, the one or more substrates may comprise a glassy substance. In some embodiments, the one or more substrates may comprise a polymeric substance. In some embodiments, the one or more substrates may comprise a hydrogel. In some embodiments, the one or more substrates may comprise any one or combination of substrates presented in this disclosure.


Nicking Enzyme Amplification Reaction (NEAR)

In some embodiments, a target nucleic acid may be amplified using a nicking enzyme amplification reaction (NEAR). NEAR may be used to amplify a region of a nucleic acid comprising a target nucleic acid. NEAR may comprise a forward primer and a reverse primer that at least partially ° ls to complementary strands of a target nucleic acid 3′ of the region to be amplified. The forward primer and the reverse primer may comprise a stabilizing region that is not complementary to the target sequence. The forward primer and the reverse primer may comprise recognition regions that may be nicked by a nicking enzyme. A polymerase may polymerize a nucleic acid 5′ to 3′ from the 3′ end of the forward primer or the 3′ end of the reverse primer, using the strand to which the primer is annealed as a template, resulting in a double stranded nucleic acid product or amplicon. The newly synthesized strand may comprise a restriction site and may then serve as a template. The resulting double stranded nucleic acid amplicon may comprise nicking sites in both strands. A nicking enzyme may nick a single strand of the double stranded product or amplicon. The polymerase may polymerize a nucleic acid 5′ to 3′ from the 3′ end of the nucleic acid amplicon 5′ of the nick. The process may be repeated, thereby amplifying the target nucleic acid.


As described herein, a target nucleic acid may be detected using a DNA-activated programmable RNA nuclease (e.g., a Cas13), a DNA-activated programmable DNA nuclease (e.g., a Cas12), or an RNA-activated programmable RNA nuclease (e.g., a Cas13) and other reagents disclosed herein (e.g., RNA components). The target nucleic acid may be detected using DETECTR, as described herein. The target nucleic acid may be an RNA, reverse transcribed RNA, DNA, DNA amplicon, amplified DNA, synthetic nucleic acids, or nucleic acids found in biological or environmental samples. In some embodiments, the target nucleic acid is amplified prior to or concurrent with detection. In some embodiments, the target nucleic acid is reverse transcribed prior to amplification. The target nucleic acid may be amplified via NEAR of a target nucleic acid sequence. In some embodiments, the nucleic acid is amplified using NEAR coupled with reverse transcription (RT-NEAR). The NEAR amplification may be performed independently, or the NEAR amplification may be coupled to DETECTR for detection of the target nucleic acid. The RT-NEAR amplification may be performed independently, or the RT-NEAR amplification may be coupled to DETECTR for detection of the target nucleic acid. The DETECTR reaction may be performed using any method consistent with the methods disclosed herein.


NEAR may be performed as an isothermal reaction, for example NEAR may be performed at from about 37° ° C. to about 42° C. In some embodiments, NEAR may be performed at from about 15° C. to about 60° C., from about 15° C. to about 55° C., from about 15° C. to about 50° C., from about 15° C. to about 45° C., from about 15° C. to about 40° C., from about 15° C. to about 35° C., from about 15° C. to about 30° ° C., from about 15° C. to about 25° C., from about 15° C. to about 20° C., from about 20° C. to about 60° C., from about 20° C. to about 55° C., from about 20° ° C. to about 50° C., from about 20° C. to about 45° C., from about 20° C. to about 40° C., from about 20° ° C. to about 35° ° C., from about 20° ° C. to about 30° C., from about 20° C. to about 25° C., from about 25° C. to about 60° C., from about 25° C. to about 55° C., from about 25° C. to about 50° C., from about 25° C. to about 45° C., from about 25° C. to about 40° C., from about 25° C. to about 35° C., from about 25° C. to about 30° C., from about 30° C. to about 60° C., from about 30° ° C. to about 55° C., from about 30° ° C. to about 50° C., from about 30° C. to about 45° C., from about 30° C. to about 40° C., from about 30° C. to about 35° C., from about 35° C. to about 60° C., from about 35° C. to about 55° C., from about 35° C. to about 50° C., from about 35° C. to about 45° C., from about 35° C. to about 40° C., from about 40° C. to about 60° C., from about 40° C. to about 55° C., from about 40° C. to about 50° C., from about 40° C. to about 45° C., from about 45° C. to about 60° C., from about 45° C. to about 55° C., from about 45° C. to about 50° C., from about 50° ° C. to about 60° C., from about 50° C. to about 55° C., or from about 55° C. to about 60° C. In some embodiments, NEAR may be performed above about 15° C., above about 20° C., above about 25° C., above about 30° C., above about 35° C., above about 40° C., above about 45° C., or above about 50° C. In some embodiments, an SDA reaction may be performed below about 60° C., below about 55° C., below about 50° C., below about 45° C., below about 40° C., below about 35° C., below about 30° C., below about 25° C., below about 20° C., or below about 15° C. In some embodiments, NEAR may be performed at about room temperature. In some embodiments, a nucleic acid sample may be heated prior to isothermal amplification. In some embodiments, the nucleic acid sample heated prior to isothermal amplification may comprise one or more primers. The nucleic acid sample may be heated to about 95° C. prior to isothermal amplification. The nucleic acid sample may be heated to a temperature sufficient to dissociate two strands of a double stranded nucleic acid sequence.


NEAR may amplify a target nucleic acid to detectable levels within about 30 seconds, 5 minutes, about 10 minutes, about 15 minutes, about 20 minutes, about 25 minutes, about 30 minutes, about 35 minutes, about 40 minutes, about 45 minutes, about 50 minutes, about 55 minutes, or about 60 minutes. NEAR may amplify a target nucleic acid to detectable levels within about 1 hour, about 1.1 hours, about 1.2 hours, about 1.3 hours, about 1.4 hours, about 1.5 hours, about 1.6 hours, about 1.7 hours, about 1.8 hours, about 1.9 hours, about 2 hours, about 2.2 hours, about 2.4 hours, about 2.5 hours, about 2.6 hours, about 2.8 hours, about 3 hours, about 3.5 hours, about 4 hours, about 4.5 hours, or about 5 hours. NEAR may amplify a target nucleic acid to detectable levels within from about 0.1 hours to about 0.5 hours, from about 0.1 hours to about 1 hour, from about 0.1 hours to about 1.5 hours, from about 0.1 hours to about 2 hours, from about 0.1 hours to about 2.5 hours, from about 0.1 hours to about 3 hours, from about 0.1 hours to about 3.5 hours, from about 0.1 hours to about 4 hours, from about 0.1 hours to about 4.5 hours, from about 0.1 hours to about 5 hours, from about 0.5 hours to about 1 hour, from about 0.5 hours to about 1.5 hours, from about 0.5 hours to about 2 hours, from about 0.5 hours to about 2.5 hours, from about 0.5 hours to about 3 hours, from about 0.5 hours to about 3.5 hours, from about 0.5 hours to about 4 hours, from about 0.5 hours to about 4.5 hours, from about 0.5 hours to about 5 hours, from about 1 hour to about 1.5 hours, from about 1 hour to about 2 hours, from about 1 hour to about 2.5 hours, from about 1 hour to about 3 hours, from about 1 hour to about 3.5 hours, from about 1 hour to about 4 hours, from about 1 hour to about 4.5 hours, from about 1 hour to about 5 hours, from about 1.5 hours to about 2 hours, from about 1.5 hours to about 2.5 hours, from about 1.5 hours to about 3 hours, from about 1.5 hours to about 3.5 hours, from about 1.5 hours to about 4 hours, from about 1.5 hours to about 4.5 hours, from about 1.5 hours to about 5 hours, from about 2 hours to about 2.5 hours, from about 2 hours to about 3 hours, from about 2 hours to about 3.5 hours, from about 2 hours to about 4 hours, from about 2 hours to about 4.5 hours, from about 2 hours to about 5 hours, from about 2.5 hours to about 3 hours, from about 2.5 hours to about 3.5 hours, from about 2.5 hours to about 4 hours, from about 2.5 hours to about 4.5 hours, from about 2.5 hours to about 5 hours, from about 3 hours to about 3.5 hours, from about 3 hours to about 4 hours, from about 3 hours to about 4.5 hours, from about 3 hours to about 5 hours, from about 3.5 hours to about 4 hours, from about 3.5 hours to about 4.5 hours, from about 3.5 hours to about 5 hours, from about 4 hours to about 4.5 hours, from about 4 hours to about 5 hours, or from about 4.5 hours to about 5 hours.


NEAR Amplification and Detection Reaction Mixtures

NEAR reaction components may comprise a polymerase, a nicking enzyme, dNTPs, and one or more nucleic acid primers. In some embodiments, the reaction may further comprise a reverse transcriptase as described herein. In some embodiments, the polymerase may be an exo-Klenow polymerase. The nicking enzyme may be capable of nicking a single strand of a double stranded nucleic acid sequence. In some embodiments, the nicking enzyme may be capable of nicking an unthiolated strand of a double stranded nucleic acid sequence comprising a thiolated strand and an unthiolated strand. In some embodiments, the nicking enzyme may be capable of nicking a single strand comprising an unthiolated region of a double stranded nucleic acid comprising at one or more thiolated regions and one or more unthiolated regions. In some embodiments, the nicking enzyme is a restriction enzyme capable of nicking a single strand of a double stranded nucleic acid sequence. In some embodiments, the nicking enzyme is a modified restriction enzyme. The nicking enzyme may be a strand-limited restriction enzyme. The restriction enzyme may be HincII. In some embodiments, the restriction enzyme may be AluI, BamHI, EcoP15I, EcoRI, EcoRII, EcoRV, HaeIII, HgaI, HindII, HindIII, HinFI, KpnI, NotI, PstI, PvuII, SacI, SalI, Sau3, Scal, SmaI, SpeI, SphI, StuI, TaqI, or XbaI, or the like. The nicking enzyme may be Nt.BspQI, Nt.CvPII, Nt.BstNBI, Nb.BsrDI, Nb.BtsI, Nt.AlwI, Nb.BbvCI, Nb.BsmI, Nb.BssSI, Nt.BsmAI, Nb.Bpu10I, Nt.Bpu110I, Nb.Mva1269I, or I-HmuI, or the like. The one or more nucleic acid primers may comprise two primers. For example, the one or more nucleic acid primers may comprise a first primer (e.g., an S1 primer) and a second primer (e.g., an S2 primer). The target nucleic acid may be single stranded DNA or double stranded DNA. In some embodiments, a target nucleic acid comprising RNA may be reverse transcribed into DNA using a reverse transcriptase prior to NEAR amplification. A reverse transcription reaction may comprise primers, dNTPs, and a reverse transcriptase. In some embodiments, the reverse transcription reaction and the NEAR amplification reaction may be performed in the same reaction. A combined RT-NEAR reaction may comprise NEAR primers, reverse transcription primers, dNTPs, a reverse transcriptase, a polymerase, and dNTPs. In some embodiment, the NEAR primers may comprise the reverse transcription primers.


A DETECTR reaction to detect the target nucleic acid sequence may comprise a guide nucleic acid comprising a segment that is reverse complementary to a segment of the target nucleic acid and a programmable nuclease. The programmable nuclease when activated, as described elsewhere herein, exhibits sequence-independent cleavage of a reporter (e.g., a nucleic acid comprising a moiety that becomes detectable upon cleavage of the nucleic acid by the programmable nuclease). The programmable nuclease is activated upon the guide nucleic acid hybridizing to the target nucleic acid. A combined NEAR DETECTR reaction may comprise a polymerase, a restriction enzyme, dNTPs, one or more nucleic acid primers, a guide nucleic acid, a programmable nuclease, and a substrate nucleic acid. A combined RT-NEAR DETECTR reaction may comprise reverse transcription primers, a reverse transcriptase, a polymerase, a restriction enzyme, dNTPs, one or more nucleic acid primers, a guide nucleic acid, a programmable nuclease, and a substrate nucleic acid. In some embodiment, the primers may comprise the reverse transcription primers. NEAR and DETECTR can be carried out in the same sample volume. NEAR and DETECTR can be carried out concurrently in separate sample volumes or in the same sample volume. RT-NEAR and DETECTR can be carried out in the same sample volume. RT-NEAR and DETECTR can be carried out concurrently in separate sample volumes or in the same sample volume. A NEAR reaction may be multiplexed to amplify a plurality of target nucleic acid sequences in a single reaction.


NEAR Primers and Guide Nucleic Acids

A number of NEAR primers and NEAR primer design methods are consistent with the methods compositions, reagents, enzymes, and kits disclosed herein. NEAR may comprise a set of primers. In some embodiments, NEAR may be an RT-NEAR reaction, a NEAR DETECTR reaction, or an RT-NEAR DETECTR reaction. The set of NEAR primers may comprise two primers, a first primer and a second primer. In some embodiments, a first primer may comprise a sequence of the first region at the 3′ end of the first primer. The sequence of the first region may be from about 16 nucleic acids to about 25 nucleic acids long, or about 20 nucleic acids long. The 3′ end of the first primer may hybridize to the first complementary region of the target. The first complementary region may be 3′ of the target nucleic acid. The first complementary region may be 3′ of a sequence reverse complementary to the target nucleic acid. The first primer may further comprise a cut site 5′ of the sequence of the first region that may be recognized and cleaved by a nicking enzyme. The 3′ end of the first primer may further comprise a recognition site for a nicking enzyme. In some embodiments, the 3′ end of the first primer may further comprise a nicking enzyme stabilization region. The sequence of the second region may be from about 30 nucleic acids to about 38 nucleic acids long. The 3′ end of the second primer may hybridize to the second complementary region. The second complementary region may be 3′ of a sequence reverse complementary to the target nucleic acid. The second complementary region may be 3′ of the target nucleic acid. The second complementary region may be 3′ of a sequence reverse complementary to the target nucleic acid. The second primer may further comprise a cut site 5′ of the sequence of the second region that may be recognized and cleaved by a nicking enzyme. The 3′ end of the second primer may further comprise a recognition site for a nicking enzyme. In some embodiments, the 3′ end of the second primer may further comprise a nicking enzyme stabilization region.


The NEAR primers are designed depending on the site of the optimal guide RNA placement, which may or may not be determined by an available PAM sequence. When performing a NEAR-DETECTR reaction, single-stranded DNA is produced by the designed primers. Because the DETECTR reaction will detection single stranded DNA species, the amplification reaction can be biased to produce more of the particular strand than another. This can be done through changing of the ratio of the forward and reverse primer concentrations. In some embodiments, the concentration of forward primer can be 5 times, 4 times, 3 times, 2 times, or equal to the concentration of reverse primer. In some embodiments, the concentration of reverse primer can be 5 times, 4 times, 3 times, 2 times, or equal to the concentration of forward primer.


In some embodiments, the first region, the second region, or both may be about 8 nucleic acids, about 10 nucleic acids, about 12 nucleic acids, about 14 nucleic acids, about 16 nucleic acids, about 18 nucleic acids, about 20 nucleic acids, about 22 nucleic acids, about 24 nucleic acids, about 26 nucleic acids, about 28 nucleic acids, about 30 nucleic acids, about 32 nucleic acids, about 34 nucleic acids, about 36 nucleic acids, about 38 nucleic acids, about 40 nucleic acids, about 42 nucleic acids, about 44 nucleic acids, about 46 nucleic acids, about 48 nucleic acids, or about 50 nucleic acids long.


In some embodiments, the first region, the second region, or both may be from about 8 to about 12, from about 8 to about 16, from about 8 to about 20, from about 8 to about 24, from about 8 to about 28, from about 8 to about 30, from about 8 to about 32, from about 8 to about 34, from about 8 to about 36, from about 8 to about 38, from about 8 to about 40, from about 8 to about 42, from about 8 to about 44, from about 8 to about 48, from about 8 to about 50, from about 12 to about 16, from about 12 to about 20, from about 12 to about 24, from about 12 to about 28, from about 12 to about 30, from about 12 to about 32, from about 12 to about 34, from about 12 to about 36, from about 12 to about 38, from about 12 to about 40, from about 12 to about 42, from about 12 to about 44, from about 12 to about 48, from about 12 to about 50, from about 16 to about 20, from about 16 to about 24, from about 16 to about 28, from about 16 to about 30, from about 16 to about 32, from about 16 to about 34, from about 16 to about 36, from about 16 to about 38, from about 16 to about 40, from about 16 to about 42, from about 16 to about 44, from about 16 to about 48, from about 16 to about 50, from about 20 to about 24, from about 20 to about 28, from about 20 to about 30, from about 20 to about 32, from about 20 to about 34, from about 20 to about 36, from about 20 to about 38, from about 20 to about 40, from about 20 to about 42, from about 20 to about 44, from about 20 to about 48, from about 20 to about 50, from about 24 to about 28, from about 24 to about 30, from about 24 to about 32, from about 24 to about 34, from about 24 to about 36, from about 24 to about 38, from about 24 to about 40, from about 24 to about 42, from about 24 to about 44, from about 24 to about 48, from about 24 to about 50, from about 28 to about 30, from about 28 to about 32, from about 28 to about 34, from about 28 to about 36, from about 28 to about 38, from about 28 to about 40, from about 28 to about 42, from about 28 to about 44, from about 28 to about 48, from about 28 to about 50, from about 30 to about 32, from about 30 to about 34, from about 30 to about 36, from about 30 to about 38, from about 30 to about 40, from about 30 to about 42, from about 30 to about 44, from about 30 to about 48, from about 30 to about 50, from about 32 to about 34, from about 32 to about 36, from about 32 to about 38, from about 32 to about 40, from about 32 to about 42, from about 32 to about 44, from about 32 to about 48, from about 32 to about 50, from about 34 to about 36, from about 34 to about 38, from about 34 to about 40, from about 34 to about 42, from about 34 to about 44, from about 34 to about 48, from about 34 to about 50, from about 36 to about 38, from about 36 to about 40, from about 36 to about 42, from about 36 to about 44, from about 36 to about 48, from about 36 to about 50, from about 38 to about 40, from about 38 to about 42, from about 38 to about 44, from about 38 to about 48, from about 38 to about 50, from about 40 to about 42, from about 40 to about 44, from about 40 to about 48, from about 40 to about 50, from about 42 to about 44, from about 42 to about 48, from about 42 to about 50, from about 44 to about 48, from about 44 to about 50, from about 48 to about 50 nucleic acids long.


In some embodiments, the first region, the second region, or both may comprise a GC content of about 1%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, or about 99%. In some embodiments, the first region, the second region, or both may comprise a GC content of from about 1% to about 5%, from about 1% to about 10%, from about 1% to about 15%, from about 1% to about 20%, from about 1% to about 25%, from about 1% to about 30%, from about 1% to about 40%, from about 1% to about 50%, from about 1% to about 60%, from about 1% to about 70%, from about 1% to about 80%, from about 1% to about 90%, from about 1% to about 95%, from about 1% to about 99%, from about 5% to about 10%, from about 5% to about 15%, from about 5% to about 20%, from about 5% to about 25%, from about 5% to about 30%, from about 5% to about 40%, from about 5% to about 50%, from about 5% to about 60%, from about 5% to about 70%, from about 5% to about 80%, from about 5% to about 90%, from about 5% to about 95%, from about 5% to about 99%, from about 10% to about 15%, from about 10% to about 20%, from about 10% to about 25%, from about 10% to about 30%, from about 10% to about 40%, from about 10% to about 50%, from about 10% to about 60%, from about 10% to about 70%, from about 10% to about 80%, from about 10% to about 90%, from about 10% to about 95%, from about 10% to about 99%, from about 15% to about 20%, from about 15% to about 25%, from about 15% to about 30%, from about 15% to about 40%, from about 15% to about 50%, from about 15% to about 60%, from about 15% to about 70%, from about 15% to about 80%, from about 15% to about 90%, from about 15% to about 95%, from about 15% to about 99%, from about 20% to about 25%, from about 20% to about 30%, from about 20% to about 40%, from about 20% to about 50%, from about 20% to about 60%, from about 20% to about 70%, from about 20% to about 80%, from about 20% to about 90%, from about 20% to about 95%, from about 20% to about 99%, from about 25% to about 30%, from about 25% to about 40%, from about 25% to about 50%, from about 25% to about 60%, from about 25% to about 70%, from about 25% to about 80%, from about 25% to about 90%, from about 25% to about 95%, from about 25% to about 99%, from about 30% to about 40%, from about 30% to about 50%, from about 30% to about 60%, from about 30% to about 70%, from about 30% to about 80%, from about 30% to about 90%, from about 30% to about 95%, from about 30% to about 99%, from about 40% to about 50%, from about 40% to about 60%, from about 40% to about 70%, from about 40% to about 80%, from about 40% to about 90%, from about 40% to about 95%, from about 40% to about 99%, from about 50% to about 60%, from about 50% to about 70%, from about 50% to about 80%, from about 50% to about 90%, from about 50% to about 95%, from about 50% to about 99%, from about 60% to about 70%, from about 60% to about 80%, from about 60% to about 90%, from about 60% to about 95%, from about 60% to about 99%, from about 70% to about 80%, from about 70% to about 90%, from about 70% to about 95%, from about 70% to about 99%, from about 80% to about 90%, from about 80% to about 95%, from about 80% to about 99%, from about 90% to about 95%, from about 90% to about 99%, or from about 95% to about 99%.


In some embodiments, the first region, the second region, or both may have a melting temperature of about 38° C., about 40° C., about 42° ° C., about 44° C., about 46° C., about 48° C., about 50° C., about 52° C., about 54° C., about 56° C., about 58° C., about 60° C., about 62° C., about 64° C., about 66° C., about 68° C., about 70° C., about 72° C., about 74° C., about 76° C., about 78° C., about 80° C., about 82° C., about 84° C., about 86° C., about 88° C., about 90° C., or about 92° ° C. In some embodiments, the first region, the second region, or both may have a melting temperature of from about 35° C. to about 40° C., from about 35° C. to about 45° C., from about 35° C. to about 50° C., from about 35° C. to about 55° C., from about 35° C. to about 60° C., from about 35° C. to about 65° C., from about 35° C. to about 70° C., from about 35° C. to about 75° C., from about 35° C. to about 80° C., from about 35° C. to about 85° C., from about 35° C. to about 90° C., from about 35° C. to about 95° C., from about 40° C. to about 45° C., from about 40° C. to about 50° C., from about 40° C. to about 55° C., from about 40° C. to about 60° ° C., from about 40° C. to about 65° C., from about 40° C. to about 70° C., from about 40° C. to about 75° C., from about 40° ° C. to about 80° C., from about 40° C. to about 85° C., from about 40° C. to about 90° C., from about 40° ° C. to about 95° C., from about 45° C. to about 50° C., from about 45° C. to about 55° ° C., from about 45° ° C. to about 60° C., from about 45° C. to about 65° C., from about 45° C. to about 70° C., from about 45° C. to about 75° C., from about 45° C. to about 80° C., from about 45° C. to about 85° C., from about 45° C. to about 90° C., from about 45° C. to about 95° C., from about 50° C. to about 55° C., from about 50° ° C. to about 60° C., from about 50° C. to about 65° C., from about 50° ° C. to about 70° C., from about 50° C. to about 75° C., from about 50° C. to about 80° C., from about 50° ° C. to about 85° C., from about 50° C. to about 90° C., from about 50° C. to about 95° C., from about 55° C. to about 60° C., from about 55° C. to about 65° C., from about 55° C. to about 70° C., from about 55° ° C. to about 75° C., from about 55° C. to about 80° C., from about 55° C. to about 85° C., from about 55° C. to about 90° C., from about 55° C. to about 95° C., from about 60° C. to about 65° C., from about 60° C. to about 70° C., from about 60° C. to about 75° C., from about 60° C. to about 80° C., from about 60° C. to about 85° C., from about 60° C. to about 90° C., from about 60° C. to about 95° C., from about 65° C. to about 70° C., from about 65° ° C. to about 75° C., from about 65° C. to about 80° C., from about 65° C. to about 85° C., from about 65° C. to about 90° C., from about 65° C. to about 95° C., from about 70° C. to about 75° C., from about 70° ° C. to about 80° ° C., from about 70° C. to about 85° C., from about 70° C. to about 90° C., from about 70° C. to about 95° C., from about 75° C. to about 80° C., from about 75° C. to about 85° C., from about 75° C. to about 90° C., from about 75° C. to about 95° C., from about 80° C. to about 85° C., from about 80° C. to about 90° C., from about 80° C. to about 95° C., from about 85° C. to about 90° C., from about 85° C. to about 95° C., or from about 90° C. to about 95° C.


A set of NEAR primers may be designed for use in combination with a DETECTR reaction. Exemplary NEAR primer sequences are provided in Table 19 below. The amplified nucleic acid sequence may comprise a sequence that hybridizes to a guide RNA. The amplified nucleic acid sequence may comprise a target nucleic acid. The guide RNA may hybridize to the target nucleic acid. The amplified nucleic acid sequence may comprise corresponding to a guide RNA. The amplified nucleic acid sequence may comprise a sequence reverse complementary to the target nucleic acid. All or part of the guide RNA may be reverse complementary to all or part of the target nucleic acid. The amplified nucleic acid sequence may comprise a protospacer adjacent motif (PAM) positioned next to or near the target sequence. The PAM may be 3′ of the target nucleic acid. In some embodiments, a portion of a sequence that hybridizes the guide RNA may be located between the first region and the second complementary region. The portion of a sequence that hybridizes the guide RNA located between the first region and the second complementary region may comprise at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or more of the sequence that hybridizes guide RNA. In some embodiments, the 5′ end of the sequence that hybridizes the guide RNA is 3′ of the 3′ end of the first region and 3′ end of the sequence that hybridizes the guide RNA is 5′ of the 5′ end of the second complementary region. In some embodiments, a portion of a sequence that hybridizes the guide RNA may be located between the second region and the first complementary region. The portion of a sequence that hybridizes the guide RNA located between the second region and the first complementary region may comprise at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or more of the sequence that hybridizes the guide RNA. In some embodiments, the 5′ end of the sequence that hybridizes the guide RNA is 3′ of the 3′ end of the second region and 3′ end of the sequence that hybridizes the guide RNA is 5′ of the 5′ end of the first complementary region.


In some embodiments, a sequence that hybridizes the guide RNA may overlap the first region, the first complementary region, the second region, or the second complementary region by no more than no more than about 5%, no more than about 10%, no more than about 15%, no more than about 20%, no more than about 25%, no more than about 30%, no more than about 35%, no more than about 40%, no more than about 45%, no more than about 50%, no more than about 55%, no more than about 60%, no more than about 65%, no more than about 70%, no more than about 75%, no more than about 80%, no more than about 85%, no more than about 90%, or no more than about 95%. In some embodiments, the sequence that hybridizes the guide RNA does not overlap the first region, the first complementary region, the second region, or the second complementary region. In some embodiments, the guide RNA does not hybridize to the first primer or the second primer.


In some embodiments, a NEAR primer set may be designed using a commercially available primer design software. A NEAR primer set may be designed for use in combination with a DETECR reaction, a reverse transcription reaction, or both. One or more methods of designing a set of NEAR primers may be readily apparent to one skilled in the art and may be employed in any of the compositions, kits and methods described herein.









TABLE 19







Exemplary NEAR Primers











PRIMER
ALTERNATIVE



SEQ ID NO:
NAME
NAME
SEQUENCE





SEQ ID NO: 377
M2805
CoV2-M1
TTGCTTTCGTGGTA





TT





SEQ ID NO: 378
M2811
CoV2-M2
GGATGGCTAGTGT





AA





SEQ ID NO: 379
R1763 F8
CoV2-F8
TTTGCCCCCAGCG





CTT





SEQ ID NO: 380
R1763 R8
CoV2-R8
ATGCGCGACATTC





CGA





SEQ ID NO: 381
R1763 F7
CoV2-F7
ATTTGCCCCCAGC





GCT





SEQ ID NO: 382
R1763 R7
CoV2-R7
TGCGCGACATTCC





GAA





SEQ ID NO: 383
R1763 F6
CoV2-F6
AATTTGCCCCCAG





CGC





SEQ ID NO: 384
R1763 R6
CoV2-R6
GCGCGACATTCCG





AAG





SEQ ID NO: 385
R1763 F5
CoV2-F5
CAATTTGCCCCCA





GCG





SEQ ID NO: 386
R1763 R5
CoV2-R5
CGCGACATTCCGA





AGA





SEQ ID NO: 387
R1763 F4
CoV2-F4
ACAATTTGCCCCC





AGC





SEQ ID NO: 388
R1763 R4
CoV2-R4
GCGACATTCCGAA





GAA





SEQ ID NO: 389
R1763 F3
CoV2-F3
CACAATTTGCCCC





CAG





SEQ ID NO: 390
R1763 R3
CoV2-R3
CGACATTCCGAAG





AAC





SEQ ID NO: 391
R1763 F2
CoV2-F2
TCACAATTTGCCC





CCA





SEQ ID NO: 392
R1763 R2
CoV2-R2
GACATTCCGAAGA





ACG





SEQ ID NO: 393
R1763 F1
CoV2-F1
TTCACAATTTGCC





CCC





SEQ ID NO: 394
R1763 R1
CoV2-R1
ACATTCCGAAGAA





CGC





SEQ ID NO: 395
EXAMPLE
EXAMPLE
TAGCCAAACAGGA



PRIMER 1F
PRIMER 1F
ACA





SEQ ID NO: 396
EXAMPLE
EXAMPLE
GCCAGCAATCTGA



PRIMER 2R
PRIMER 2R
GGA









Amplification and Detection of a Gene of Interest

A DETECTR reaction may be used to detect the presence of a specific target gene in the same. The DETECTR reaction may produce a detectable signal, as described elsewhere herein, in the presence of a target nucleic acid sequence comprising a target gene. The DETECTR reaction may not produce a signal in the absence of the target nucleic acid or in the presence of a nucleic acid sequence that does not comprise the specific SNP allele or comprises a different SNP allele. In some embodiments, a DETECTR reaction may comprise a guide RNA reverse complementary to a portion of a target nucleic acid sequence comprising a specific SNP allele. The guide RNA and the target nucleic acid comprising the specific SNP allele may bind to and activate a programmable nuclease, thereby producing a detectable signal as described elsewhere herein. The guide RNA and a nucleic acid sequence that does not comprise the specific SNP allele may not bind to or activate the programmable nuclease and may not produce a detectable signal. In some embodiments, a target nucleic acid sequence that may or may not comprise a specific SNP allele may be amplified using, for example, a LAMP amplification reaction, an RPA amplification reaction, an SDA amplification reaction, a NEAR amplification reaction, or any other amplification method. In some embodiments, the amplification reaction may be combined with a reverse transcription reaction, a DETECTR reaction, or both. For example, the amplification reaction may be an RT-NEAR reaction, a NEAR DETECTR reaction, or an RT-NEAR DETECTR reaction. In some embodiments, the target nucleic acid sequence can comprise a SNP. In some embodiments, the target nucleic acid sequence can comprise a sequence indicative of a human disease.


A DETECTR reaction, as described elsewhere herein, may produce a detectable signal specifically in the presence of a target nucleic acid sequence comprising a target gene. In addition to the DETECTR reaction, the target nucleic acid having the target gene may be concurrently, sequentially, concurrently together in a sample, or sequentially together in a sample be carried out alongside NEAR or RT-NEAR. For example, the reactions can comprise NEAR and DETECTR reactions, or RT-NEAR and DETECTR reactions. Performing a DETECTR reaction in combination with a NEAR reaction may result in an increased detectable signal as compared to the DETECTR reaction in the absence of the NEAR reaction. In some embodiments, the target nucleic acid sequence can comprise a SNP. In some embodiments, the target nucleic acid sequence can comprise a sequence indicative of a human disease.


In some embodiments, the detectable signal produced in the DETECTR reaction may be higher in the presence of a target nucleic acid comprising target nucleic acid than in the presence of a nucleic acid that does not comprise the target nucleic acid. In some embodiments, the DETECTR reaction may produce a detectable signal that is at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 10-fold, at least 20-fold, at least 30-fold, at least 40-fold, at least 50-fold, at least 100-fold, at least 200-fold, at least 300-fold, at last 400-fold, at least 500-fold, at least 1000-fold, at least 2000-fold, at least 3000-fold, at least 4000-fold, at least 5000-fold, at least 6000-fold, at least 7000-fold, at least 8000-fold, at least 9000-fold, at least 10000-fold, at least 50000-fold, at least 100000-fold, at least 500000-fold, or at least 1000000-fold greater in the presence of a target nucleic acid comprising a target nucleic acid than in the presence of a nucleic acid that does not comprise the target nucleic acid. In some embodiments, the DETECTR reaction may produce a detectable signal that is from 1-fold to 2-fold, from 2-fold to 3-fold, from 3-fold to 4-fold, from 4-fold to 5-fold, from 5-fold to 10-fold, from 10-fold to 20-fold, from 20-fold to 30-fold, from 30-fold to 40-fold, from 40-fold to 50-fold, from 50-fold to 100-fold, from 100-fold to 500-fold, from 500-fold to 1000-fold, from 1000-fold to 10,000-fold, from 10,000-fold to 100,000-fold, or from 100,000-fold to 1,000,000-fold greater in the presence of a target nucleic acid comprising a specific SNP allele than in the presence of a nucleic acid that does not comprise the specific SNP allele. In some embodiments, the target nucleic acid sequence can comprise a SNP. In some embodiments, the target nucleic acid sequence can comprise a sequence indicative of a human disease.


A DETECTR reaction may be used to detect the presence of a target nucleic acid associated with a disease or a condition in a nucleic acid sample. The DETECTR reaction may reach signal saturation within about 30 seconds, about 5 minutes, about 10 minutes, about 15 minutes, about 20 minutes, about 25 minutes, about 30 minutes, about 35 minutes, about 40 minutes, about 45 minutes, about 50 minutes, about 55 minutes, about 60 minutes, about 65 minutes, about 75 minutes, about 80 minutes, or about 85 minutes and be used to detect the presence of a target gene associated with an increased likelihood of developing a disease or a condition in a nucleic acid sample. The DETECTR reaction may be used to detect the presence of a target gene associated with a phenotype in a nucleic acid sample. For example, a DETECTR reaction may be used to detect target gene associated with a disease such as phenylketonuria (PKU), cystic fibrosis, sickle-cell anemia, albinism, Huntington's disease, myotonic dystrophy type 1, hypercholesterolemia, neurofibromatosis, polycystic kidney disease, hemophilia, muscular dystrophy, hypophosphatemic rickets, Rett's syndrome, or spermatogenic failure. A DETECTR reaction may be used to detect a SNP allele associated with an increased risk of cancer, for example bladder cancer, brain cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, gallbladder cancer, stomach cancer, leukemia, liver cancer, lung cancer, oral cancer, esophageal cancer, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, testicular cancer, thyroid cancer, neuroblastoma, or lymphoma. A DETECTR reaction may be used to detect a SNP allele associated with an increased risk of a disease, for example Alzheimer's disease, Parkinson's disease, amyloidosis, heterochromatosis, celiac disease, macular degeneration, or hypercholesterolemia. A DETECTR reaction may be used to detect a SNP allele associated with a phenotype, for example, eye color, hair color, height, skin color, race, alcohol flush reaction, caffeine consumption, deep sleep, genetic weight, lactose intolerance, muscle composition, saturated fat and weight, or sleep movement. A DETECTR reaction may also be used to detect the presence of a pathological organism. In some embodiments, the pathological organism is a prokaryote, eukaryote, or a protozoa. In some embodiments, the pathological organism is a virus, an opportunistic pathogen, a parasite, a bacterium, or any combination thereof. In some embodiments, the pathological organism is SARS-COV-2 or Streptococcus pyogenes.


Detection Devices

In another aspect, the present disclosure provides a detection device or system for target detection. The detection device can be configured for multiplexed target detection. The detection device can be used to collect one or more samples, prepare or process the one or more samples for detection, and optionally divide the one or more samples into a plurality of droplets, aliquots, or subsamples for amplification of one or more target sequences or target nucleic acids. The target sequences may comprise, for example, a biological sequence. The biological sequence can comprise a nucleic acid sequence or an amino acid sequence. In some embodiments, the target sequences are associated with an organism of interest, a disease of interest, a disease state of interest, a phenotype of interest, a genotype of interest, or a gene of interest.


The detection device can be configured to amplify target nucleic acids contained within the plurality of droplets, aliquots, or subsamples. The detection device can be configured to amplify the target sequences or target nucleic acids contained within the plurality of droplets by individually processing each of the plurality of droplets (e.g., by using a thermocycling process or any other suitable amplification process as described in greater detail below). In some cases, the plurality of droplets can undergo separate thermocycling processes. In some cases, the thermocycling processes can occur simultaneously. In other cases, the thermocycling processes can occur at different times for each droplet.


The detection device can be further configured to remix the droplets, aliquots, or subsamples after the target nucleic acids in each of the droplets undergo amplification. The detection device can be configured to provide the remixed sample comprising the droplets, aliquots, or subsamples to a detection chamber of the device. The detection chamber can be configured to direct the remixed droplets, aliquots, or subsamples to a plurality of programmable nuclease complexes (i.e., programmable nuclease in complex with a guide nucleic acid). The detection chamber can be configured to direct the remixed droplets, aliquots, or subsamples along one or more fluid flow paths such that the remixed droplets, aliquots, or subsamples are positioned adjacent to and/or in contact with the one or more programmable nuclease complexes (i.e., programmable nuclease in complex with a guide nucleic acid). In some cases, the detection chamber can be configured to recirculate or recycle the remixed droplets, aliquots, or subsamples such that the remixed droplets, aliquots, or subsamples are repeatedly placed in contact with one or more programmable nuclease complexes (i.e., programmable nuclease in complex with a guide nucleic acid) over a predetermined period of time.


The detection device can comprise one or more sensors. The one or more sensors of the detection device can be configured to detect one or more signals that are generated after one or more programmable nucleases of the one or more programmable nuclease complexes (i.e., programmable nuclease in complex with a guide nucleic acid) become activated due to a binding of a guide nucleic acid of the programmable nuclease complexes (i.e., programmable nuclease in complex with a guide nucleic acid) with a target nucleic acid present in the sample. As described elsewhere herein, the activated programmable nuclease can cleave the target nucleic acid, which can result in a trans cleavage activity. Trans cleavage activity can be a non-specific cleavage of nearby single-stranded nucleic acids by the activated programmable nuclease, such as trans cleavage of target nucleic acids with a detection moiety. Once the target nucleic acids are cleaved by the activated programmable nucleases, the detection moiety can be released or separated from the reporter, thereby generating one or more detectable signals. The one or more sensors of the detection device can be configured to register and/or process the one or more detectable signals to confirm a presence and/or an absence of a particular target (e.g., a target nucleic acid).


A number of detection devices, systems, and methods are consistent with methods disclosed herein. In some embodiments, a detection device may be used to identify and/or quantify a detectable signal produced by a DETECTR reaction in which amplification and detection are performed in a single buffer. A detection device may procure a signal from a container (e.g., a cuvette, a well, a tube, a centricon, a fluidic channel, a fluidic chamber, or the like) or plurality of containers. For example, a detection device may be capable of detecting signals (e.g., fluorescence signals) from a plurality of wells in a multi well plate. A container may be sealed during detection. For example, a container may comprise a septum or film that the sample can be inserted (e.g., syringed) through, and the container may be placed next to or inside of the detection device after insertion of the sample, without removal of the seal. A device may be configured to simultaneously monitor multiple containers, such as multiple wells within a well plate. A device may perform real time signal monitoring.


A device and/or instrument/system thermally-coupled thereto may be configured to heat a sample (e.g., a sample container). A device may be configured to enact a programmed heating sequence (e.g., holding a sample at a first temperature for a first length of time and then holding the sample at a second temperature for a second length of time). For a sample comprising amplification and programmable nuclease-based detection (e.g., DETECTR) reagents, the programmed heating sequence may first hold the sample temperature or thermocycle the sample at an optimum temperature(s) for amplification and then transition the sample to an optimum temperature for detection (e.g., between 60 and 65° C. for an amplification reaction and between 30 and 40° ° C. for a DETECTR reaction). In another example, the programming sequence may hold the sample mixture comprising amplification and programmable nuclease-based detection (e.g., DETECTR) reagents at a temperature selected to enable simultaneous amplification and detection (e.g., between 50 to 65° C. for a HotPot reaction). A device may perform simultaneous heating and signal detection. For example, a device may comprise a heating block and a fluorescence imaging system.


A device can measure or detect a calorimetric, potentiometric, amperometric, optical (e.g., fluorescent, colorimetric, etc.), or piezo-electric signal. Often a calorimetric signal is heat produced after cleavage of the reporters. Sometimes, a calorimetric signal is heat absorbed after cleavage of the reporters. A potentiometric signal, for example, is electrical potential produced after cleavage of the reporters. An amperometric signal can be movement of electrons produced after the cleavage of a reporter. Often, the signal is an optical signal, such as a colorimetric signal or a fluorescence signal. An optical signal is, for example, a light output produced after the cleavage of the reporters. Sometimes, an optical signal is a change in light absorbance between before and after the cleavage of reporters. Often, a piezo-electric signal is a change in mass between before and after the cleavage of the reporter. Sometimes, the reporter is protein-nucleic acid. Often, the protein-nucleic acid is an enzyme-nucleic acid.


The results from the detection region (e.g., from a container) from a completed assay can be detected and analyzed in various ways, for example, by a glucometer. In some cases, the positive control spot and the detection spot in the detection region is visible by eye, and the results can be read by the user. In some cases, the positive control spot and the detection spot in the detection region is visualized by an imaging device or other device depending on the type of signal. Often, the imaging device is a digital camera, such a digital camera on a mobile device. The mobile device may have a software program or a mobile application that can capture an image of the support medium, identify the assay being performed, detect the detection region and the detection spot, provide image properties of the detection spot, analyze the image properties of the detection spot, and provide a result. Alternatively, or in combination, the imaging device can capture fluorescence, ultraviolet (UV), infrared (IR), or visible wavelength signals. The imaging device may have an excitation source to provide the excitation energy and captures the emitted signals. In some cases, the excitation source can be a camera flash and optionally a filter. In some cases, the imaging device is used together with an imaging box that is placed over the support medium to create a dark room to improve imaging. The imaging box can be a cardboard box that the imaging device can fit into before imaging. In some instances, the imaging box has optical lenses, mirrors, filters, or other optical elements to aid in generating a more focused excitation signal or to capture a more focused emission signal. Often, the imaging box and the imaging device are small, handheld, and portable to facilitate the transport and use of the assay in remote or low resource settings.


The assay described herein can be visualized and analyzed by a mobile application (app) or a software program. Using the graphic user interface (GUI) of the app or program, an individual can take an image of the support medium, including the detection region, barcode, reference color scale, and fiduciary markers on the housing, using a camera on a mobile device. The program or app reads the barcode or identifiable label for the test type, locate the fiduciary marker to orient the sample, and read the detectable signals, compare against the reference color grid, and determine the presence or absence of the target nucleic acid, which indicates the presence of the gene, virus, or the agent responsible for the disease. The mobile application can present the results of the test to the individual. The mobile application can store the test results in the mobile application. The mobile application can communicate with a remote device and transfer the data of the test results. The test results can be viewable remotely from the remote device by another individual, including a healthcare professional. A remote user can access the results and use the information to recommend action for treatment, intervention, cleanup of an environment.


Methods of Making Polymer Matrices with Immobilized Reporters



FIG. 78 shows an exemplary polymer immobilization matrix (14901) comprising a plurality of immobilized DETECTR reaction components. The DETECTR reaction components may comprise one or more reporters, one or more programmable nucleases, and/or one or more guide nucleic acids. In some embodiments, the polymer matrix may comprise a hydrogel. In the exemplary embodiment shown in FIG. 78, a plurality of reporters (14902) may be immobilized within a hydrogel (14901) matrix. In some embodiments, methods of immobilizing a reporter (14902) and/or other DETECTR reaction component may comprise (a) providing a polymerizable composition comprising: (i) a plurality of oligomers, (ii) a plurality of polymerizable (e.g., functionalized) oligomers, (iii) a set of polymerizable (e.g., functionalized) reporters (and/or other DETECTR reaction components), and (iv) a set of polymerization initiators; and (b) initiating the polymerization reaction by providing an initiation stimulus.


Co-polymerization of the reporter into the hydrogel may result in a higher density of reporter/unit volume or reporter/unit area than other immobilization methods utilizing surface immobilization (e.g., onto beads). Co-polymerization of the reporter into the hydrogel may result in less undesired release of the reporter (e.g., during an assay, a measurement, or on the shelf), and thus may cause less background signal, than other immobilization strategies (e.g., conjugation to a pre-formed hydrogel, bead, etc.). In at least some instances this may be due to better incorporation of reporters into the hydrogel as a co-polymer and fewer “free” reporter molecules retained on the hydrogel via non-covalent interactions or non-specific binding interactions.


In some embodiments, the plurality of oligomers and the plurality of polymerizable oligomers may comprise an irregular or non-uniform mixture. The irregularity of the mixture of polymerizable oligomers and unfunctionalized oligomers may allow pores to form within the hydrogel (i.e., the unfunctionalized oligomers may act as a porogen). For example, the irregular mixture of oligomers may result in phase separation during polymerization that allows for the generation of pores of sufficient size for programmable nucleases to diffuse into the hydrogel and access internal reporter molecules. The relative percentages and/or molecular weights of the oligomers may be varied to vary the pore size of the hydrogel. For example, pore size may be tailored to increase the diffusion coefficient of the programmable nucleases.


In some embodiments, the functional groups attached to the reporters may be selected to preferentially incorporate the reporters into the hydrogel matrix via covalent binding at the functional group versus other locations along the nucleic acid of the reporter. In some embodiments, the functional groups attached to the reporters may be selected to favorably transfer free radicals from the functionalized ends of polymerizable oligomers to the functional group on the end of the reporter (e.g., 5′ end), thereby forming a covalent bond and immobilizing the reporter rather than destroying other parts of the reporter molecules.


In some embodiments, the polymerizable composition may further comprise one or more polymerizable nucleic acids. In some embodiments, the polymerizable nucleic acids may comprise guide nucleic acids (e.g., guide nucleic acids 15003a, 15003b, or 15003c shown in FIGS. 79A-79B). In some embodiments, the polymerizable nucleic acids may comprise linker or tether nucleic acids. In some embodiments, the polymerizable nucleic acids may be configured to bind to a programmable nuclease (e.g., programmable nuclease 15004a, 15004b, or 15004c shown in FIGS. 79A-79B). In some embodiments, the programmable nuclease may be immobilized in the polymer matrix.


In some embodiments, the oligomers may form a polymer matrix comprising a hydrogel. In some embodiments, the oligomers may comprise poly(ethylene glycol) (PEG), poly(siloxane), poly(hydroxyethyl acrylate, poly(acrylic acid), poly(vinyl alcohol), poly(butyl acrylate), poly(2-ethylhexyl acrylate), poly(methyl acrylate), poly(ethyl acrylate), poly(acrylonitrile), poly(methyl methacrylate), poly(acrylamide), poly(TMPTA methacrylate), chitosan, alginate, or the like, or any combination thereof. One of ordinary skill in the art will recognize that the oligomers may comprise any oligomer or mix of oligomers capable of forming a hydrogel.


In some embodiments, the oligomers may comprise polar monomers, nonpolar monomers, protic monomers, aprotic monomers, solvophobic monomers, or solvophillic monomers, or any combination thereof.


In some embodiments, the oligomers may comprise a linear topology, branched topology, star topology, dendritic topology, hyperbranched topology, bottlebrush topology, ring topology, catenated topology, or any combination thereof. In some embodiments, the oligomers may comprise 3-armed topology, 4-armed topology, 5-armed topology, 6-armed topology, 7-armed topology, 8-armed topology, 9-armed topology, or 10-armed topology.


In some embodiments, the oligomers may comprise at least about 2 monomers, at least about 3 monomers, at least about 4 monomers, at least about 5 monomers, at least about 6 monomers, at least about 7 monomers, at least about 8 monomers, at least about 9 monomers, at least about 10 monomers, at least about 20 monomers, at least about 30 monomers, at least about 40 monomers, at least about 50 monomers, at least about 60 monomers, at least about 70 monomers, at least about 80 monomers, at least about 90 monomers, at least about 100 monomers, at least about 200 monomers, at least about 300 monomers, at least about 400 monomers, at least about 500 monomers, at least about 600 monomers, at least about 700 monomers, at least about 800 monomers, at least about 900 monomers, at least about 1000 monomers, at least about 2000 monomers, at least about 3000 monomers, at least about 4000 monomers, at least about 5000 monomers, at least about 6000 monomers, at least about 7000 monomers, at least about 8000 monomers, at least about 9000 monomers, at least about 10000 monomers, at least about 20000 monomers, at least about 30000 monomers, at least about 40000 monomers, at least about 50000 monomers, at least about 60000 monomers, at least about 70000 monomers, at least about 80000 monomers, at least about 90000 monomers, or at least about 100000 monomers.


In some embodiments, the oligomers may comprise a homopolymer, a copolymer, a random copolymer, a block copolymer, an alternative copolymer, a copolymer with regular repeating units, or any combination thereof.


In some embodiments, the oligomers may comprise 1 type of monomer, 2 types of monomers, 3 types of monomers, 4 types of monomers, 5 types of monomers, 6 types of monomers, 7 types of monomers, 8 types of monomers, 9 types of monomers, or 10 types of monomers.


The polymerizable oligomers may comprise any of the oligomers described herein. In some embodiments, the polymerizable oligomers may comprise one or more functional groups. In some embodiments, the functional group may comprise an acrylate group, N-hydroxysuccinimide ester group, thiol group, carboxyl group, azide group, alkyne group, an alkene group, or any combination thereof. One of ordinary skill in the art will recognize that a variety of functional groups may be used to functionalize oligomers into polymerizable oligomers depending on the desired properties of the polymerizable oligomers.


In some embodiments, the polymerizable oligomers may form a polymer matrix comprising a hydrogel. In some embodiments, the polymerizable oligomers may comprise PEG, poly(siloxane), poly(hydroxyethyl acrylate, poly(acrylic acid), poly(vinyl alcohol), or any combination thereof. One of ordinary skill in the art will recognize that the set of polymerizable oligomers may comprise any polymer capable of forming a hydrogel.


In some embodiments, the set of polymerizable oligomers comprises polar monomers, nonpolar monomers, protic monomers, aprotic monomers, solvophobic monomers, or solvophillic monomers.


In some embodiments, the set of polymerizable oligomers comprises a linear topology, branched topology, star topology, dendritic topology, hyperbranched topology, bottlebrush topology, ring topology, catenated topology, or any combination thereof. In some embodiments, the set of polymerizable oligomers comprises 3-armed topology, 4-armed topology, 5-armed topology, 6-armed topology, 7-armed topology, 8-armed topology, 9-armed topology, or 10-armed topology.


In some embodiments, the set of polymerizable oligomers comprises at least about 2 monomers, at least about 3 monomers, at least about 4 monomers, at least about 5 monomers, at least about 6 monomers, at least about 7 monomers, at least about 8 monomers, at least about 9 monomers, at least about 10 monomers, at least about 20 monomers, at least about 30 monomers, at least about 40 monomers, at least about 50 monomers, at least about 60 monomers, at least about 70 monomers, at least about 80 monomers, at least about 90 monomers, at least about 100 monomers, at least about 200 monomers, at least about 300 monomers, at least about 400 monomers, at least about 500 monomers, at least about 600 monomers, at least about 700 monomers, at least about 800 monomers, at least about 900 monomers, at least about 1000 monomers, at least about 2000 monomers, at least about 3000 monomers, at least about 4000 monomers, at least about 5000 monomers, at least about 6000 monomers, at least about 7000 monomers, at least about 8000 monomers, at least about 9000 monomers, at least about 10000 monomers, at least about 20000 monomers, at least about 30000 monomers, at least about 40000 monomers, at least about 50000 monomers, at least about 60000 monomers, at least about 70000 monomers, at least about 80000 monomers, at least about 90000 monomers, or at least about 100000 monomers. As used herein, “about” may mean plus or minus 1 monomer, plus or minus 10 monomers, plus or minus 100 monomers, plus or minus 1000 monomers, plus or minus 10000 monomers, or plus or minus 100000 monomers.


In some embodiments, the set of polymerizable oligomers comprises a homopolymer, a copolymer, a random copolymer, a block copolymer, an alternative copolymer, a copolymer with regular repeating units, or any combination thereof.


In some embodiments, the set of polymerizable oligomers comprises 1 type of monomer, 2 types of monomers, 3 types of monomers, 4 types of monomers, 5 types of monomers, 6 types of monomers, 7 types of monomers, 8 types of monomers, 9 types of monomers, or 10 types of monomers.


In some embodiments, the polymerizable composition may comprise a mix of unfunctionalized or unmodified oligomers and polymerizable oligomers as described herein. In some embodiments, the unfunctionalized or unmodified oligomers may act as porogens to generate pores within the polymer matrix.


The polymerizable reporters may comprise any of the reporters described herein. In some embodiments, the set of polymerizable reporters may comprise one or more functional groups. In some embodiments, the functional group may comprise a single stranded nucleic acid, a double stranded nucleic acid, an acrydite group, a 5′ thiol modifier, a 3′ thiol modifier, an amine group, a I-Linker™ group, methacryl group, or any combination thereof. One of ordinary skill in the art will recognize that a variety of functional groups may be used with the set of polymerizable reporters depending on the desired properties of the polymerizable reporters.


In some embodiments, the set of initiators may comprise one or more photoinitiators or thermal initiators. In some embodiments, the set of initiators may comprise cationic initiators, anionic initiators, or radical initiators. In some embodiments, the set of initiators may comprise AIBN, AMBN, ADVN, ACVA, dimethyl 2,2′-azo-bis(2methylpropionate), AAPH, 2,2′-azobis[2-(2-imidazolin-2-yl)-propane] dihydrochloride, TBHP, cumene hydroperoxide, di-tert-butyl peroxide, dicumyl peroxide, BPO, dicyandamide, cyclohexyl tosylate, diphenyl(methyl)sulfonium tetrafluoroborate, benzyl(4-hydroxyphenyl)-methylsulfonium hexafluoroantimonate, (4-hydroxyphenyl)methyl-(2-methylbenzyl)sulfonium hexafluoroantimonate, camphorquinone, acetophenone, 3-acetophenol, 4-acetophenol, benzophenone, 2-methylbenzophenone, 3-methylbenzophenone, 3-hydroxybenzophenone, 3,4-dimethylbenzophenone, 4-hydroxybenzophenone, 4-benzoylbenzoic acid, 2-benzoylbenzoic acid, methyl 2-benzoylbenzoate, 4,4′-dihydroxybenzophenone, 4-(dimethylamino)-benzophenone, 4,4′-bis(dimethylamino)-benzophenone, 4,4′-bis(diethylamino)-benzophenone, 4,4′-dichlorobenzophenone, 4-(p-tolylthio)benzophenone, 4-phenylbenzophenone, 1,4-dibenzoylbenzene, benzil, 4,4′-dimethylbenzil, p-anisil, 2-benzoyl-2-propanol, 2-hydroxy-4′-(2-hydroxyethoxy)-2-methylpropiophenone, 1-benzoylchclohexanol, benzoin, anisoin, benzoin methyl ether, benzoin ethyl ether, benzoin isopropyl ether, benzoin isobutyl ether, o-tosylbenzoin, 2,2-diethoxyacetophenone, benzil dimethylketal, 2-methyl-4′-(methylthio)-2-morpholinopropiophenone, 2-benzyl-2-(dimethylamino)-4′-morpholinobutyrophenone, 2-isonitrosopropiophenone, anthraquinone, 2-ethylantraquinone, sodium anthraquinone-2-sulfonate monohydrate, 9,10-phenanthrenequinone, 9, 10-phenanthrenequinone, dibenzosuberenone, 2-chlorothioxanthone, 2-isopropylthioxanthone, 2,4-diethylthioxanthen-9-one, 2,2′bis(2-chlorophenyl)-4,4′,5,5′-tetraphenyl-1,2′-biimidazole, diphenyl(2,4,6-trimethyl-benzoyl)phosphine oxide, phenylbis(2,4,6-trimethyl-benzoyl)phosphine oxide, lithium phenyl(2,4,6-trimethylbenzoyl)phosphinate, diphenyliodonium trifluoromethanesulfonate, diphenyliodonium hexafluorophosphate, diphenyliodonium hexafluoroarsenate, bis(4-tert-butylphenyl)-iodonium triflate, bis(4-tert-butylphenyl)iodonium hexafluorophosphate, 4-isopropyl-4′-methyl-diphenyliodonium tetrakis(pentafluorophenyl)borate, [4-[(2-hydroxytetradecyl)-oxy]phenyl]phenyliodonium hexafluoroantimonate, bis[4-(tert-butyl)phenyl]-iodonium tetra(nonafluoro-tert-butoxy)aluminate, cyclopropyldiphenylsulfonium tetrafluoroborate, triphenylsulfonium bromide, triphenylsulfonium tetrafluoroborate, tri-p-tolylsulfonium triflate, tri-p-tolylsulfonium hexafluorophosphate, 4-nitrobenzenediazonium tetrafluoroborate, 2-(4-methoxyphenyl)-4,6-bis(trichloromethyl)-1,3,5-triazine, 2-(1,3-benzodioxol-5-yl)-4,6-bis(trichloromethyl)-1,3,5-triazine, 2-(4-methoxystyryl)-4,6-bis(trichloromethyl)-1,3,5-triazine, 2-(3,4-dimethoxystyryl)-4,6-bis(trichloromethyl)-1,3,5-triazine, 2-[2-(Furan-2-yl)vinyl]-4,6-bis(trichloromethyl)-1,3,5-triazine, 2-[2-(5-methylfuran-2-yl)vinyl]-4,6-bis(trichloromethyl)-1,3,5-triazine, 2-(9-oxoxanthen-2-yl)proprionic acid 1,5,7-triazabicyclo[4.4.0]dec-5-ene salt, 2-(9-oxoxanthen-2-yl)proprionic acid 1,5-diazabicyclo[4.3.0]non-5-ene salt, 2-(9-oxoxanthen-2-yl)proprionic acid 1,8-diazabicyclo[5.4.0]-undec-7-ene salt, acetophenone O-benzoyloxime, 2-nitrobenzyl cyclohexylcarbamate, 1,2-bis(4-methoxyphenyl)-2-oxoethyl cyclohexylcarbamate, tert-amyl peroxybenzoate, 4,4-azobis(4-cyanovaleric acid), 1,1′-azobis(cyclohexanecarbonitrile), 2,2′-azobisisobutyronitrile, benzoyl peroxide, 2,2-bi(tert-butylperoxy)butane, 1,1-bis(tert-butylperoxy)cyclohexane, 2,5-bis(tert-butylperoxy)-2,5-dimethylhexane, bis(1-(tert-butylperoxy)-1-methylethyl)benzene, 1,1-bis(tert-butylperoxy)-3,3,5-trimethylcyclohexane, tert-butyl hydroperoxide, tert-butyl peracetate, tert-butyl peroxide, tert-butyl peroxybenzoate, tert-butylperoxy isopropyl carbonate, cumene hydroperoxide, cyclohexanone peroxide, dicumyl peroxide, lauroyl peroxide, 2,4-pentanedione peroxide, peracetic acid, potassium persulfate, 2-Hydroxy-2-methylpropiophenone, or any combination thereof. One of ordinary skill in the art will recognize that a variety of initiators may be used depending on the desired reaction conditions and chemistries.


In some embodiments, the initiation stimulus is UV light. In some embodiments, the initiation stimulus is UV light through a photomask. In some embodiments, the initiation stimulus is heat.


In some embodiments, the hydrogel may comprise a circular cross-sectional shape, a rectangular cross-sectional shape, a star cross-sectional shape, a dollop shape, an amorphous shape, or any shape of interest, or any combination thereof (e.g., as shown in FIGS. 79A-79B).


In some embodiments, a mask may be used to shape the initiation stimulus deposition on the polymerizable components (e.g., oligomers, etc.) and thereby shape the resulting polymer matrix. In some embodiments, the mask may comprise a circular shape, a rectangular shape, a star shape, a dollop shape, an amorphous shape, or any shape of interest, or any combination thereof.


Hydrogel Compositions with Immobilized Reporters



FIG. 78 and FIGS. 79A-79B show examples of hydrogels comprising immobilized reporters. In some aspects, provided herein are compositions comprising a hydrogel (14901) comprising (a) a network of covalently bound oligomers (14903) and (b) immobilized reporters (14902) covalently bound to said network (14903).



FIG. 78 shows an exemplary hydrogel (14901) comprising a plurality of reporters (14902) co-polymerized with a plurality of oligomers (modified and unmodified) to form a network or matrix (14903). FIGS. 79A-79B show exemplary multiplexing schemes utilizing hydrogel-immobilized reporters which may be implemented in any of the devices or methods described herein. Multiplexing could be distinguished through spatial multiplexing by knowing the location of hydrogels functionalized with each guide nucleic acid and/or through shape, by using different shapes of hydrogel for each guide nucleic acid.


In some embodiments, the composition may comprise a hydrogel (15001) comprising (a) a polymer network comprising covalently bound oligomers co-polymerized with reporters (15002) to covalently bind and immobilize the reporters to said network, and (b) immobilized programmable nuclease complexes covalently bound to said network (e.g., via co-polymerization or after reporter-immobilized polymer formation), wherein said programmable nuclease complexes may comprise a programmable nuclease (15004) and a guide nucleic acid (15003). In some embodiments, the guide nucleic acid (15003) and/or the programmable nuclease (15004) may be immobilized to or in the hydrogel as described herein (e.g., during or after formation of the hydrogel).


In some embodiments, the network of covalently bound oligomers may comprise a network formed by polymerizing one or more PEG species. In some embodiments, the network of covalently bound oligomers may comprise a network formed by polymerizing PEG comprising acrylate functional groups. In some embodiments, the acrylate functional groups may be PEG end groups. In some embodiments, the network may be formed by polymerizing PEG comprising acrylate functional groups with unmodified PEG. The molecular weight of the acrylate-modified PEG (e.g., PEG-diacrylate) and the unmodified PEG may be the same or different.


In some embodiments, the network of covalently bound oligomers may comprise a network formed from polymerizing one or more PEG species, wherein each PEG species may comprise a linear topology, branched topology, star topology, dendritic topology, hyperbranched topology, bottlebrush topology, ring topology, catenated topology, or any combination thereof. In some embodiments, the network of covalently bound oligomers may comprise a network formed from polymerizing one or more PEG species comprising a 3-armed topology, a 4-armed topology, a 5-armed topology, a 6-armed topology, a 7-armed topology, a 8-armed topology, a 9-armed topology, or a 10-armed topology.


In some embodiments, the immobilized reporter may comprise a reporter molecule covalently bound to a linker molecule, wherein the linker molecule is covalently bound to the hydrogel (e.g., via co-polymerization with the oligomers as described herein). In some embodiments, the linker molecule may comprise a single stranded nucleic acid, a double stranded nucleic acid, an acrydite group, a 5′ thiol modifier, a 3′ thiol modifier, an amine group, a I-Linker™ group, or any combination thereof. One of ordinary skill in the art will recognize that a variety of linker molecules may be used.


In some cases, the immobilized guide nucleic acid may comprise a guide nucleic acid covalently bound to a linker molecule, wherein the linker molecule is covalently bound to the hydrogel. In some embodiments, the linker molecule may comprise a single stranded nucleic acid, a double stranded nucleic acid, an acrydite group, a 5′ thiol modifier, a 3′ thiol modifier, an amine group, a I-Linker™ group, or any combination thereof. One of ordinary skill in the art will recognize that a variety of linker molecules may be used.


In some cases, the immobilized programmable nuclease may comprise a programmable nuclease covalently bound to a linker molecule, wherein the linker molecule is covalently bound to the hydrogel. In some embodiments, the linker molecule may comprise a single stranded nucleic acid, a double stranded nucleic acid, an acrydite group, a 5′ thiol modifier, a 3′ thiol modifier, an amine group, a I-Linker™ group, or any combination thereof. One of ordinary skill in the art will recognize that a variety of linker molecules may be used.


Methods of Using Hydrogels with Immobilized Reporters


Any of the methods described herein may utilize hydrogels (14901) with immobilized reporters (14902) for target detection assays. In some embodiments, the hydrogel (14901) comprises (a) a network of covalently bound oligomers (14903) and (b) immobilized reporters (14902) covalently bound to said network (14903) as shown in FIG. 78. A solution comprising target nucleic acid molecules and programmable nuclease complexes may be applied to the hydrogel (e.g., by pipetting or flowing over the hydrogel). The immobilized reporters (14902) may comprise a nucleic acid with a sequence cleavable by the programmable complex when the programmable nuclease complex is activated by binding of its associated guide nucleic acid to a target nucleic acid molecule as described herein. When activated, the programmable nuclease complex may trans-cleave the cleavable nucleic acid of the reporter molecule and generates a detectable signal as described herein. For example, the reporter may comprise a detection moiety which may be release upon cleavage of the reporter as described herein. The detection moiety may comprise FAM-biotin which may be captured by one or more capture molecules coupled to a substrate (e.g., a lateral flow assay strip) at a detection location as described herein. Detection of the detectable signal generated at the detection location by the detection moiety may indicate the presence of the target nucleic acid in the sample as described herein.


Any of the multiplexing methods described herein may utilize hydrogels (15001a, 15001b, 15001c, etc.) with immobilized reporters (15002) for multiplexed target detection assays. In some embodiments, each hydrogel (15001a, 150001b, 15001c, etc.) may comprise (a) a polymer network of covalently bound oligomers co-polymerized with reporters (15002) to covalently bind and immobilize the reporters to said network, and (b) one or more immobilized programmable nuclease complexes covalently bound to said network as shown in FIGS. 79A-79B. Each of the programmable nuclease complexes may comprise a programmable nuclease (15004a, 15004b, 15004c, etc.) and a guide nucleic acid (15003a, 15003b, 15003c, etc.). In some embodiments, the guide nucleic acid (15003) and/or the programmable nuclease (15004) may be immobilized to or in the hydrogel as described herein (e.g., during or after formation of the hydrogel). In some embodiments, multiplexing for a plurality of different targets may be facilitated by providing a plurality different and/or spatially separated hydrogels comprising a plurality of different DETECTR reaction components. In some embodiments, each hydrogel may comprise a different programmable nuclease as described herein. Alternatively, or in combination, each hydrogel may comprise a different guide nucleic acid configured to bind to a different target nucleic acid sequence as described herein. Alternatively, or in combination, each hydrogel may comprise a different reporter as described herein. Alternatively, or in combination, each hydrogel may comprise a different shape and be deposited on a substrate at different detection locations. For example, as shown in FIGS. 79A-79B, a first hydrogel (15001a) may comprise a first programmable nuclease (15004a), a first guide nucleic acid (15003a) configured to bind a first target nucleic acid, and a first reporter (15002). A second hydrogel (15001b) may comprise a second programmable nuclease (15004b), a second guide nucleic acid (15003b) configured to bind a second target nucleic acid, and a second reporter (15002). A third hydrogel (15001c) may comprise a third programmable nuclease (15004c), a third guide nucleic acid (15003c) configured to bind a third target nucleic acid, and a third reporter (15002). The programmable nucleases (15004a, 15004b, 15004c) may be the same programmable nuclease or different programmable nuclease. The guide nucleic acids (15003a, 15003b, 15003c) may be different guide nucleic acids configured to recognize different target nucleic acids. The reporters (15002) may be the same reporter or different reporters. A solution comprising one or more target nucleic acid molecules may be applied to the hydrogels (15001a, 15002b, 15003c), e.g., by pipetting or flowing over the hydrogels. The immobilized reporters (15002) may comprise a nucleic acid with a sequence cleavable by the programmable nuclease complexes (15004a, 15004b, 15004c) when the programmable nuclease complexes are activated by binding of their respective guide nucleic acids (15003a, 15003b, 15003c) to their respective target nucleic acid molecules as described herein. When activated, the programmable nuclease complexes may trans-cleave the cleavable nucleic acid of the reporter molecule and generates a detectable signal at the detection location as described herein. For example, the reporter may comprise a detection moiety which may be release upon cleavage of the reporter as described herein. The detection moiety may comprise FAM-biotin as shown in FIG. 79A which may be captured by one or more capture molecules coupled to a substrate (e.g., a lateral flow assay strip) at a detection location as described herein. Alternatively, the detection moiety may comprise a quencher moiety which may be released from the hydrogel upon cleavage of the reporter, thereby allowing a fluorescent moiety on the other end of the reporter to fluoresce at the detection location comprising the hydrogel as shown in FIG. 79B. Detection of the detectable signal generated at the detection locations by the detection moiety may indicate the presence of the target nucleic acid in the sample as described herein. Each hydrogel (15001a, 15001b, 15001c) may have a different shape and detection of a target nucleic acid may comprise detecting a particular fluorescent shape corresponding to the hydrogel shape at the detection location.


Devices Comprising Hydrogels with Immobilized Reporters


Any of the systems or devices described herein may comprise one or more hydrogels with immobilized reporters.


In some embodiments, the systems and devices described herein may comprise a plurality of hydrogels each comprising reporter molecules (e.g., in order to facilitate multiplexing and/or improve signal). In some embodiments, a first hydrogel may comprise a shape different from a shape of a second hydrogel. In some embodiments, the first hydrogel may comprise a plurality of first reporter molecules different from a plurality of second reporter molecules of the second hydrogel. In some embodiments, the reporters are the same in the first and second hydrogels. In some embodiments, the first hydrogel may comprise a circular shape, a square shape, a star shape, or any other shape distinguishable from a shape of the second hydrogel. In some embodiments, the plurality of first reporter molecules may each comprise a sequence cleavable by a programmable nuclease complex comprising a first programmable nuclease and a first guide nucleic acid. In some embodiments, the plurality of second reporter molecules may each comprise a sequence not cleavable by the first programmable nuclease complex.


Any of the systems or devices described herein may comprise a plurality of hydrogels each comprising reporter molecules. For example, a first hydrogel may comprise a plurality of first reporter molecules different from a plurality of second reporter molecules of a second hydrogel. In some embodiments, the plurality of first reporter molecules may each comprise a first fluorescent moiety, wherein the first fluorescent moiety is different than second fluorescent moieties of in each of the plurality of second reporter molecules. In some embodiments, the plurality of first reporter molecules may each comprise a sequence cleavable by a first programmable nuclease complex comprising a first programmable nuclease and a first guide nucleic acid. In some embodiments, the plurality of second reporter molecules may each comprise a sequence cleavable by a second programmable nuclease complex comprising a second programmable nuclease and a second guide nucleic acid.


Any of the systems or devices described herein may comprise at least about 2 hydrogels, at least about 3 hydrogels, at least about 4 hydrogels, at least about 5 hydrogels, at least about 6 hydrogels, at least about 7 hydrogels, at least about 8 hydrogels, at least about 9 hydrogels, at least about 10 hydrogels, at least about 20 hydrogels, at least about 30 hydrogels, at least about 40 hydrogels, at least about 50 hydrogels, at least about 60 hydrogels, at least about 70 hydrogels, at least about 80 hydrogels, at least about 90 hydrogels, at least about 100 hydrogels, at least about 200 hydrogels, at least about 300 hydrogels, at least about 400 hydrogels, at least about 500 hydrogels, at least about 600 hydrogels, at least about 700 hydrogels, at least about 800 hydrogels, at least about 900 hydrogels, at least about 1000 hydrogels,


Any of the systems or devices described herein may comprise one or more compartments, chambers, channels, or locations comprising the one or more hydrogels. In some embodiments, two or more of the compartments may be in fluid communication, optical communication, thermal communication, or any combination thereof with one another. In some embodiments, two or more compartments may be arranged in a sequence. In some embodiments, two or more compartments may be arranged in parallel. In some embodiments, two or more compartments may be arranged in sequence, parallel, or both. In some embodiments, one or more compartments may comprise a well. In some embodiments, one or more compartments may comprise a flow strip. In some embodiments, one or more compartments may comprise a heating element.


In some embodiments, the device may be a handheld device. In some embodiments, the device may be point-of-need device. In some embodiments, the device may comprise any one of the device configurations described herein. In some embodiments, the device may comprise one or more parts of any one of the device configurations described herein.


Multiplexing

Methods consistent with the present disclosure include a multiplexing method of assaying for a target nucleic acid in a sample. A multiplexing method may comprise contacting the sample to a complex comprising a guide nucleic acid comprising a segment that is reverse complementary to a segment of the target nucleic acid and a programmable nuclease that exhibits sequence independent cleavage upon forming a complex comprising the segment of the guide nucleic acid binding to the segment of the target nucleic acid; and assaying for a signal indicating cleavage of at least some protein-nucleic acids of a population of protein-nucleic acids, wherein the signal indicates a presence of the target nucleic acid in the sample, wherein absence of the signal indicates an absence of the target nucleic acid in the sample. As another example, multiplexing method of assaying for a target nucleic acid in a sample, for example, comprises: a) contacting the sample to a complex comprising a guide nucleic acid comprising a segment that is reverse complementary to a segment of the target nucleic acid and a programmable nuclease that exhibits sequence independent cleavage upon forming a complex comprising the segment of the guide nucleic acid binding to the segment of the target nucleic acid; b) contacting the complex to a substrate; c) contacting the substrate to a reagent that differentially reacts with a cleaved substrate; and d) assaying for a signal indicating cleavage of the substrate, wherein the signal indicates a presence of the target nucleic acid in the sample and wherein absence of the signal indicates an absence of the target nucleic acid in the sample. Often, the substrate is an enzyme-nucleic acid. Sometimes, the substrate is an enzyme substrate-nucleic acid.


Multiplexing may also comprise targeting multiple sequences during amplification. Such amplification multiplexing may comprise targeting distinct sequences (e.g., multiple different genes), and may also comprise targeting different portions of the same target nucleic acid sequence. The buffers of the present disclosure allow for multiplexed amplification and multiplexed programmable nuclease-based detection (e.g., DETECTR) reactions to be performed within the same sample volume.


Multiplexing can be either spatial multiplexing wherein multiple different target nucleic acids are detected at the same time, but the reactions are spatially separated. Often, the multiple target nucleic acids are detected using the same programmable nuclease, but different guide nucleic acids. The multiple target nucleic acids sometimes are detected using the different programmable nucleases. Sometimes, multiplexing can be single reaction multiplexing wherein multiple different target acids are detected in a single reaction volume. Often, a single population of programmable nucleases is used in single reaction multiplexing. Sometimes, at least two different programmable nucleases are used in single reaction multiplexing. For example, multiplexing can be enabled by immobilization of multiple categories of reporters within a fluidic system, to enable detection of multiple target nucleic acids within a single sample.


Sometimes, multiplexing can be single reaction multiplexing wherein multiple different target acids are detected in a single reaction volume. Often, at least two different programmable nucleases are used in single reaction multiplexing. For example, multiplexing can be enabled by immobilization of multiple categories of reporters within a fluidic system, to enable detection of multiple target nucleic acids within a single fluidic system. Multiplexing allows for detection of multiple target nucleic acids in one kit or system. In some cases, the multiple target nucleic acids comprise different target nucleic acids to a virus, a bacterium, or a pathogen responsible for one disease. In some cases, the multiple target nucleic acids comprise different target nucleic acids associated with a cancer or genetic disorder. Multiplexing for one disease, cancer, or genetic disorder increases at least one of sensitivity, specificity, or accuracy of the assay to detect the presence of the disease in the sample. In some cases, the multiple target nucleic acids comprise target nucleic acids directed to different viruses, bacteria, or pathogens responsible for more than one disease. In some cases, multiplexing allows for discrimination between multiple target nucleic acids, such as target nucleic acids that comprise different genotypes of the same bacteria or pathogen responsible for a disease, for example, for a wild-type genotype of a bacteria or pathogen and for genotype of a bacteria or pathogen comprising a mutation, such as a single nucleotide polymorphism (SNP) that can confer resistance to a treatment, such as antibiotic treatment. Multiplexing, thus, allows for multiplexed detection of multiple genomic alleles. For example, multiplexing may comprise method of assaying comprising a single assay for a microorganism species using a first programmable nuclease and an antibiotic resistance pattern in a microorganism using a second programmable nuclease. Sometimes, multiplexing allows for discrimination between multiple target nucleic acids of different HPV strains, for example, HPV16 and HPV18. In some cases, the multiple target nucleic acids comprise target nucleic acids directed to different cancers or genetic disorders. Often, multiplexing allows for discrimination between multiple target nucleic acids, such as target nucleic acids that comprise different genotypes, for example, for a wild-type genotype and for SNP genotype. Multiplexing for multiple diseases, cancers, or genetic disorders provides the capability to test a panel of diseases from a single sample. For example, multiplexing for multiple diseases can be valuable in a broad panel testing of a new patient or in epidemiological surveys. Often multiplexing is used for identifying bacterial pathogens in sepsis or other diseases associated with multiple pathogens.


Furthermore, signals from multiplexing can be quantified. For example, a method of quantification for a disease panel comprises assaying for a plurality of unique target nucleic acids in a plurality of aliquots from a sample, assaying for a control nucleic acid control in a second aliquot of the sample, and quantifying a plurality of signals of the plurality of unique target nucleic acids by measuring signals produced by cleavage of reporters compared to the signal produced in the second aliquot. Often the plurality of unique target nucleic acids are from a plurality of viruses in the sample. Sometimes the quantification of a signal of the plurality correlates with a concentration of a unique target nucleic acid of the plurality for the unique target nucleic acid of the plurality that produced the signal of the plurality.


The methods, reagents, and devices described herein can be multiplexed by various configurations of the reagents and the support medium. In some cases, the kit or system is designed to have multiple support mediums encased in a single housing. Sometimes, the multiple support mediums housed in a single housing share a single sample pad. The single sample pad may be connected to the support mediums in various designs such as a branching or a radial formation. Alternatively, each of the multiple support mediums has its own sample pad. In some cases, the kit or system is designed to have a single support medium encased in a housing, where the support medium comprises multiple detection spots for detecting multiple target nucleic acids. Sometimes, the reagents for multiplexed assays comprise multiple guide nucleic acids, multiple programmable nucleases, and multiple single stranded reporters, where a combination of one of the guide nucleic acids, one of the programmable nucleases, and one of the single stranded reporters detects one target nucleic acid and can provide a detection spot on the detection region. In some cases, the combination of a guide nucleic acid, a programmable nuclease, and a single stranded reporter configured to detect one target nucleic acid is mixed with at least one other combination in a single reagent chamber. In some cases, the combination of a guide nucleic acid, a programmable nuclease, and a single stranded reporter configured to detect one target nucleic acid is mixed with at least one other combination on a single support medium. When these combinations of reagents are contacted with the sample, the reaction for the multiple target nucleic acids occurs simultaneously in the same medium or reagent chamber. Sometimes, this reacted sample is applied to the multiplexed support medium described herein. In some cases, the methods, reagents, and devices described herein can be multiplexed in a configuration lacking a support medium.


In some instances, the multiplexed devices, systems, fluidic devices, kits, and methods detect at least 2 different target nucleic acids in a single reaction. In some instances, the multiplexed devices, systems, fluidic devices, kits, and methods detect at least 3 different target nucleic acids in a single reaction. In some instances, the multiplexed devices, systems, fluidic devices, kits, and methods detect at least 4 different target nucleic acids in a single reaction. In some instances, the multiplexed devices, systems, fluidic devices, kits, and methods detect at least 5 different target nucleic acids in a single reaction. In some cases, the multiplexed devices, systems, fluidic devices, kits, and methods detect at least 6, 7, 8, 9, or 10 different target nucleic acids in a single reaction. In some instances, the multiplexed kits detect at least 2 different target nucleic acids in a single kit. In some instances, the multiplexed kits detect at least 3 different target nucleic acids in a single kit. In some instances, the multiplexed kits detect at least 4 different target nucleic acids in a single kit. In some instances, the multiplexed kits detect at least 5 different target nucleic acids in a single kit. In some instances, the multiplexed kits detect at least 6, 7, 8, 9, or 10 different target nucleic acids in a single kit. In some instances, the multiplexed kits detect from 2 to 10, from 3 to 10, from 4 to 10, from 5 to 10, from 6 to 10, from 7 to 10, from 8 to 10, from 9 to 10, from 2 to 9, from 3 to 9, from 4 to 9, from 5 to 9, from 6 to 9, from 7 to 9, from 8 to 9, from 2 to 8, from 3 to 8, from 4 to 8, from 5 to 8, from 6 to 8, from 7 to 8, from 2 to 7, from 3 to 7, from 4 to 7, from 5 to 7, from 6 to 7, from 2 to 6, from 3 to 6, from 4 to 6, from 5 to 6, from 2 to 5, from 3 to 5, from 4 to 5, from 2 to 4, from 3 to 4, or from 2 to 3 different target nucleic acids in a single kit. Multiplexing can be carried out in a single-pot or “one-pot” reaction, where reverse transcription, amplification, in vitro transcription, or any combination thereof, and detection are carried out in a single volume. Multiplexing can be carried out in a “two-pot reaction”, where reverse transcription, amplification, in vitro transcription, or any combination thereof, are carried out in a first volume and detection is carried out in a second volume.


In some cases, multiplexing can comprise detecting multiple targets with a single probe. Alternatively, multiplexing can comprise detecting multiple targets with multiple probes. The multiple probes can be configured to detect a presence of a particular sequence, target nucleic acid, or a plurality of different target sequences or nucleic acids.


In some cases, the combination of a guide nucleic acid, a programmable nuclease, and a single stranded reporter configured to detect one target nucleic acid is provided in its own reagent chamber or its own support medium. In this case, multiple reagent chambers or support mediums are provided in the device, kit, or system, where one reagent chamber is designed to detect one target nucleic acid. In this case, multiple support mediums are used to detect the panel of viral infections, or other diseases of interest.


Kits

Disclosed herein are kits, reagents, methods, and systems for use to detect a target nucleic acid. A target nucleic acid can be assayed for using the compositions and methods disclosed herein and used in a kit as described herein.


Disclosed herein are kits, reagents, methods, and systems for use to detect a target nucleic acid. In some cases, the target nucleic acid is from a coronavirus. In some cases, the target nucleic acid is from the SARS-COV-2 coronavirus. Any nucleic acid of the SARS-COV-2 can be assayed for using the compositions and methods disclosed herein and used in a kit as described herein. In some embodiments, the target nucleic acid comprises the N gene or the E gene of coronavirus and can be assayed for using the compositions and methods disclosed herein and used in a kit as described herein.


In some embodiments, the kit comprises the reagents and a support medium. The reagent may be provided in a reagent chamber or on the support medium. Alternatively, the reagent may be placed into the reagent chamber or the support medium by the individual using the kit. Optionally, the kit further comprises a buffer and a dropper. The reagent chamber be a test well or container. The opening of the reagent chamber may be large enough to accommodate the support medium.


Optionally, the kit further comprises a dropper. The reagent chamber be a test well or container. The opening of the reagent chamber may be large enough to accommodate the support medium. The buffer may be provided in a dropper bottle for ease of dispensing. The dropper can be disposable and transfer a fixed volume. The dropper can be used to place a sample into the reagent chamber or on the support medium. A kit may comprise a plurality of pipettes. The plurality of pipettes may be sufficient for performing one or more than one assays. A plurality of pipettes may comprise a disposable pipette.


A kit may comprise a container (e.g., a PCR tube) comprising reagents for amplification and programmable nuclease-based detection (e.g., DETECTR) reactions. The container may comprise all reagents required for the amplification and programmable nuclease-based detection reactions, or lack a particular reagent that can be provided to initiate a reaction (e.g., a LAMP activator or dNTPs for amplification, reporters for DETECTR reactions, etc.). A kit may comprise separate containers comprising amplification and programmable nuclease-based detection reagents, which can be added at specified times to a reaction chamber or vessel containing the sample. For example, a kit may comprise separate containers for amplification reagents, programmable nuclease-based detection reagents, and a buffer capable of supporting both reactions. In such a case, the sample, amplification reagents, and programmable nuclease-based detection reagents may be added to a container at different times. A kit may comprise reverse transcription reagents, such as a reverse transcriptase, an oligonucleotide primer, and dNTPs. A kit may comprise in vitro transcription reagents, such as transcription reagents comprise an RNA polymerase, a transcription primer, and nucleotide triphosphates (NTPs). A kit may comprise a reagent for stabilizing a target nucleic acid. A kit may comprise a buffer. A kit may comprise a pipette. A kit may comprise a programmable nuclease. A kit may comprise a plurality of containers comprising separate reagents. For example, a kit may comprise an amplification reagent mix within a first vial and the detection reagent mix within a second vial. A kit may comprise a control amplification reagent mix in its own vial. A kit may comprise a control nucleic acid in its own vial. A kit may comprise an amplification activator mix within its own vial. A kit may comprise multiple distinct amplification, detection, control, activator, or any other reagent mixes. For example, a kit may comprise 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 20, 25, 30, or 40 separate amplification reagent mixes, each in separate containers.


Alternatively, a reagent may be stored in a container, and later placed into a reagent chamber or support medium by the individual using the kit. The container can be any suitable container for stably storing and transporting the reagent. For example, a reagent may be stored in an Eppendorf® tube, a centrifuge tube, a vial, a screw top vial, an ampule, a bottle, a bag, or any other container capable of holding the reagent. The reagent may be provided frozen, in dry form (e.g., as a powder such as a lyophilized powder), or as a solution. A reagent may be divided between multiple containers, or may be provided in a single container.


Conversely, a container may comprise a complete set of reagents for amplification and programmable nuclease-based detection reactions targeting a nucleic acid sequence. The sample may be added to the container to initiate the reactions. A reaction within a container may be monitored in a fluorescence imaging system. A reaction within a container (e.g., a DETECTR reaction) may also be monitored by a smart phone. For example, a smartphone camera may be disposed next to a first translucent portion of a container, and a fluorophore excitation light-source (e.g., a kit-provided lamp) may be directed into a second translucent portion of a container. In some cases, a fluorescence change can be monitored just with a smart phone and ambient lighting. In some cases, an optical signal (e.g., a colorimetric change) can be monitored solely with a smartphone.


A container may comprise a seal. The seal may be irreversibly removable (e.g., a film that can be pealed from an opening into the container), reversibly removable (e.g., a lid that can be removed and refastened over an opening in the container), or puncturable (e.g., a septum or membrane that a needle may be inserted through). A puncturable seal may reseal after puncturing. A container may be provided comprising reagents for a reaction, and a seal to secure the reagents within the container.


A container may comprise multiple compartments. The compartments may be physically separated by puncturable or breakable barriers. A sample may be added to the first chamber, and after a specified period of time the membrane may be broken to release the contents of the first chamber into the second chamber. For example, a container may comprise a first chamber comprising amplification reagents separated from a second chamber comprising programmable nuclease-based detection reagents by a breakable membrane. A sample may first be added to the first chamber for amplification, and then later released into the second chamber (e.g., by breaking or piercing the breakable membrane) to initiate a programmable nuclease-based detection reaction.


In some embodiments, the kit comprises the reagents and a support medium. The reagent may be provided in a reagent chamber or on the support medium. The medium may comprise multiple partitions or chambers that are pre-loaded with reagents. A partition or chamber may comprise a seal, which may be removable or pierceable (e.g., a septum that is pierceable by a syringe). For example, a kit may comprise a well plate that is pre-loaded with reagents and covered with a removable seal.


A support medium may comprise a plurality of containers. A plurality of containers may comprise two containers comprising reagents that target different nucleic acid sequences. A support medium may provide an array of containers comprising reagents targeting different nucleic acid sequences. For example, a kit may comprise a multi-well plate comprising 24, 48, 96, 192, or 384 wells comprising reagents for detecting (e.g., amplification and programmable nuclease-based detection reagents) different nucleic acid sequences. A single sample may be partitioned between each of the wells (e.g., by a multi-channel pipette provided in the kit), the support medium may be placed within or disposed next to a detector (e.g., a fluorescence imaging system) that can generate a real-time fluorescence readout from each well, and the change in signal identified by the detector (e.g., a change in fluorescence intensity) within each well can be correlated with the presence, absence, or concentration of a particular nucleic acid sequence from the sample. The support medium may be disposable (e.g., single use).


Alternatively, a reagent may be stored in a container, and later placed into a reagent chamber or support medium by the individual using the kit. The container can be any suitable container for stably storing and transporting the reagent. For example, a reagent may be stored in an Eppendorf® tube, a centrifuge tube, a vial, a screw top vial, an ampule, a bottle, a bag, or any other container capable of holding the reagent. The reagent may be provided frozen, in dry form (e.g., as a powder such as a lyophilized powder), or as a solution. A reagent may be divided between multiple containers or may be provided in a single container.


In some embodiments, a kit for detecting a target nucleic acid comprising a support medium; a guide nucleic acid targeting a target nucleic acid segment; a programmable nuclease capable of being activated when complexed with the guide nucleic acid and the target nucleic acid segment; and a single stranded reporter comprising a detection moiety, wherein the reporter is capable of being cleaved by the activated nuclease, thereby generating a first detectable signal.


In some embodiments, a kit for detecting a target nucleic acid comprising a PCR plate; a guide nucleic acid targeting a target nucleic acid segment; a programmable nuclease capable of being activated when complexed with the guide nucleic acid and the target nucleic acid segment; and a single stranded reporter comprising a detection moiety, wherein the reporter is capable of being cleaved by the activated nuclease, thereby generating a first detectable signal. The wells of the PCR plate can be pre-aliquoted with the guide nucleic acid targeting a target nucleic acid segment, a programmable nuclease capable of being activated when complexed with the guide nucleic acid and the target sequence, and at least one population of a single stranded reporter comprising a detection moiety. A user can thus add the biological sample of interest to a well of the pre-aliquoted PCR plate and measure for the detectable signal with a fluorescent light reader or a visible light reader.


In some instances, such kits may include a package, carrier, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein. Suitable containers include, for example, test wells, bottles, vials, and test tubes. In one embodiment, the containers are formed from a variety of materials such as glass, plastic, or polymers.


The kit or systems described herein contain packaging materials. Examples of packaging materials include, but are not limited to, pouches, blister packs, bottles, tubes, bags, containers, bottles, and any packaging material suitable for intended mode of use.


A kit typically includes labels listing contents and/or instructions for use, and package inserts with instructions for use. A set of instructions will also typically be included. In one embodiment, a label is on or associated with the container. In some instances, a label is on a container when letters, numbers or other characters forming the label are attached, molded or etched into the container itself; a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert. In one embodiment, a label is used to indicate that the contents are to be used for a specific therapeutic application. The label also indicates directions for use of the contents, such as in the methods described herein.


After packaging the formed product and wrapping or boxing to maintain a sterile barrier, the product may be terminally sterilized by heat sterilization, gas sterilization, gamma irradiation, or by electron beam sterilization. Alternatively, the product may be prepared and packaged by aseptic processing.


A kit may be designed for long term storage. A kit may lose less than 5% accuracy after 3 months of storage. A kit may lose less than 5% accuracy after 6 months of storage. A kit may lose less than 5% accuracy after 9 months of storage. A kit may lose less than 5% accuracy after 12 months of storage. A kit may lose less than 5% accuracy after 18 months of storage. A kit may lose less than 5% accuracy after 24 months of storage. A kit may lose less than 5% accuracy after 36 months of storage. A kit may lose less than 10% accuracy after 3 months of storage. A kit may lose less than 20% accuracy after 3 months of storage. A kit may have a shelf life of at least 3 months after storage at room temperature storage. A kit may have a shelf life of at least 3 months after storage between 1° ° C. and 30° C. A kit may have a shelf life of at least 3 months after storage between 10° C. and 30° C. A kit may have a shelf life of at least 3 months after storage between 15° C. and 35° C. A kit may have a shelf life of at least 3 months after storage between 20° C. and 40° ° C. A kit may have a shelf life of at least 3 months after storage between 1° C. and 15° C. A kit may have a shelf life of at least 3 months after storage between 1° C. and 10° C. A kit may be designed for sub 0° C. storage. For example, a kit may comprise storage containers that do not break, seals that do not fail, and reagents that do not lose meaningful activity (e.g., <5%) after multiple freeze-thaw cycles. The kit may be shipped and stored at room temperature. The kit may be shipped and stored at a temperature between 1 and 10° C. The kit may be shipped and stored below 0° C., and then thawed prior to use.


In some embodiments, a kit for detecting a target nucleic acid comprises: an amplification reagent mix comprising target primers designed to hybridize to the target nucleic acid, and a polymerase; and a detection reagent mix comprising a programmable nuclease, a target guide nucleic acid designed to hybridize to the target nucleic acid, and a reporter.


In some embodiments, the kit further comprises: a control amplification mix comprising control primers designed to hybridize to a control nucleic acid, and the polymerase; and a control detection reagent mix comprising a programmable nuclease, a control guide nucleic acid designed to hybridize to the control nucleic acid, and the reporter.


In some embodiments, the target nucleic acid is a viral nucleic acid. In some embodiments, the target nucleic acid is a coronavirus nucleic acid. In some embodiments, the target nucleic acid is from a SARS-COV-2 N gene or a SARS-COV-2 E gene. In some embodiments, the target nucleic acid is selected from any one of SEQ ID NO: 179-SEQ ID NO: 184. In some embodiments, the guide nucleic acid has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, at least 99%, or 100% sequence identify to any one of SEQ ID NO: 318-SEQ ID NO: 327. In some embodiments, the guide nucleic acid is selected from any one of SEQ ID NO: 318-SEQ ID NO: 327.


In some embodiments, the kit further comprises: a second amplification reagent mix comprising second target primers designed to hybridize to a second target nucleic acid, and the polymerase; and a second detection reagent mix comprising the programmable nuclease, a second target guide nucleic acid designed to hybridize to the second target nucleic acid, and the reporter. In some embodiments, the second target nucleic acid is a viral nucleic acid. In some embodiments, the second target nucleic acid is a coronavirus nucleic acid. In some embodiments, the second target nucleic acid is from a SARS-COV-2 N gene or a SARS-COV-2 E gene. In some embodiments, the second target nucleic acid is selected from any one of SEQ ID NO: 179-SEQ ID NO: 184.


In some embodiments, the second target guide nucleic acid comprises a sequence that has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, at least 99%, or 100% sequence identity to any one of SEQ ID NO: 318-SEQ ID NO: 327. In some embodiments, the second target guide nucleic acid is selected from any one of SEQ ID NO: 318-SEQ ID NO: 327. In some embodiments, the control nucleic acid is from an RNase P nucleic acid. In some embodiments, the RNase P nucleic acid is an RNase P POP7 nucleic acid. In some embodiments, the control nucleic acid has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, at least 99%, or 100% sequence identify to SEQ ID NO: 220. In some embodiments, the control guide nucleic acid has at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, at least 99%, or 100% sequence identify to any one of SEQ ID NO: 320, SEQ ID NO: 326, or SEQ ID NO: 327.


In some embodiments, the kit further comprises in vitro transcription reagents. In some embodiments, the in vitro transcription reagents comprise an RNA polymerase, a transcription primer, and nucleotide triphosphates (NTPs). In some embodiments, the target primers are designed for thermal cycling amplification of the target nucleic acid. In some embodiments, the target primers are designed for isothermal amplification of the target nucleic acid. In some embodiments, the isothermal amplification comprises transcription mediated amplification (TMA), helicase dependent amplification (HDA), circular helicase dependent amplification (cHDA), strand displacement amplification (SDA), loop mediated amplification (LAMP), exponential amplification reaction (EXPAR), rolling circle amplification (RCA), ligase chain reaction (LCR), simple method amplifying RNA targets (SMART), single primer isothermal amplification (SPIA), multiple displacement amplification (MDA), nucleic acid sequence based amplification (NASBA), hinge-initiated primer-dependent amplification of nucleic acids (HIP), nicking enzyme amplification reaction (NEAR), or improved multiple displacement amplification (IMDA). In some embodiments, the target primers comprise a FIP primer, a BIP primer, a LF primer, and a LB primer. In some embodiments, the kit further comprises reverse transcription reagents. In some embodiments, the reverse transcription reagents comprise a reverse transcriptase, an oligonucleotide primer, and dNTPs.


In some embodiments, the amplification reagent mix is contained within a first vial and the detection reagent mix is contained within a second vial. In some embodiments, the control amplification mix is contained within a third vial. In some embodiments, the control nucleic acid is contained within a fourth vial. In some embodiments, the amplification activator is contained within a fifth vial. In some embodiments, the second amplification reagent mix is contained within a sixth vial and the second detection reagent mix is contained within a seventh vial. In some embodiments, the contents of the first vial, the second vial, the third vial, the fourth vial, the fifth vial, the sixths vial, the seventh vial, or combinations thereof are frozen. In some embodiments, the kit further comprising a pipette, a multi-well plate, or both. In some embodiments, the pipette is a disposable pipette.


In some aspects, a method of diagnosing a disease or condition comprises contacting a sample to the kit of any one of previous embodiments. In some embodiments, the method comprises contacting the sample to the amplification reagent mix, the detection reagent mix, or both. In some embodiments, the method comprises contacting the sample to the second amplification reagent mix, the second detection reagent mix, or both. In some embodiments, the method comprises the control nucleic acid to the control amplification mix, the control detection reagent mix, or both. In some embodiments, the method comprises cleavage of the reporter by the programmable nuclease upon contacting the control nucleic acid to the control detection reagent mix. In some embodiments, the method comprises cleavage of the reporter by the programmable nuclease upon contacting the sample to the detection reagent mix, wherein the sample comprises the target nucleic acid. In some embodiments, the method comprises cleavage of the reporter by the programmable nuclease upon contacting the sample to the second detection reagent mix, wherein the sample comprises the target nucleic acid. In some embodiments, the method comprises producing a detectable signal upon cleavage of the reporter by the programmable nuclease.


EXAMPLES

The following examples are illustrative and non-limiting to the scope of the devices, methods, reagents, systems, compositions, and kits described herein.


Example 1
Optimization of an RT-LAMP Buffer

This example describes the optimization of a buffer for use in amplification reactions. Various buffer components were interrogated through a design of experimentation (DOE) methodology to maximize the rate of a reverse transcription loop-mediated isothermal amplification (RT-LAMP). In total, 23 phosphate buffers with varying pH, ammonium sulfate concentration, potassium chloride concentration, and magnesium salt-type (summarized in TABLE 6 below) were tested for their abilities to support RT-LAMP reactions.
















TABLE 6





FORMULATION
BUFFER

(NH4)2SO4
KCL
MG(OAC)2

TWEEN-20


NAME
SYSTEM
PH
(MM)
(MM)
(MM)
MGSO4
(%)






















Test 1
Phosphate
8.2
8
38
2
0
0.1


Test 2
Phosphate
8.6
8
2
2
0
0.1


Test 3
Phosphate
8.6
2
38
2
0
0.1


Test 4
Phosphate
8.8
10
50
2
0
0.1


Test 5
Phosphate
8.8
4
14
2
0
0.1


Test 6
Phosphate
8.2
8
50
2
0
0.1


Test 7
Phosphate
8.2
2
14
2
0
0.1


Test 8
Phosphate
8.8
2
2
2
0
0.1


Test 9
Phosphate
8.6
10
50
2
0
0.1


Test 10
Phosphate
8.0
4
14
2
0
0.1


Test 11
Phosphate
8.0
4
2
2
0
0.1


Test 12
Phosphate
8.0
10
38
2
0
0.1


Test 13
Phosphate
8.0
2
50
2
0
0.1


Test 14
Phosphate
8.6
4
38
0
2
0.1


Test 15
Phosphate
8.4
2
2
0
2
0.1


Test 16
Phosphate
8.0
2
50
0
2
0.1


Test 17
Phosphate
8.2
10
2
0
2
0.1


Test 18
Phosphate
8.2
4
50
0
2
0.1


Test 19
Phosphate
8.6
10
14
0
2
0.1


Test 20
Phosphate
8.8
2
38
0
2
0.1


Test 21
Phosphate
8.0
10
26
0
2
0.1


Test 22
Phosphate
8.8
8
14
0
2
0.1


Test 23
Phosphate
8.0
8
2
0
2
0.1


Commercial
Tris
8.8
10
50
0
2
0.1


Control

















FIG. 1 shows RT-LAMP results for each buffer, measured as time to final product. The reactions were performed with two separate dilutions (Dil-1 and Dil-2) of SARS-COV-2 N-gene RNA, as well as in the absence of target nucleic acid (NTC). The reactions were performed at 62° ° C. with 0.2 μM forward and backward outer primers, 1.6 μM forward and backward inner primers, and 0.8 μM loop forward and loop backward primers. In addition to the buffer constituents listed in TABLE 6, each reaction contained 1.4 mM dNTP, 0.8 unit/μl NEB Bst 2.0 DNA polymerase; 1.4 mM dNTP, 0.56 unit/μL NEB Warmstart® RTx reverse transcriptase, 0.8 unit/μl NEB Murine Rnase inhibitor, 1 μM Invitrogen SYT09 green fluorescent nucleic acid stain, and 4.5 mM magnesium sulfate (in addition to the amount of magnesium sulfate listed in TABLE 6). A number of buffer formulations had comparable times to final product as the commercial control NEB 1 LAMP buffer (e.g., tests 12, 13 & 16).



FIG. 2 provides an effect summary for the parameters varied in the assay. The table provides the statistical significance of a variable or set of variables (listed in the ‘Source’ column) on the time to final product values shown in FIG. 1. Some rows provide a coupled effect for two variables (e.g., ‘pH*Mg source’), while others list the significance for a single variable (e.g., [KCl](2,50), which refers to KCl concentration). The variables are listed in order of statistical significance, with ‘LogWorth’ showing the negative Log10 of the p-value for each variable. The vertical line in the ‘LogWorth’ column corresponds a p-value of 0.01. As is shown in FIG. 2, KCl concentration and Mg source were found to be significant at a p-0.01 level under the conditions tested.



FIG. 3 provides a graphical representation of the anticipated effect of different buffer components on RT-LAMP reaction times. The first three columns show the effects of pH, ammonium sulfate concentration, and potassium chloride (x-axes) on RT-LAMP time to final result (y-axes). The fourth column shows a binary comparison between magnesium acetate and magnesium sulfate as the Mg2+ source on the RT-LAMP time to result. The ‘Desirability’ column shows the weighting function applied against the measured reaction rates used to generate the bottom (‘Desirability) row of FIG. 3, which charts parameter optimizations for each variable. As can be seen from the chart, higher pH, higher ammonium sulfate concentration, and lower KCl concentration may correlate with faster RT-LAMP reactions, while no effect was observed for switching Mg2+ source under the conditions tested.


Example 2
Optimization of a Buffer for Detection of a Target Nucleic Acid

This example describes the optimization of a buffer for use in CRISPR-based detection (e.g., DETECTR) reactions. Test buffers were generated by varying the composition of a buffer capable of supporting DETECTR reactions. Fifteen phosphate buffers with varying pH, ammonium sulfate concentrations, potassium acetate concentrations, detergents, and detergent concentrations (provided in TABLE 7 below) were tested for their abilities to support DETECTR reactions. In addition to the reagents listed, all buffers contained 5 mM magnesium acetate and 1% glycerol.















TABLE 7











DETERGENT


FORMULATION
BUFFER

(NH4)2SO4
KOAC

CONCENTRATION


NAME
SYSTEM
PH
(MM)
(MM)
DETERGENT
(% V/V)





















Test 1
Phosphate
8.25
0
10
Triton x-100
0.00016


Test 2
Phosphate
7.5
2
30
Triton x-100
0.00016


Test 3
Phosphate
7.75
0
2
Tween 20
0.10000


Test 4
Phosphate
8
0
20
Tween 20
0.10000


Test 5
Phosphate
8.25
2
2
Tween 20
0.10000


Test 6
Phosphate
7.5
0
10
Tween 20
0.10000


Test 7
Phosphate
7.5
0
2
Triton x-100
0.00016


Test 8
Phosphate
7.75
2
30
Tween 20
0.10000


Test 9
Phosphate
7.75
2
10
Triton x-100
0.00016


Test 10
Phosphate
7.5
2
20
Tween 20
0.10000


Test 11
Phosphate
8
0
30
Triton x-100
0.00016


Test 12
Phosphate
8.25
0
30
Tween 20
0.10000


Test 13
Phosphate
8
2
2
Triton x-100
0.00016


Test 14
Phosphate
7.75
0
20
Triton x-100
0.00016


Test 15
Phosphate
8.25
2
20
Triton x-100
0.00016


Commercial
HEPES
7.5
0
2
Triton x-100
0.00016


Control
















FIG. 4 shows the results of DETECTR reactions performed at 37° C. with the modified DETECTR buffers listed in TABLE 7, SEQ ID NO: 28 targeting SARS-COV-2 N-gene, and two separate dilutions of SARS-COV-2 N-gene LAMP amplicons, ‘Dil 1’ & ‘Dil 2’, corresponding to 1000 and 500 starting copies of the SARS-COV-2 N gene template, respectively. Controls were performed with conditions lacking target nucleic acid (NTC). As can be seen in the figure, Tests 1, 2, 4, 5 & 7-15 performed faster DETECTR reactions than the commercially available control buffer. Tests 3 & 6 did not support reactions.



FIG. 5 provides an effect summary for the parameters varied in the assay of FIG. 4. The vertical line in the ‘LogWorth’ column corresponds to a p-value of 0.01. Among the variables, only detergent was significant to a p=0.01 level, indicating that 0.1% (v/v) Tween 20 supports faster DETECTR reaction rates than 0.00016% (v/v) Triton X-100. The remaining variables had weaker correlations with DETECTR reaction rates under the conditions tested.


However, as can be seen in FIG. 6, which provides a graphical representation of measured effect sizes for various buffer constituents on DETECTR reaction times, a number of weakly correlating variables had considerable effect sizes. For example, while pH and potassium acetate concentration had p-values of 0.5345 and 0.40378, respectively, both had changes in desirability scores of nearly 0.5 in going from the lowest to highest measured concentrations. Conversely, detergent type had a p-value of 0.00874, but a change in desirability of 0.25, with Tween 20 supporting faster DETECTR reactions than Triton x-100. The absence of ammonium sulfate led to slightly faster DETECTR reactions.


Example 3
Optimization of a Dual Amplification, CRISPR Reaction Buffer

This example describes the optimization of a buffer that can support both amplification and DETECTR reactions, enabling assays that perform both reactions in a single buffer. For low titer samples (e.g., low viral titer samples), DETECTR reactions often require an amplification step prior to detection. In such cases, the amplification and DETECTR reactions are typically performed in separate buffers, potentially leading to sample loss, contamination, and potentially high user input requirements. This example provides a single buffer capable of supporting both amplification and DETECTR reactions, without hindering the kinetics of either reaction. The single buffer platform is useful for streamlining the assay process, and may reduce the time, complexity, and/or contamination during the assay step.


Buffers for amplification and DETECTR reactions are often optimized separately for dual assays. In such cases, the products of the amplification reaction need to be exchanged into a new buffer capable of supporting the DETECTR reaction prior to the detection step. For example, the inventors previously showed that RT-LAMP could be performed at 60-62° C. in 20 mM, pH 8.8 Tris buffer containing 10 mM (NH4)2SO4, 50 mM KCl, 6.5 mM MgSO4, 0.1% Tween20, and that the RT-LAMP products could be buffer exchanged into 20 mM, pH 7.5 HEPES containing 20 mM KOAc, 5 mM Mg(OAc)2, 1% glycerol, and 0.00016% Triton X-100 for 37° C. DETECTR reactions. However, these two buffer systems were not optimized for cross-compatibility.


In this example, multiple phosphate buffers were formulated based on the results from Examples 1 and 2 indicating the buffer constituents necessary for RT-LAMP and DETECTR. Five buffers were generated with varying concentrations of ammonium sulfate, potassium acetate, potassium chloride, magnesium acetate, magnesium sulfate, detergent and glycerol, and varying pH, summarized in TABLE 8 below. The rates of RT-LAMP and DETECTR were measured in each buffer, and compared against an assay utilizing separate buffers optimized for RT-LAMP and DETECTR reactions (NEB IsoAmp Tris buffer and Mg(OAc)2 containing HEPES buffer, respectively). SARS-COV-2 N-gene was the target nucleic acid in the amplification and DETECTR reactions. The DETECTR assays utilized SEQ ID NO: 28 as a programmable nuclease. Each buffer was tested with seven separate dilutions of the target nucleic acid (Dilution 1-7) and against a control sample lacking the target nucleic acid (NTC). DETECTR reactions were performed by adding 2 uL RT-LAMP product to 18 uL of DETECTR reagents in the same type of buffer (or different type of buffer for the separate buffer comparator).



















TABLE 8














DETERGENT



FORMULATION
BUFFER

(NH4)2SO4
KOAc
KCl
Mg(OAc)2
MgSO4

CONC
Glycerol


NAME
SYSTEM
PH
(mM)
(mM)
(mM)
(mM)
(mM)
DETERGENT
(%)
(%)

























Single 8
PO43−
8
2
25
0
2
0
Tween 20
0.1
1


Single 9
PO43−
8
2
20
0
5
0
Tween 20
0.1
1


Single 10
PO43−
8.25
2
15
0
5
0
Tween 20
0.1
1


DET 4
PO43−
8
0
20
0
5
0
Tween 20
0.1
1


DET 8
PO43−
7.75
2
30
0
5
0
Tween 20
0.1
1


NEB IsoAmp
Tris
8.8
10
0
50
0
2
Tween 20
0.1
0


MBuffer3
HEPES
7.5
0
2
0
5
0
Triton x-100
0.00016
1









As is shown in FIG. 7, four of the five buffers tested supported RT-LAMP and DETECTR. The total assay times for these four formulations were ˜1.5-2.5 greater than for the two buffer system. The formulation ‘DET 4’ had the shortest average total assay time of around 25 minutes, as compared to 17 minutes for the 2-buffer control system. These results not only show that it is possible to formulate a single buffer system capable of supporting separate amplification and CRISPR reactions, but also that such a buffer can enable comparably fast amplification and CRISPR reaction rates. These results also highlight pH 8.0 phosphate buffer containing 20 mM potassium acetate, 5 mM magnesium acetate, 1% glycerol and 0.1% Tween-20 as an effective buffer for amplification and CRISPR reactions under the conditions tested.


Example 4
Formulation of Tris Buffers for Amplification Assays

This example describes the formulation of a buffer RT-LAMP. This assay built on the results of Example 3, which found pH 8.0 buffer with potassium acetate, magnesium acetate, glycerol and Tween-20 to be capable of supporting amplification and CRISPR reactions. While the previous examples cover optimization of phosphate buffers, this example tested the efficacy of various Tris buffer formulations.


The rates of RT-LAMP reactions were measured in four different pH 8.0 Tris-buffer systems. Each buffer contained 20 mM potassium acetate, 5 mM magnesium acetate, 1% glycerol and 0.1% Tween-20. The buffer in test 1 contained 20 mM Tris. The buffer in test 2 contained 20 mM Tris and 2 mM ammonium sulfate. The buffer in test 3 contained 10 mM Tris. The buffer in test 4 contained 10 mM Tris and 2 mM ammonium sulfate. The rates of the four test reactions were compared against the rates of control reactions performed with NEB IsoAMP buffer (TABLE 3).



FIG. 8 shows the mean reaction times of the RT-LAMP assays. The rates of RT-LAMP were measured with seven separate dilutions of SARS-COV-2 N-gene (Dil-1 through Dil-7) and with sample containing no target nucleic acid (NTC). The rates of the four test reactions were approximately half that of the reaction performed in the NEB IsoAmp control buffer. The fastest rates were observed in test 2, which utilized 20 mM Tris buffer with 2 mM ammonium sulfate.


Example 5
Buffer Formulations for Dual Amplification, CRISPR Assays

This example describes various buffer formulations for dual amplification, CRISPR-based assays. These assays used low input titers comprising 1000 starting copies of the target nucleic acid. Dual RT-LAMP, DETECTR assays were performed in four separate buffer systems, with SARS-COV-2 RNA as the target nucleic acid and SEQ ID NO: 28 used as the programmable nuclease. The rates of the four assays were compared against an assay performed with separate buffers for the RT-LAMP and DETECTR reactions (NEB IsoAmp and MBuffer, respectively).


The compositions of the four buffers interrogated are provided in TABLE 9, with each additionally containing 1% glycerol, 0.1% (v/v) Tween 20, and 5 mM magnesium acetate. As is shown in FIG. 9, formulations 1, 3 & 4 supported assays with about half the rate of the two-buffer system. These results show that ammonium salt may not be a necessary component for phosphate buffers used in combined RT-LAMP and DETECTR assays.













TABLE 9





FORMULATION
BUFFER

(NH4)2SO4
KOAC


NAME
SYSTEM
PH
(MM)
(MM)



















1
Phosphate
8
0
20


2
Phosphate
7.75
2
30


3
Phosphate
8.25
0
30


4
Tris
8
2
20









Example 6
Buffer-Type Optimization for Amplification Reactions

This example describes the formulation of buffers that can be used for amplification reactions. Amplification reaction rates are heavily dependent on buffer conditions. Thus, buffer optimization is an important component of assay optimization. Previous examples have covered buffer optimization for a single type of buffer system (e.g., phosphate, Tris, etc.). This example covers buffer optimization across multiple types of buffer systems.


The rate of RT-LAMP was measured in ten separate buffer formulations. The RT-LAMP was performed with SARS-COV-2 N-gene as the target nucleic acid, using input titers with 1000 target nucleic acid copies. Each buffer was set to a pH of 8.0 and contained 20 mM potassium acetate, 5 mM magnesium acetate, 1% glycerol and 0.1% Tween-20. The types and concentrations of ammonium salt and buffer varied between each of the ten formulation, as is shown in TABLE 10, with the buffer designation ‘Filtered’ (formulation 10) corresponding to 10 mM phosphate buffer passed through a 0.22 micron membrane filter. A control RT-LAMP reaction was performed in NEB IsoAmp Buffer (TABLE 3).










TABLE 10








FORMULATION NAME


















1
2
3
4
5
6
7
8
9
10





Buffer
10 mM
20 mM
20 mM
20 mM
20 mM
20 mM
20 mM
20 mM
20 mM
10 mM


System
Phosphate
HEPES
HEPES
Imidazole
Imidazole
HEPES
Imidazole
Tris
Phosphate
Filtered


Ammonium
2 mM
2 mM
0 mM
2 mM
0 mM
2 mM
2 mM
2 mM
2 mM
2 mM


Salt
(NH4)2SO4
(NH4)2SO4
NH4+
(NH4)2SO4
NH4+
NH4OAc
NH4OAc
NH4OAc
NH4OAc
NH4OAc









As is shown in FIG. 10, the rates of the RT-LAMP reactions varied from 2 to only 1.1 times the rate of the RT-LAMP reaction performed in the NEB IsoAmp Buffer. Formulation 9, which contained 20 mM phosphate buffer and ammonium acetate, supported an RT-LAMP reaction that was only three minutes slower than the reaction performed in NEB IsoAmp buffer, and twice as fast as the reaction performed with half the phosphate buffer concentration and ammonium sulfate (Formulation 1). The highest performing formulation (Formulation 10 with filtered buffer) facilitated an RT-LAMP reaction that was only one minute slower than the reaction performed in the NEB IsoAmp buffer.


Example 7
Buffer-System Optimization for Low Titer Amplification Reactions

This example describes the effect of buffer-system on total assay time for assays that involve both amplification and CRISPR reactions. The rates of amplification and CRISPR reactions are strongly dependent on solution composition and reaction conditions. While a number of buffers have been optimized for fast amplification or fast CRISPR reactions, these buffers are often only compatible for use in a single type of reaction. As there is a need for buffers capable of supporting fast rates for amplification and CRISPR reactions, this example explores the buffer-system dependence on amplification and CRISPR reaction rates.


Four separate buffers were formulated for use in dual RT-LAMP, DETECTR reactions. Building on the findings of the previous examples, each buffer was set to pH 8.0 and contained 2 mM Ammonium Acetate, 20 mM Potassium Acetate, 5 mM Magnesium Acetate, 1% Glycerol (v/v), and 0.1% Tween-20 (v/v). The RT-LAMP and DETECTR reactions targeted SARS-COV-2 N-gene, and the DETECTR reactions utilized SEQ ID NO: 28 as a programmable nuclease. The assays were performed with variable input titers ranging from 250 to 1000 copies of the target nucleic acid. The times required to complete the single buffer assays were compared against the times required to complete corresponding two buffer assays utilizing NEB IsoAmp for RT-LAMP reactions and MBuffer3 for DETECTR reactions (TABLE 3).


As is shown in FIG. 11, HEPES, Imidazole, and Tris buffer systems were capable of supporting dual amplification, CRISPR reaction assays with about ⅔ the rate of the control assays performed in two separate, single-reaction optimized buffers. The total assay time varied with input titer copy number for HEPES, indicating that amplification was likely rate limiting for the HEPES buffer system. The total assay time was nearly invariant for the imidazole and Tris buffer systems, indicating that the DETECTR reaction was rate limiting for assays performed in those buffer systems. The phosphate buffer systems were able to support amplification, but failed to enable DETECTR reactions under the conditions tested.


Example 8
Phosphate Buffer Optimization Amplification and CRISPR Reactions

This example describes the optimization of a buffer for use in amplification and CRISPR reactions. The solution and condition requirements for amplification and CRISPR reactions can be divergent, often necessitating the use of separate buffers for the two reactions. Using multiple buffers in a single assay increases the assay time and user input requirements, increases the chances for contamination and sample loss, and complicates the process of automating the assay.


Four separate buffers (outlined below in TABLE 11) were formulated with a range of buffer systems, potassium concentrations, and pH values. In addition to the constituents listed in TABLE 11, each buffer contained 5 mM Mg(OAc)2, 0.1% Tween-20 (v/v), and 1% glycerol (v/v). Each buffer was used in dual RT-LAMP, DETECTR assays with varying target nucleic acid input titers (1000 copies, 500 copies, 250 copies, or 0 copies (NTC)). The rates of the reactions were compared against RT-LAMP reactions performed in NEB IsoAmp Buffer (TABLE 3) and DETECTR reactions performed in MBuffer3 (TABLE 3). The reactions targeted the SARS-COV-2 N-gene, and the DETECTR reactions utilized SEQ ID NO: 28 as a programmable nuclease.













TABLE 11





FORMULATION
BUFFER

(NH4)2SO4
KOAC


NAME
SYSTEM
PH
(MM)
(MM)



















DET 4
Phosphate
8
0
20


DET 8
Phosphate
7.75
2
30


DET 12
Phosphate
8.25
0
30


DET 4 with Tris +
Tris
8
2
20


Ammonium









As can be seen in FIG. 12 panel A, the three phosphate buffers all had comparable RT-LAMP reaction times, all of which were about 5 minutes slower than the RT-LAMP reactions performed in NEB IsoAmp Buffer. The DET 4 formulation with Tris buffer had the slowest RT-LAMP rates, requiring about twice as much time to complete RT-LAMP. However, as is shown in FIG. 12 panel B, the DET 4 formulation with Tris buffer had the fastest DETECTR reaction rates. While the RT-LAMP times varied minimally between the three phosphate buffers tested, the DETECTR reaction rates supported by the three buffers varied considerably, with DET 8 supporting the slowest reactions and DET 4 supporting the fastest reactions. These results highlight the potential importance of high pH and low ammonium sulfate content for phosphate buffers used in DETECTR reactions.


Example 9
Design of Experimentation Approach to Buffer Optimization

This example describes a multivariate optimization of a buffer for dual use in amplification and CRISPR reactions. Amplification and CRISPR reaction rates depend on solution composition and constituents. In many cases, a buffer optimized for use in an amplification or CRISPR reaction is unable to support the other type of reaction. Optimization of a buffer suitable for amplification and CRISPR reactions is a complex process, as the buffers for each type of reaction tend to be complex, and the effects imparted by individual constituents (e.g., a particular magnesium salt) can be affected by other components present in the buffer.


Thus, a design of experimentation approach was taken to separate and deconvolute the features necessary for supporting amplification and CRISPR reactions. RT-LAMP was performed at 62° C. in the buffers summarized in TABLE 6 with 0.2 μM forward and backward outer primers, 1.6 μM forward and backward inner primers, and 0.8 μM loop forward and loop backward primers. In addition to the buffer constituents listed in TABLE 6, each reaction contained 1.4 mM dNTP, 0.8 unit/μl NEB Bst 2.0 DNA polymerase; 1.4 mM dNTP, 0.56 unit/μL NEB Warmstart® RTx reverse transcriptase, 0.8 unit/μl NEB Murine Rnase inhibitor, 1 μM Invitrogen SYT09 green fluorescent nucleic acid stain, and 4.5 mM magnesium sulfate (in addition to the amount of magnesium sulfate listed in TABLE 6). DETECTR was performed at 37° ° C. with SEQ ID NO: 28 in the buffers summarized in TABLE 7. Both reactions targeted SARS-Cov-2 N gene.



FIG. 13 shows the effect size and statistical significance of different buffer parameters on the rates of RT-LAMP reactions. Panel A provides a graphical view of the anticipated effect of each buffer component on RT-LAMP times. The effects were calibrated to desirability curves that assigned higher values to faster RT-LAMP reaction times. As can be seen in the bottom row (titled ‘desirability), high pH, ammonium sulfate concentrations approaching 5 mM, high potassium chloride concentrations, and the use of magnesium sulfate as the magnesium source correlated with faster RT-LAMP reaction rates. As can be seen in panel B, potassium chloride concentration and pH were found to be statistically significant beyond a p-value threshold of 0.01.



FIG. 14 shows the effect size and statistical significance of buffer parameters on the rates of DETECTR reactions. As can be seen in panel A, increased potassium acetate concentration, the presence of ammonium, and the use of Tween-20 (rather than Triton x-100) as detergent correlated with faster DETECTR reaction rates, while pH (over the 7.5-8.25 range tested) did not correlate with DETECTR reaction rate. As can be seen in panel B, only detergent type was found to have a statistically significant effect beyond a p-value threshold of 0.01.


Example 10

Screening Buffers and Alternative Polymerases that Enable RT-LAMP to Work at Lower Temperatures (LowLAMP)


This example describes the effect of varying buffer parameters, such as higher amounts of Mg2+ and lower amounts of salt effect an RT-LAMP assay (compared to NEB IsoAmp). The following variations to the buffer compositions were tested:

    • Isothermal Buffer “IB” 1—potassium acetate instead of potassium chloride, magnesium acetate instead of magnesium sulfate
    • IB2—higher Mg2+
    • IB3—lower potassium acetate
    • IB4—with 0.25% PEG-8000


RT-LAMP was performed on the buffers listed above, with both Klenow and Bsu polymerases, the results of which are shown in FIG. 15A. LAMP mixtures were then prepared. 7 uL of the LAMP mixtures were aliquoted into wells of a 96 well plate as shown in FIG. 16 and 1 uL of isothermal buffer was pipetted into the wells of the 06 well plate, as shown in FIG. 16. 2 μL of target nucleic acid was added to each well at 500 copies/μ L and 1000 copies/rxn. The reaction was run on a qPCR machine for 90 minutes at the following temperatures—37, 40, 42, 45, 50, and 45 degrees Celsius.


Next a DETECTR assay was performed, the results of which are shown in FIG. 15B. A complexing reaction was performed using the complexing mix comprising 40 nM each of a Cas12 variant (SEQ ID NO:28) and a guide RNA. The reaction was incubated at 37 C for 30 minutes, after which a reporter substrate was added. The RT-LAMP reactions were then added as follows: 14 μL of 1× MBuffer 3 was added in wells on a 384-well plate, after which 5 μL of complexing reaction was added to the wells. Finally, 1 μL of the LowLAMP product was added to the wells on a 384-well plate.


The plate was read on a plate reader with extended gain setting for 30 minutes at 37 C. FIGS. 15A and 15B show the RT-LAMP data and DETECTR data, respectively, for all four buffers with the target nucleic acid at 1000 copies and a control. For some embodiments, RT-LAMP can be performed at lower temperatures by using Klenow(exo-) or Bsu polymerases: LowLAMP. RT-LAMP is normally performed at temperatures between 55C-70 C. Results can be seen in FIG. 15A. This temperature range is influenced by the polymerase used. In standard RT-LAMP, a Bst or Bsm polymerase is typically used, which shows peak activity above 55 C. In this embodiment, the performance of RT-LAMP at temperatures from 37 C to 50 C was evaluated with different reagents and conditions. Klenow(exo-) and Bsu polymerases were evaluated and functional RT-LAMP was demonstrated on an RNA target of SARS-COV-2 N-gene at temperatures as low as 40 C under the conditions tested. For this embodiment, the performance of these enzymes in four different buffers was tested and peak activity for Klenow(exo-) was observed at 50 C, and peak activity for Bsu was at 45 C under the conditions tested. For some embodiments, this method can be referred to as LowLAMP, as it functions at lower temperatures than standard RT-LAMP.


The RT-LowLAMP data further showed that the addition of 0.25% PEG-8000 to the buffer helps speed up amplification with the Bsu polymerase especially at the lower temperatures, under the conditions tested. Increasing Mg2+ appeared to reduce the overall reaction performance under the conditions tested while lowering the total salt content did not have much effect on overall reaction performance under the conditions tested.


Example 11
Evaluating the Performance of Cas Variants on Putative Target Sites in SARS-COV-2

This example demonstrates that Cas14a.1 (SEQ ID NO: 63) and Cas14a.3 (SEQ ID NO: 61) programmable nucleases may target and cleave distinct sequences from separate organisms. Six different DETECTR assays were performed with either Cas14a.1 (SEQ ID NO: 63) or Cas14a.3, and with one of three following three targets: (1) the N-gene of SARS-COV-2; (2) a sequence from the Mammuthus primigenius mitochondrial DNA; and (3) POP7, a target site in the human gene POP7 (RNase P). Guide nucleic acids were provided in the form of separate crRNA and tracrRNA molecules.


Ribonucleoproteins (RNPs, e.g. programmable nuclease complexes) were prepared by complexing 40 nM of a programmable nuclease with equimolar equivalents of crRNA targeting one of the three target sequences and one of two different tracrRNAs. The programmable nucleases, crRNA, and tracrRNA were incubated at 37° ° C. for 30 minutes in nuclease-free water comprising pH 7.2 phosphate and EDTA buffered saline and bovine serum albumin. The resulting ribonucleoproteins were then mixed with 100 μM of a reporter nucleic acid and transferred to ice for storage.


Assays were performed by mixing 5 μL of the ribonucleoproteins with 2 μL of 10 nM target nucleic acid and 13 μL pH 7.2 phosphate EDTA buffer in wells of a 96-well plate incubated over ice. Control reactions were prepared by instead mixing 5 μL of the ribonucleoproteins with 15 μL pH 7.2 phosphate EDTA buffer and no target nucleic acid. The plates were sealed and spun at 2000 rcf for 30 seconds, and then incubated at 50° C. over 90 minutes fluorescence readouts of the DETECTR reactions.



FIG. 17A provides results for the DETECTR reactions with a first of the tracrRNAs. As can be seen from the figure, Cas14a.3 (SEQ ID NO: 61)


was inactive in the presence of all three target sequences. Cas14a.1 (SEQ ID NO: 63) had the highest activity in the presence of the SARS COV-2 N-gene, with complete reporter cleavage in around 25 minutes. Cas14a. 1 exhibited moderate activity in the presence of the Mammuthus gene, and no activity in the presence of Human RNAse PoP P 7 under the conditions tested. The controls performed in the absence of target nucleic acid confirmed negligible resting transcollateral cleavage activity for Cas14a.1 and Cas14a.3 (SEQ ID NO: 61).



FIG. 17B provides results for the DETECTR reactions with the second of the tracrRNAs. The second tracrRNA imparted activity for Cas14a.1 (SEQ ID NO: 63) and Cas14a.3 (SEQ ID NO: 61). Cas14a.1 and Cas14a.3 exhibited activity in the presence of the target nucleic acid SARS COV-2 N-gene and the Mammuthus gene, with Cas14a.1 (SEQ ID NO: 63) exhibiting faster kinetics in the presence of the SARS COV-2 N-gene and Cas14a.3 exhibiting faster kinetics in the presence of the Mammuthus gene. Neither programmable nuclease exhibited activity in the presence of Human RNAse PoP P 7.


Example 12

HotPot: Integrating LAMP Amplification with Cas14a.1 DETECTR in a Single Reaction (One-Pot) Volume


This example describes one approach to combining isothermal amplification protocols described herein with new CRISPR systems to create one-pot DETECTR and other assays, as outlined by the strategy in FIG. 18. In particular, this example demonstrates that Cas14a.1 (SEQ ID NO: 63) programmable nucleases are compatible with LowLAMP conditions such that one may amplify a and detect target sequences in a one-pot reaction using these techniques. To develop a one-pot Cas14a. 1 LowLAMP protocol (HotPot), various DETECTR assays were performed to determine feasible one-pot conditions, as described herein. As used herein, “HotPot” refers to a one-pot reaction in which both amplification (e.g., LowLAMP) and detection (e.g., DETECTR) reactions occur simultaneously. In many embodiments, a HotPot reaction may utilize a thermostable programmable nuclease which exhibits trans cleavage activity at elevated temperatures (e.g., greater than 37 C).



FIGS. 19A-19B presents results for an embodiment of HotPot involving LAMP amplification with Cas14a.1 (SEQ ID NO: 63) DETECTR in single reaction volume (one-pot). As shown in the figure, Cas14a. 1 failed to function in one-pot LAMP conditions. Specifically, Cas14a.1 was tested to see if it can function in a one-pot reaction using LowLAMP at 50 C using Klenow(exo-) as the DNA polymerase. FIG. 19A shows the signal from SYTO9, a DNA binding dye, that indicates the production of DNA by LAMP. It was confirmed that LowLAMP was able to generate DNA amplicon in the presence of Cas14 in three different buffers. FIG. 19B shows the signal from the fluorophore-quencher (FQ) reporter in the one-pot reaction condition shown in FIG. 19A. No signal was detected in the one-pot reaction, even though DNA was generated. The results of this method suggest that Cas14 was inhibited in the LAMP reaction under the conditions tested.



FIG. 20 presents the results from screening various buffers for HotPot, Cas14a. 1 (SEQ ID NO: 63) and LowLAMP, compatibility. LowLAMP and Cas14a.1 were found to be optimally functional in different buffers. The performance of LowLAMP at 50 C in buffers that are optimal for LowLAMP (IB1, IB13, IB14) and buffers that are optimal for Cas14a. 1 (A3, H2, TM) was evaluated as well as the performance of Cas14a. 1 in the same buffers at 50 C. Results in FIG. 20 show that both LowLAMP and Cas14a. 1 were functional in the LowLAMP optimal buffers, but Cas14a.1 was most compatible with buffer IB14. This work identified a buffer condition that was compatible with both isothermal amplification and Cas14a.1 DETECTR, a key step in moving towards a one-pot reaction.



FIG. 21 shows that primers and dNTPs were found to have the greatest inhibitory effect on Cas14 performance in one-pot reactions. The impact of individual components of LowLAMP on the performance of Cas14 were tested to determine which components might be responsible for the inhibition seen when attempting one-pot reactions. Each component was added to the Cas14a.1 (SEQ ID NO: 63) reaction with 1 nM of target dsDNA and the reaction was run for 60 minutes at 50 C. The results indicate that primers and dNTPs contribute to the inhibition of LAMP. Therefore, it was necessary to investigate whether lower concentrations of these components could be tolerated in LowLAMP.


In some embodiments, LAMP functions with lower concentrations of dNTPs and primers. FIG. 22 shows that LowLAMP functions with lower concentrations of dNTPs and primers as compared to other methods described herein (e.g., Examples 1-11, etc.). The performance of lower concentrations of dNTPs and primers was tested to see the impact on the performance of LowLAMP using Klenow(exo-) at 50 C. For this embodiment, results show a time-to-result where a lower value indicates faster amplification. (a) Titration of dNTPs shows that LowLAMP functions down to 0.8 mM dNTPs. The performance of the assay was improved at 1.2 mM and less versus the standard 1.4 mM. (b) Titration of primers in LowLAMP shows that going below 0.5× of primers can have an inhibitory effect. Together these results suggest that the dNTPs and primers concentrations can be reduced without negatively impacting LowLAMP performance, which was investigated to help relieve the observed inhibition of Cas14.


The results of the primer and dNTPs titrations were applied to the one-pot Cas14a. 1 (SEQ ID NO: 63) DETECTR and LowLAMP reaction. In some embodiments, one-pot Cas14a.1 DETECTR with LowLAMP was tested at 50 C. For such embodiments, one-pot DETECTR using Cas14 and LowLAMP at 50 C using a Klenow(exo-) DNA polymerase were tested at lower primer and dNTP concentrations. As demonstrated in FIGS. 23A-23B, one-pot DETECTR using Cas14a.1 (SEQ ID NO: 63) and LowLAMP at 50 C using a Klenow(exo-) DNA polymerase was shown to be functional under lower primer and dNTPs conditions. Other examples have suggested that Cas14a.1 may be inhibited at the standard 1.4 mM and 1× primer concentration. FIG. 23A shows signal from Cas14a.1 (SEQ ID NO: 63) one-pot. One-pot DETECTR reactions using a Cas14 were either complexed with a SARS-COV-2 N-gene crRNA that targeted the amplicon generated by LowLAMP, or complexed with a non-target crRNA that targeted a gene in the Mammuthus primigenius mtDNA. Results from this embodiment, showed a signal only in the reaction that were run with target RNA and the crRNA targeting the amplicon. No signal was seen when there was no target present, or the Cas14a. 1 crRNA was targeting the Mammuthus gene FIG. 23B shows the generation of the target of interest by LAMP, amplicons generated in the one-pot reaction were run in a Cas12 DETECTR reaction using a SARS-COV-2 N-gene crRNA to confirm the results in FIG. 23A. These results demonstrated that the amplicon was generated when the target RNA was present. Furthermore, the results indicate that the one-pot reaction is functional at 0.8 and 0.6 mM dNTPS, but not at 1.4 mM dNTPs at least in part due to Cas14a.1 inhibition at this concentration of dNTPs under the conditions tested.


In some embodiments, Cas14a. 1, or another thermostable Cas effector protein, may be used with a polymerase (e.g., a Bsm DNA polymerase, a Bst DNA polymerase, a Klenow(exo-) DNA polymerase, or a Bsu DNA polymerase) (55° C.) for the HotPot assay. The one-pot reaction may be faster if the reaction temperature is increased from 50° C. to 55° C.. However, the DNA polymerase used in LowLAMP (Klenow(exo-)) was not functional at 55° C., so here Bsm DNA polymerase was used, which worked more robustly at 55° C. than other LAMP polymerases tested such as Bst. Several different concentrations of dNTPs and primers were tested and performance of the assay was assessed. Results shown in FIG. 24 indicate that a signal was generated by Cas14a.1 (SEQ ID NO: 63) when Cas14a. 1 had the crRNA that matched the target amplicon (SARS-Cov-2 N-gene) and when the target RNA was present in the reaction. The speed of the reaction was fastest with 1 mM dNTPs, but the overall signal was lower than when 0.8 and 0.6 mM dNTPs were used.


The initial performance of the one-pot DETECTR reaction, called HotPot, was then evaluated, as demonstrated in FIG. 25. In this experiment, a limit of detection experiment for two different DNA polymerases at 55 C was performed. Cas14a. 1 crRNAs (SEQ ID NO: 63) targeting either the SARS-COV-2 N-gene or an off-target gene were included in the experiment. The assay at 1000, 500, 250, or 125 input copies/rxn of SARS-COV-2 RNA genome was tested. Robust performance of the assay at the lowest tested concentration of 125 copies/rxn for 3/3 replicates is shown in FIG. 25.


Limit of detection experiments were performed using two different DNA polymerases at 55° C. as shown in FIG. 26. Cas14a. 1 crRNAs (SEQ ID NO: 63) targeting either the SARS-CoV-2 N-gene or an off-target gene were included in the experiments and the assay at 100, 75, 50, 10, or 1 input copies/rxn of SARS-COV-2 RNA genomes were tested. Robust performance of the assay at 100 copies/rxn for 3/3 replicates was observed. At 75 copies and below several replicates did not show up, but detection down to 10 copies/rxn for 2/3 replicates was observed


The effect of different tracrRNA and sgRNA on one-pot DETECR performance were investigated. As shown in FIG. 27, a new tracrRNA R4614 was designed to be more thermostable than the native tracrRNA (R1518) for Cas14a.1 (SEQ ID NO: 63), for one-pot DETECTR uses. R4614 was used in the improved conditions previously described herein while modifying temperature to investigate whether a more thermostable tracrRNA would allow for higher reaction temperature conditions. As shown in FIG. 27, R4614 performed better than the native tracrRNA at 50 C and 55 C. Neither tracrRNA enabled a signal from either the 60 C or 65 C conditions. Similarly, a sgRNA was tested against the use of native tracrRNA and crRNA two RNA component system for Cas14a.1 (SEQ ID NO: 63) in a one-pot DETECTR reaction, as shown in FIG. 28. From this analysis, we observed that the sgRNA outperformed the two RNA component system at both 50 C and 55 C. These results suggest that sgRNA and thermostable non-native tracrRNAs can improve Cas14a.1 (SEQ ID NO: 63) performance in DETECR assays under the conditions tested.


Example 13
One-Pot NECTR: NEAR Amplification+Cas14a.1 DETECTR in Single Reaction Volume

This example describes one approach to combining isothermal amplification protocols described herein with new CRISPR systems to create one-pot DETECTR and other assays, as outlined by the strategy in FIG. 18. In particular, this example demonstrates that Cas14a. 1 programmable nucleases are compatible with NEAR conditions such that one may amplify a and detect target sequences in a one-pot reaction using these techniques. To develop a one-pot Cas14a.1 NEAR protocol (NECTR), various DETECTR assays were performed to determine feasible one-pot conditions, as described herein.


For some embodiments, replacing Bst polymerase in NEAR can enable SARS-COV-2 detection at lower temperatures as shown in FIG. 29. Many NEAR protocol use Bst 2.0 to generate an amplicon from SARS-COV-2 at 60 C. This polymerase also functions at 55 C, but at reduced capacity. Accordingly, the performance of Bsu and Klenow(-exo) polymerases at lower temperatures was evaluated for these embodiments. It was found that Klenow(-exo) polymerase enabled robust amplification at 55 C and 50 C. FIG. 29 shows a Cas14a.1 DETECTR reaction following a NEAR amplification reaction. Amplification reactions were performed at the indicated temperature for 10 minutes in a buffer that was composed of a mixture of 1× IsoAmp buffer+0.5× CutSmart buffer. It was observed in this embodiment that Cas14a. 1 shows peak activity at 55 C and 50 C, but not at 60 C.


For some embodiments, NEAR amplification functions in Cas14a. 1 optimal buffers as shown in FIG. 30. The optimal buffer for Cas14a. 1 has less salt and higher Mg2+ than the optimal buffer for NEAR amplification. Therefore, the performance of NEAR amplification in two buffers for Cas14a: 1× TM Buffer and 1× H2 Buffer was investigated. Amplification reactions were performed with the indicated polymerase, buffer, and reaction temperature with Nt.BstNBI, Omniscript RT, M2805 FWD, M2811 REV. FIG. 30 shows the result of a Cas14a.1 DETECTR reaction to evaluate the amount of amplicon produced. For this embodiment, results indicate that TM Buffer and H2 Buffer may function for NEAR amplification. However, results suggest that the reactions produced ˜10 times less amplicon than an optimal NEAR reaction. This suggests room for further buffer optimization. 1× TM Buffer used in the experiments contained: 20 mM Tricine, pH 8.5; 2 mM KOAc; 100 μg/mL BSA; 15 mM MgOAc. 1× H2 Buffer used in the experiments contained: 20 mM Tris-HCl, pH 8.8; 2 mM KOAc; 0.1% Tween-20; 17.5 mM MgOAc.


Cas14a. 1 functioned in a range of KOAc salt concentrations as shown in FIG. 31. In some embodiments, Cas14a. 1 reaction buffer (H2 Buffer) contains 2 mM KOAc. In some embodiments, amplification reactions often require higher concentrations for optimal efficiency. In some embodiments, the performance of a Cas14a.1 DETECTR reaction in increasing concentrations of KOAc was investigated. In some embodiments, Cas14a.1 DETECTR reaction was evaluated with tracrRNA R1518, crRNA R2424, at a target concentration of 1 nM, and a reaction temperature of 50 C. Results shown in FIG. 31 indicate that Cas14a. 1 is maximally active at 2-10 mM KOAc, but remains robustly active up to 60 mM. At 70 mM KOAc performance of Cas14a. 1 begins to drop off. In the experiments 1× H2 Buffer was composed of: 20 mM Tris-HCl; pH 8.8; 2 mM KOAc; 0.1% Tween-20; and 17.5 mM MgOAc.


In some embodiments, increasing concentrations of KOAc may improve NEAR performance in Cas14a. 1 optimal buffers, as seen in FIG. 32. NEAR amplification in Cas14a.1 optimal reaction buffers was initially seen to be less efficient than when amplification is performed in the optimal NEAR buffer (1× IsoAmp+0.5× CutSmart). The impact of increasing amounts of KOAc on the performance of NEAR amplification in the background of a Cas14a. 1 optimal amplification buffer (H2 Buffer) was investigated. As seen in FIG. 32 results show how Cas14a.1 DETECTR assay performed after NEAR amplification to measure amplification efficiency. Results indicate that increasing KOAc to up to 60 mM improved the amount of NEAR amplicon generated under the conditions tested. From 2 mM to 60 mM KOAc were functional ranges for Cas14a.1 under the conditions tested. In these experiments 1× H2 Buffer contained: 20 mM Tris-HCl, pH 8.8; 2 mM KOAc; 0.1% Tween-20; 17.5 mM MgOAc. In some embodiments, it was also shown that increasing concentrations of KOAc improved NEAR performance in Cas14a.1 optimal buffers as seen in FIG. 33.


Performance of Cas14a.1 crRNAs on SARS-COV-2 E-gene amplicon are shown in FIGS. 34A-34B. In some embodiments, Cas14a.1 crRNAs, were designed to target the SARS-CoV-2 E-gene NEAR amplicon generated by primers M2805/M2811. Results are shown in FIG. 34. FIG. 34A shows the position of the crRNAs and primers. R1765 and R1764 are Cas14a. 1 crRNAs that target the amplicon. PAM sequences for Cas14a. 1 are indicated, but there are no TTTR PAMs for Cas14a. 1 to use within this amplicon. FIG. 34B shows the performance of Cas14a. 1 crRNAs on NEAR amplicon (generated at 60 C for 15 minutes). All tested crRNAs worked robustly with the least amount of background from R3960, R3961, and R3962.


In some embodiments, the performance of Klenow(exo-) NEAR assay in IB13 buffer at decreasing salt concentrations was evaluated as shown in FIG. 35. In these embodiments, Standard NEAR reaction buffer was composed of a mixture of 1× IsoAmp buffer and 0.5× NEBuffer 3.1, which gave a final salt concentration of 100 mM. Variations of the IB13 buffer, with various concentrations of KOAc as the salt, were tested. In these buffer variations the NEAR assay using Klenow(exo-) as the polymerase was run at 55 C for 20 minutes. To readout the amount of amplicon produced, a Cas14a. 1 DETECTR assay on 2 μL of the NEAR amplicon was performed. The results indicate that the best NEAR performance was with 100 mM KOAc, similar to the standard NEAR reaction buffer, and that the amount of amplicon produced was reduced as the salt is reduced. The assay has acceptable performance at 80-70 mM KOAc. In these experiments, the IB13 buffer had a composition of: 20 mM Tris-HCl, pH 8.8; 10 mM (NH4)2SO4; 50 mM KOAc; 5 mM MgOAc; 1% Tween-20; 1 mg/mL BSA.


Example 14

One-Pot sRCA: Rolling Circle Amplification with Cas14a.1, a Cas12 Variant, or a Cas13 DETECTR Reaction in Single Reaction Volume


This example describes one approach to combining isothermal amplification protocols described herein with new CRISPR systems to create one-pot DETECTR and other assays, as outlined by the strategy in FIG. 18. In particular, this example demonstrates that Cas14a. 1 programmable nucleases are compatible with RCA conditions such that one may amplify a and detect target sequences in a one-pot reaction using these techniques. To develop a one-pot Cas14a. 1 RCA protocol (One-pot sRCA), various DETECTR assays were performed to determine feasible one-pot conditions, as described herein.



FIG. 36 presents an overview of sRCA. In this system, a target nucleic acid is added to a system that contains components for a one-pot RCA+Cas protein reaction. The RCA portion of the system is composed of a dumbbell-shaped DNA template, a primer, and a DNA polymerase. For some embodiments, the DETECTR portion of the reaction is composed of a Cas protein, such as a Cas12 or Cas14a.1, a crRNA that targets the amplicon generated by RCA, but not the dumbbell-shaped DNA template, and a FQ ssDNA reporter. A target nucleic acid (e.g. viral RNA), which is capable of binding to the dumbbell DNA template, is added to the system. The target nucleic acid base pairs with the DNA template. The more extensive base pairing between the target and the DNA template causes the internal base pairing of the dumbbell to be disrupted, which opens up a binding site for the primer. The DNA polymerase can then use this primer to begin RCA. In some embodiments, As RCA proceeds, the amplicon is generated which contains the target site for the Cas protein. The Cas protein recognizes this site through base pairing with the crRNA, and initiates trans-cleavage of the FQ ssDNA reporter. In some embodiments, the system contains fewer components than other one-pot approaches and does not require a RT enzyme.


In some embodiments, dumbbell DNA templates were screened for sRCA performance, as shown in FIG. 37. In some embodiments, four dumbbell DNA templates for RCA contained a Cas12 or Cas14a. 1 target sites. For these embodiments, A DNA binding dye, SYTO9, was used to monitor whether these dumbbells were functional in RCA at a variety of temperatures for 1 hour. Results as seen in FIG. 37 indicate that only dumbbell 4 was functional in generating DNA by RCA. The peak performance of the system was seen at 35-45 C.


The performance of Cas14a. 1 to detect product of RCA reaction was monitored, as seen in FIG. 38. It was shown that Dumbbell 4 was functional in RCA and that Dumbbell 1 was not functional under the conditions tested. Either 2 μL or 5 μL of the completed RCA reactions were added to a 20 μL Cas14a.1 DETECTR reaction that contained a crRNA that was capable of detecting the amplicon generated by RCA, but not the Dumbbell DNA template. The results indicate that Cas14a.1 is capable of detecting the amplicon as expected, and that increased performance is seen with lower amounts of RCA added to the reaction.


In some embodiments of the One-Pot assay sRCA Cas14a. 1 was used, as shown in FIG. 39. The reaction was performed at 45 C in two conditions as two embodiments. In a first embodiment, the DNA template, Dumbbell 4, did not contain a trigger oligo and in the second embodiment the DNA template did have a trigger oligo. In the first embodiment with the trigger oligo condition, which initiates RCA, higher signal was observed as compared to the second embodiment condition without the trigger oligo. This demonstrates that Cas14a.1 is capable of detecting the RCA amplicon in a one-pot reaction, and that the sRCA reaction can be controlled by the presence of the trigger oligo.


In some embodiments, a trigger oligo was titrated for the Cas14a. 1 One-Pot SRCA assay. In this embodiment, the minimal concentration of trigger oligo that was required to initiate the one-pot Cas14a. 1 sRCA reaction was determined. Results shown in FIG. 40 indicate that at least 0.5 nM of trigger oligo was required to initiate the sRCA reaction under the conditions tested.


In another example, a Cas12 variant enzyme (SEQ ID NO: 28) was used in the one-pot sRCA assay. In other embodiments, it has been shown that Cas14a. 1 was capable of functioning in a one-pot sRCA reaction. In this embodiment, it was shown that the Cas12 variant was also capable of functioning in this assay at 45 C. The results from the cleavage of ssDNA FQ reporter included in the sRCA reaction are shown in FIG. 41. The results for such an embodiment indicate that Cas12 variant was capable of functioning in the one-pot sRCA format. For such an embodiments, the concentration of stock trigger oligo was: (1) 40 nM stock=2 nM final conc; (2) 20 pM stock=1 pM final conc.; and (3) 20 fm stock=1 fM final conc.



FIG. 42 presents an overview of an RCA-based positive feedback system for a Cas13 effector protein. In some embodiments, Cas13 may be programmed to recognize an RNA target. When the viral RNA target is present, a blocking motif on the 3′ end of the primer is removed. After removal of this blocking motif, the primer can then serve to open up the circular template and allow for amplification by RCA using a DNA polymerase. As the amplicon is generated the same target sequence as the original RNA is generated. This ssDNA target sequence is then capable of being recognized by Cas13 which can either remove additional blocking groups from the primer or cleave a FQ reporter that generates a fluorescent signal. This system forms a positive feedback loop for Cas13. In general, a “blocking motif” refers to a moiety at the 3′ end of a polynucleotide that impedes formation of a covalent bond between the 3′ hydroxyl moiety of the nucleotide and the 5′ phosphate of another nucleotide. A variety of suitable blocking motifs are available, non-limiting examples of which are described in US20210198730, which is incorporated herein by reference.



FIG. 43 presents results for the evaluation of Cas13-compatible DNA templates for RCA. In some embodiments, two dumbbell DNA templates for RCA contained a Cas13 ssDNA targeting site. In some embodiments, a DNA binding dye, SYTO9, was used to monitor whether these two dumbbells were functional in RCA at 30 C. The performance of the two templates was monitored with a titration of the primers that were used to trigger the amplification. The results indicate that Dumbbell 7, but not Dumbbell 8 is compatible with RCA as shown in FIG. 43 under the conditions tested.


In some embodiments, Cas13-compatible DNA template was used for RCA. FIG. 44 presents results for such embodiments, to determine whether Cas13-compatible DNA template was functional in RCA. In some embodiments, a circular DNA dumbbell for sRCA had a Cas13 ssDNA target site, known as dumbbell 7. A DNA binding dye, SYTO9, was used to monitor whether the DNA template was functional in RCA at various temperatures for two different polymerases using 2 nM of trigger oligo (the primer for sRCA). The results shown in FIG. 44 indicate that dumbbell 7 was capable of generating amplicon at temperatures from 30 to 55 C. These conditions, consisting of the utilization of dumbbell 7 for generating amplicons from 30 to 55 C, enabled for the use of the Cas13 enzyme in the OnePot reaction.


In some embodiments, a Cas13 effector protein (SEQ ID NO: 154) was used in the one-pot sRCA reaction. FIG. 45 presents results for such an embodiment, where the amplicon generated by RCA contained a ssDNA region that was capable of being recognized by a Cas13 gRNA. As the reaction proceeded, additional ssDNA target was produced, while the activated the Cas13 cleaved a FQ RNA reporter to generate a signal. The performance of this reaction at a variety of temperatures was evaluated and it was shown that the Cas13 was capable of detecting this ssDNA region of the RCA amplicon from 30-40 C, which aligns with the previously established active temperatures for the Cas13. The performance of two different polymerases at the temperatures of interest was also compared. The results shown in FIG. 79 suggest that Cas13 can be used in a one-pot reaction where RCA is the amplification method, and that the Cas13's ability to detect ssDNA was preserved in this embodiment under the conditions tested.


Example 15

CasPin: Cas13 Positive-Feedback Loop Leveraging Cas13 ssDNA Targeting


This example describes one approach to combining isothermal amplification protocols described herein with new CRISPR systems to create one-pot DETECTR and other assays, as outlined by the strategy in FIG. 18. In particular, this example demonstrates that Cas13 programmable nucleases can be used to create a feedback loop amplification system. To develop such a system (CasPin), various DETECTR assays were performed to determine feasible conditions, as described herein.



FIG. 46 presents an overview of CasPin. In some embodiments, the CasPin system uses two populations of Cas13. One is programmed with a crRNA that targets an RNA of interest (primary target), such as a viral genome. The other population is programmed with a crRNA that is optimal for ssDNA detection. The systems also contain a hairpin-shaped oligo that is composed of both DNA and RNA. Finally, in some embodiments, there is a FQ RNA reporter that is used to readout the result of the assay. When Cas13 detects the RNA of interest, it can either cleave the FQ RNA reporter or the RNA on the hairpin oligo. When the RNA on the hairpin oligo is cleaved, it dissociates from the DNA revealing a ssDNA target site (secondary target) that can be recognized by the other population of Cas13 RNPs. This initiates a positive feedback loop where Cas13 recognizes the ssDNA target and cleaves more hairpin molecules, which increases the overall amount of target in the system (either primary target or secondary target) and leads to further activation of the system. As this process continues more and more FQ RNA reporter is cleaved, which is the ultimate readout of the assay.



FIG. 47 presents some exemplary structures of hairpins for CasPin. In some embodiments, the target ssDNA sequence is indicated by the rectangle. RNA loop structures may occur on either side of the target strand (either 5′, 3′ or both). The strand that is complementary to the target site may be DNA or RNA. The strand that is complementary to the target site may also be a perfect match to the target site, be shorter than the target site, or contain mismatches to help destabilize or promote trans-cleavage by Cas13.


In some embodiments, two hairpins were used on either end of the target site. FIG. 48 presents results indicating capability for blocking Cas13 from recognizing the ssDNA target site. CasPin oligos had varying lengths of hairpin stems. In some embodiments, CasPin oligo did not have stem structures. In other embodiments, CasPin oligos contained another DNA sequence. These were evaluated and found to not be recognized by the crRNA. Both raw oligos from the manufacture and those that had been denatured and refolded at 25 C in a Cas13 DETECTR reaction were tested. The results of this experiment, shown in FIG. 48, demonstrate that Cas13 was able to recognize the target site regardless of the stem length. In some embodiments, longer stem length oligos block Cas13 recognition without RNA cleavage to release the structure, suggesting that it may be desirable to have longer hairpins.


Example 16
DETECTR Immobilization

In some embodiments, NHS-Amine chemistry may be used for immobilization of DETECTR components. FIG. 49 presents a schematic of combined gRNA and reporter immobilization and results. In this embodiment, a functional DETECTR reaction was immobilized to solid substrate (NHS plate) using primary amine modified reporters and gRNAs. In the example shown, an amine-modified reporter molecule (rep111) was bound to the surface in combination with either an unmodified crRNA (R1763) or a modified crRNA (mod027). After incubating these nucleic acids on the surface, the surface was washed 3 times, and then a Cas12 variant (SEQ ID NO: 28) was added with the target dsDNA. The immobilized DETECTR reaction was then incubated in a plate reader at 37 C for 60 minutes with continuous monitoring of the fluorescence.



FIG. 50 presents results for various embodiments involving the optimizing conjugation buffer to reduce non-specific binding of DETECTR reagents. For some embodiments, 1× Conjugation Buffer 3 (CB3) was selected as the buffer to perform binding studies. It was found that CB3 improved the binding of the amine-modified reporter (rep111) and reduced the binding of a biotinylated reporter (rep117) which should not bind to NHS covalently. The wash buffer used was 1× MB3. In some embodiments, 1× MB3: 20 mM HEPES, pH 7.2, 2 mM KOAc, 5 mM MgAc, 1% Glycerol, 0.0016% Triton X-100 was used. In other embodiments, 1× CB2: 20 mM HEPES, pH 8.0, 2 mM KOAc, 5 mM MgAc, 1% Glycerol, 0.0016% Triton X-100 was used. In this embodiment, 1× CB3: 100 mM HEPES, pH 8.0, 10 mM KOAc, 25 mM MgAc was used.


In this embodiment, different combinations of reporter+guide+the Cas12 variant may be immobilized. FIG. 51 presents results of such embodiments, involving the optimization of the assay. For such embodiments, it was found that immobilizing guide and reporter first followed by the addition of the Cas12 variant and target at the same time gave sufficient signal.


The results for optimizing gRNA and target concentrations to improve single-to-noise ratio for immobilized DETECTR assay are shown in FIG. 52. In this embodiment, guide RNA concentration is increased while keeping reporter concentration constant at 0.5 μM, as seen on the left of FIG. 52, the signal is not changed very much. In some embodiments, as seen on the right of FIG. 52, increasing target concentration 2-fold helped improve the overall signal with rep135. Additionally, for such embodiments, rep135 gave a better signal strength compared to rep111. The sequences for the two reporters were: rep111: 5AmMC6T//i6-FAMK/TTTTTTTTTTTT/3IABkFQ/(SEQ ID NO: 425) and rep135: 5AmMC12//i6-FAMK/TTTTTTTTTTTTTTTTTTT/3IABKFQ/(SEQ ID NO: 426).


In some embodiments, amino modifications were used for DETECTR immobilization, as presented in FIG. 53. For each subplot raw fluorescence was plotted against time in minutes. Each of the four subplots represent different modifications. The dashed trace represents the no target control (NTC) and the solid trace represents a 1:10 dilution of target -GF676.


Example 17
Rapid PCR (FASTR) Thermocycling

In some embodiments, rapid thermocycling and CRISPR diagnostics were used to detect SARS-COV-2. Results are shown in FIG. 54. For some embodiments, polymerase and buffer combinations were identified that enabled the rapid amplification of SARS-COV-2 using the N2 primers from the CDC assay. Assays were performed at two target concentrations: 2 input copies/rxn and 10 input copies/rxn. Reaction conditions were as follows: initial denaturation at 98 C for 30 seconds, followed by 45 cycles consisting of 1 second at 98 C and 3 seconds at 65 C. Following thermocycling, amplicon was transferred to a Cas12 variant (SEQ ID NO: 28) detection reaction for 30 minutes at 37 C. Best performing enzyme/buffer pairs shown in FIG. 54 were those that gave strong signal in both tested concentrations.


The top enzymes and buffers identified previously at various concentrations and with multiple replicates were tested for the FASTR assay. In some embodiments, the best performing enzymes and buffers as identified in the previously disclosed screening studies were used. Results of such embodiments are shown in FIG. 55. Reaction conditions of such embodiments were as follows: initial denaturation at 98 C for 30 seconds, followed by 45 cycles consisting of 1 second at 98 C and 3 seconds at 65 C. Primers used were from the CDC N2 assay for SARS-COV-2. Following thermocycling, amplicon was transferred to a Cas12 variant (SEQ ID NO: 28) detection reaction for 30 minutes at 37 C. The data present is the signal from the CRISPR reaction. Best performing enzyme/buffer pairs were those that gave strong signal at the lowest tested concentrations and with consistent detection across replicates.


For some embodiments, single copy detection of SARS-COV-2 with FASTR assay was demonstrated as shown in FIG. 56. The limit of detection of the FASTR assay was evaluated using solutions composed of 1000 copies of SARS-COV-2 per reaction to 1 copy per reaction. Reaction conditions were as follows: reverse transcription at 55 C for 60 seconds, initial denaturation at 98 C for 30 seconds, followed by 45 cycles consisting of 1 second at 98 C and 3 seconds at 65 C. Primers used were from the CDC N2 assay for SARS-COV-2. Following thermocycling, amplicon was transferred to a Cas12 variant (SEQ ID NO: 28) detection reaction for 30 minutes at 37 C. The data presented in FIG. 56 is the signal from the CRISPR reaction. It was found that the limit of detection of the CRISPR assay was 1 copy of SARS-COV-2 per reaction.


Rapid cycling times were varied to evaluate denaturation and annealing/extension for the FASTR assay, as shown in FIG. 57. Reverse transcription was run at 55 C for 60 seconds and initial denaturation at 98 C for 30 seconds. In some embodiments, the tested cycling conditions were: 98 C for 1 second, 65 C for 3 seconds; 98 C for 2 seconds, 65 C for 2 seconds; or 98 C for 1.5 seconds, 65 C for 1.5 seconds. Primers used were from the CDC N2 assay for SARS-COV-2. Following thermocycling, amplicon was transferred to a Cas12 variant (SEQ ID NO: 28) detection reaction for 30 minutes at 37 C. The results shown in FIG. 57 indicate that >2 seconds of annealing/extension time at 65 C was able to generate robust sensitivity.



FIG. 58 presents results for Minimizing RT time for FASTR. The performance of the FASTR assay was evaluated, for various embodiments, where the reverse transcription incubation times were varied holding temperature at 55 C. The results shown in FIG. 58 indicate the assay is most robust above 30 seconds of reverse transcription.



FIG. 59 presents results for Higher pH buffers improve FASTR performance. The FASTR assay utilized buffers with pH of either 9.2 or pH 7.8. The FASTR assay was evaluated using these buffer pH values. The results as shown in FIG. 59 indicate that the higher pH buffer produced superior results in terms of amplicon yield and sensitivity.


The FASTR assay compatibility with crude lysis buffers was investigated. Results are shown in FIG. 60 where there are three row groups, each consisting of two sub rows representing a buffer and control from top to bottom respectively. The buffers, VTES, A3 and Elution buffer are plotted against a control, from top to bottom, respectively. In FIG. 60 there are also seven subgroups showing the number of copies decreasing from left to right. The performance of the FASTR assay, when combined with various crude lysis buffers, was evaluated, where crude lysis buffers VTE5, A3, and the Elution Buffer from the ChargeSwitch kit (Thermo) were tested. The FASTR assay performed best for VTE5, but was performed slightly less robustly in the A3 buffer and the Elution Buffer from the ChargeSwitch kit performed similarly to the control reactions (water).


Non-optimized multiplexing of FASTR was demonstrated as shown in FIG. 61. In FIG. 61, raw fluorescence is plotted in the y-axis and time is plotted in the x-axis for each sub-plot. Each column illustrates a particular guide RNA sequence: R1965 and R1763, respectively Each row represents duplex, RNase P, N2 and the no target control, from top to bottom respectively. Initial testing of multiplexed FASTR for SARS-COV-2 and RNase P POP7 (endogenous control) showed that while the single-plex assays generated a robust signal in DETECTR, the duplex assay tended to generate a weak signal for SARS-COV-2 (R1763) and almost no signal for RNase P (R1965). Reaction conditions were as follows: reverse transcription at 55 C for 60 seconds, initial denaturation at 98 C for 30 seconds, followed by 45 cycles consisting of 1 second at 98 C and 3 seconds at 65 C. Primers from the CDC N2 assay for SARS-CoV-2, and M3637/M3638 were used.


FASTR can be used for multiplexed detection, as shown in FIG. 62. The components of a FASTR reaction, such as primer concentration, dNTP concentration, presence/absence of DMSO, and other factors, can impact the performance of a multiplex FASTR reaction. FIG. 62 shows 18 different experimental conditions for a multiplex FASTR reaction targeting human RNase P POP7 and SARS-COV-2. In FIG. 62 each row of the y-axis represents experimental runs 1-18 and each column represents the detection signal from a particular crRNA at a time point of 30 minutes in the reaction. The shading represents the value of the raw fluorescence. In some embodiments, the multiplexed FASTR assay for SARS-COV-2 and RNase P, comprise a set of SARS-COV-2 primers (M3257/M3258). A series of experiments of such embodiments was performed with varied reaction conditions containing different combinations of buffers, primer concentrations, dNTPs, and DMSO. Results identified two reaction conditions that performed robustly for the multiplex reaction. In one of these embodiments, reaction 4, conditions consisted of: 1× FastBuffer 2; 1 uM RNase P primers; 0.5 uM CoV primers; 0.2 mM dNTPs; and 2% DMSO. In another embodiment, reaction 9, conditions consisted of: 1× Klentaq1 buffer; 1 uM RNase P primers; 0.5 uM CoV primers; 0.4 mM dNTPs; and 0% DMSO. In some embodiments, normal reaction conditions consisted of reverse transcription at 55 C for 60 seconds, initial denaturation at 98 C for 30 seconds, followed by 45 cycles consisting of 1 second at 98 C and 3 seconds at 65 C. In various aspects, permissive reaction conditions consisted of reverse transcription at 55 C for 60 seconds, initial denaturation at 98 C for 30 seconds, followed by 45 cycles consisting of 3 seconds at 98 C and 5 seconds at 65 C.


The FASTR assay enables multiplexed detection. Results of a limit of detection (LOD) study of such embodiments are shown in FIG. 63. In FIG. 63, the x-axis shows the number of copies per reaction for viral RNA and the y-axis of each subplot identifies the particular crRNA. Each subplot shows nanograms of human RNA per reaction, decreasing in concentration from left to right. The 4th subplot contains no human RNA, labeled as the no target control (NTC). An optimized multiplexed FASTR assay was ran at various concentrations of human RNA and viral RNA. Results indicated that the assay performed at a range of human RNA concentrations, while maintaining a sensitivity of ˜5 copies per reaction. Results shown are from DETECTR reactions using either R1965 to detect the human RNase P, or R3185 (labeled M3309) to detect SARS-COV-2. Reaction conditions were as follows: reverse transcription at 55 C for 60 seconds, initial denaturation at 98 C for 30 seconds, followed by 45 cycles consisting of 1 second at 98 C and 3 seconds at 65 C. Primers used were M3257/M3258 (SARS-COV-2) and M3637/M3638 (RNase P). Key primers and gRNAs have the sequences as listed in FIG. 64.


Example 18
DETECTR-Based HotPot Reactions Using Thermostable Inorganic Pyrophosphatase

This example demonstrates compositions and methods for HotPot reactions using thermostable inorganic pyrophosphatase (TIPP) to improve signal generation. TIPP is an enzyme that can catalyze the hydrolysis of inorganic pyrophosphates. This example demonstrates compositions and methods for HotPot reactions using thermostable inorganic pyrophosphatase (TIPP) to amplify output signals.


HotPot reactions were carried out under with various compositions as shown in Table 12. In the Experimental Condition, the HotPot reaction was carried out in a solution comprising both TIPP and target nucleic acid. In Control A, the HotPot reaction was carried out with target nucleic acid but without TIPP. In Control B, HotPot reaction was carried out without target RNA. In Control C, HotPot reaction was carried out without TIPP and without target RNA.


Table 12 shows example amounts of components used in the HotPot reactions.















Amount in (μL)












Experimental





Component
Condition
Control A
Control B
Control C














Nuclease-free water
1.975
1.975
1.975
1.975


5X IB15
2
2
2
2


10 mM dNTP (NEB)
1
1
1
1


100 μM rep033
0.1
0.1
0.1
0.1


RNase Inhibitor,
0.05
0.05
0.05
0.05


Murine (NEB)


Bsm (Thermo)
0.5
0.5
0.5
0.5


Warmstart RTx
0.125
0.125
0.125
0.125


(NEB)


Cas14a complexing
1
1
1
1


reaction


10X Primer Mix
1
1
1
1


TIPP (2 U/μL)
0.25

0.25



Target RNA
2
2




Total
10
9.75
8
7.75









Briefly, in a first experimental example, Cas14a.1 (SEQ ID NO. 63) effector proteins were complexed with sgRNA for 30 minutes at 37° C. The 1× concentration of proteins was 40 nM and the final concentration of sgRNA was 40 nM. 1 uL of the Cas14a complexing reaction was combined with the HotPot components listed in Table 9 for each experimental or control condition listed. Reactions were carried out at 55° ° C. for 60 minutes. Trans cleavage activity was detected by fluorescence signal upon cleavage of a fluorophore-quencher reporter in the HotPot DETECTR reaction. FIG. 65 shows HotPot reaction results under the various conditions. The experimental condition, compared to control A without TIPP, had greater than 5-fold increase in the output fluorescence signal, while control A still had significantly higher signal than control D (no TIPP or target nucleic acid) and both conditions were able to detect the target nucleic acid. Control B without target RNA had about the same output fluorescence signal as control C without TIPP and without target RNA.


In a second experimental example, a 2:1 ratio mixture of unfunctionalized PEG (MW=600 monomers) and PEG-diacrylate (MW=700 monomers) were mixed together with a photoinitiator (2-Hydroxy-2-methylpropiophenone (Darocur 1173)) and 100 μM of Acrydite-modified “Rep172” reporter (/5Acryd/TTT TTT TTT TTT TTT TTT TT/16-FAMK//3Bio/(SEQ ID NO: 428)). The mixture was exposed to UV light (365 nm, 200 ms) under a photomask to generate circular cross-section rods of hydrogel containing immobilized reporters. Cas14a. 1 effector proteins were complexed with sgRNA for 30 minutes at 37° C. The 1× concentration of proteins was 40 nM and the final concentration of sgRNA was 40 nM. 5 uL of these RNPs was combined with the following components for a final volume of 50 uL (listed at final concentration): 10 uL of target RNaseP RNA (45 pg/uL) or no target control (“(−)”), reporter-immobilized hydrogels (10 hydrogels/uL), IB15 one pot LAMP trans-cleavage buffer, dNTPs (1 mM), RNAse inhibitor, Bsm DNA polymerase, Warmstart RTx reverse transcriptase, RNase P LAMP primer mix, and TIPP (0.5 U/uL) or water (“no TIPP”). Reactions were carried out at 55° C. for 35 minutes. Trans cleavage activity was detected with lateral flow assay strips. The supernatant for each reaction was applied to the sample pad of a lateral flow assay strip containing anti-FITC conjugate particles (colloidal gold). If trans cleavage occurred, the supernatant contained cleaved FAM-biotin-labeled reporter molecules which bound to an anti-biotin (e.g., streptavidin) target line on the lateral flow strip. The anti-FITC conjugate particles bound the FAM moiety on the reporter molecules and a target band appeared on lateral flow strips at the anti-biotin target line. If trans cleavage did not occur (as in NTC or no guide RNA reactions), the supernatant did not contain any FAM-biotin-labeled molecules, and nothing bound to the anti-biotin target line. The lateral flow assay strip also contained an anti-IgG flow control line, downstream of the anti-biotin target line, which bound to the anti-FITC moiety of the conjugate particles to confirm that the lateral flow assay functioned properly. Lateral flow strips without TIPP are shown in FIG. 66A and lateral flow strips with TIPP are shown in FIG. 66B. The presence of target RNA is indicated with (+) and the absence of target RNA is indicated with (−). As in the case with a fluorescent signal output, lateral flow assay signal was improved by the addition of TIPP.


Without being bound to any particular theory, it is believed that TIPP may be responsible for processing phosphates which are produced when the LAMP reaction uses up dNTPs. Those phosphates can take Mg2+ ions out of solution, which may be cause a Cas enzyme to exhaust the remaining Mg2+ quickly and cause the reaction to plateau more rapidly than would occur with additional Mg2+ in the system. With the presence of TIPP, those phosphates may be processed and the Mg2+ may be released for use by a Cas enzyme, which can improve DETECTR signal strength as the Cas enzyme is able to react unhindered by a lack of Mg2+.


The results show that the addition of TIPP improved signal compared to without TIPP and made detection on lateral flow strips more robust. The amount of TIPP added can be varied to allow flexibility in assay designs.


Example 19
SARS-COV-2 Detection Kits

In some embodiments, the compositions herein can comprise a kit for use in detecting SARS-COV-2 from samples. In some embodiments, the methods and compositions herein may be formulated to overcome the challenges in SARS detection. For example, a challenge in detecting SARS viruses is that the virus may be present in low concentrations. For example, a buccal swab from a SARS infected patient may only comprise a couple hundred copies of the SARS genome. If such a sample is stored prior to testing, the genome copy number may be less than 100. Further augmenting the challenge of detecting SARS is the inherent instability of SARS RNA, with half-lives on the order of minutes. Thus, a number of major technical challenges have to be overcome to formulate a kit for generalized, celeritous, and facile SARS detection.


In some embodiments, the kit provides reagents for RNA amplification and detection. Given that SARS is often present in low titers, the kit provides reagents for isothermal SARS RNA amplification, split among four vials. In some embodiments, the first three vials contain reverse transcription loop-mediated isothermal amplification (RT-LAMP) reagents targeting the SARS-COV-2 N-gene, the SARS-COV-2 E-gene, and human RNase P POP 7. In some embodiments, the SARS-COV-2 E- and N-genes may comprise a sequence with at least 80% sequence similarity to any one of SEQ ID NOs: 179-184, presented in TABLE 16. The SARS-CoV-2 E- and N-genes serve as two targets for the assay, while the human RNase P POP 7 gene (GGAGTATTGAATAGTTGGGAATTGGAACCCCTCCAGGGGGAACCAAACATTGTCGT TCAGAAGAAGACAAAGAGAGATTGAAATGAAGCTGTTGATTTCAACACACAAATTC TGGTGGTAGATGAAAGCAAAGCAAGTAAGTTTCTCCGAATCCCTAGTCAACTGGAG GTAGAGACGGACTGCGCAGGTTAACTACAGCTCCCAGCATGCCTGAGGGGGGGGCT CAGCGGCTGCGCAGACTGGCGCGCGCGGACGGTCATGGGACTTCAGCATGGCGGTG TTTGCAGATTTGGACCTGCGAGCGGGTTCTGACCTGAAGGCTCTGCGCGGACTTGTG GAGACAGCCGCTCACCTTGGCTATTCAGTTGTTGCTATCAATCATATCGTTGACTTTA AGGAAAAGAAACAGGAAATTGAAAAACCAGTAGCTGTTTCTGAACTCTTCACAACT TTGCCAATTGTACAGGGAAAATCAAGACCAATTAAAATTTTAACTAGATTAACAATT ATTGTCTCGGATCCATCTCACTGCAATGTTTTGAGAGCAACTTCTTCAAGGGCCCGG CTCTATGATGTTGTTGCAGTTTTTCCAAAGACAGAAAAGCTTTTTCATATTGCTTGCA CACATTTAGATGTGGATTTAGTCTGCATAACTGTAACAGAGAAACTACCATTTTACT TCAAAAGACCTCCTATTAATGTGGCGATTGACCGAGGCCTGGCTTTTGAACTTGTCT ATAGCCCTGCTATCAAAGACTCCACAATGAGAAGGTATACAATTTCCAGTGCCCTCA ATTTGATGCAAATCTGCAAAGGAAAGAATGTAATTATATCTAGTGCTGCAGAAAGG CCTTTAGAAATAAGAGGGCCATATGACGTGGCAAATCTAGGCTTGCTGTTTGGGCTC TCTGAAAGTGACGCCAAGGCTGCGGTGTCCACCAACTGCCGAGCAGCGCTTCTCCAT GGAGAAACTAGAAAAACTGCTTTTGGAATTATCTCTACAGTGAAGAAACCTCGGCC ATCAGAAGGAGATGAAGATTGTCTTCCAGCTTCCAAGAAAGCCAAGTGTGAGGGCT GAAAAGAATGCCCCAGTCTCTGTCAGCACTCCCTTCTTCCCTTTTATAGTTCATCAGC CACAACAAAAATAAAACCTTTGTGTGATTTACTGTTTTCATTTGGAGCTAGAAATCA ATAGTCTATAAAAACAGTTTTACTTGCAATCCATTAAAACAACAAACGAAACCTAGT GAAGCATCTTTTTAAAAGGCTGCCAGCTTAATGAATTTAGATGTACTTTAAGAGAGA AAGACTGGTTATTTCTCCTTTGTGTAAGTGATAAACAACAGCAAATATACTTGAATA AAATGTTTCAGGTATTTTTGTTTCATTTTGTTTTTGAGATAGGGTCTTTGTTGCTCAGG CTGGAGTACAGTGGCATAATCACAGCTCACTGCAACCTCAATCCTGGGCTCAAGTGA TCCTCCCGCTTCAGCCTCTCAAGCAGCGGGAACTACAGGTGTGCACTACCACACCTG GCTATTTTTTTTTTTTTTTTTTTTTTCCCTTGTAGAGACATGGTCTCACTATGTTGCTGA GGCTGGTCTCAAACTCCTAGGATCAAGCCATCCTCCCGCTTTGGCCTCCTAAAGTGC TGGGATTACATGAGCCACCACATGCAGCCAGATGTTTGAATATTTTAAGAGCTTCTT TCGAAAGTTTCTTGTTCATACTCAAATAGTAGTTATTTTGAAGATATTCAAACTTATA TTGAAGAAGTGACTTTAGTTCCTCTTGTTTTAAGCTTCTTTCATGTATTCAAATCAGC ATTTTTTTCTAAGAAATTGCTATAGAATTTGTGGAAGGAGAGAGGATACACATGTAA AATTACATCTGGTCTCTTCCTTCACTGCTTCATGCCTACGTAAGGTCTTTGAAATAGG ATTCCTTACTTTTAGTTAGAAACCCCTAAAACGCTAATATTGATTTTCCTGATAGCTG TATTAAAAATAGCAAAGCATCGGACTGA, SEQ ID NO: 220) serves as an internal control. In some embodiments, the buffer in the three amplification reagent vials is pH 8.8 Tris HCl containing (NH4)2SO4, KCl, MgSO4 and Tween-20 detergent. In some embodiments, the buffers are produced with nuclease-free water mixed with RNase inhibitor to prevent target nucleic acid degradation. In some cases, the three RT-LAMP reagent vials contain all of the reagents required for RT-LAMP, including dNTPs, a reverse transcriptase, and a polymerase. In some embodiments, the RT-LAMP reagent vials lack a reverse-transcriptase activator, and may not perform RT-LAMP until mixed with contents from the fourth vial, which contains an RT-LAMP activator mix consisting of 100 mM MgSO4. The contents of the four amplification reagent vials are summarized in TABLE 13.









TABLE 13







RT-LAMP Vial Constituents












SEQ ID
Concentration


Vial
Constituent
NO
in Vial













Vial 1,
PRIMER:
300
  0.26 μM


SARS-CoV-
M1957 F3 2019-nCoV-N-set1 -




2 RT-LAMP
AACACAAGCTTTCGGCAG




Master Mix
PRIMER
301
  0.26 μM


1 - N Gene
M1958 B3 2019-nCoV-N-set1 -





GAAATTTGGATCTTTGTCATCC





PRIMER
302
  2.12 μM



M1959 FIP 2019-nCoV-N-set1 -





TGCGGCCAATGTTTGTAATCAGCCA





AGGAAATTTTGGGGAC





PRIMER
303
  2.12 μM



M1960 BIP 2019-nCoV-N-set1 -





CGCATTGGCATGGAAGTCACTTTGA





TGGCACCTGTGTAG





PRIMER
304
  1.06 μM



M1961 LF 2019-nCoV-N-set1 -





TTCCTTGTCTGATTAGTTC





PRIMER
305
  1.06 μM



M1962 LB 2019-nCoV-N-set1 -





ACCTTCGGGAACGTGGTT





dNTPs

  1.85 mM



Bst 2.0 Polymerase

  1.06 units/μl



Warmstart RTx

  0.75 units/μl



RNase Inhibitor

  1.06 units/μl



Tris HC1 pH 8.8

 26.49 mM



(NH4)2SO4

 13.25 mM



KC1

 66.23 mM



MgSO4

  2.65 mM



Tween 20

  0.13% (v/v)





Vial 2,
PRIMER -
306
  0.26 μM


SARS-CoV-
M2106 F3 2019-nCoV-E-set13




2 RT-LAMP
CCGACGACGACTACTAGC




Master Mix
PRIMER -
307
  0.26 μM


2 - E Gene
M2107 B3 2019-nCoV-E-set13





AGAGTAAACGTAAAAAGAAGGTT





PRIMER -
308
  2.12 μM



M2108 FIP 2019-nCoV-E-set13





ACCTGTCTCTTCCGAAACGAATTTGT





AAGCACAAGCTGATG





PRIMER -
309
  2.12 μM



M2109 BIP 2019-nCoV-E-set13





CTAGCCATCCTTACTGCGCTACTCAC





GTTAACAATATTGCA





PRIMER -
310
  1.06 μM



M2110 LF 2019-nCoV-E-set13





TCGATTGTGTGCGTACTGC





PRIMER
311
  1.06 μM



M2111 LB 2019-nCoV-E-set13





TGAGTACATAAGTTCGTAC





dNTPs

  1.85 mM



Bst 2.0 Polymerase

  1.06 units/μl



Warmstart RTx

  0.75 units/μl



RNase Inhibitor

  1.06 units/μl



Tris HCl pH 8.8

 26.49 mM



(NH4)2SO4

 13.25 mM



KCI

 66.23 mM



MgSO4

  2.65 mM



Tween 20

  0.13% (v/v)





Vial, SARS-
PRIMER -
312
  0.26 μM


CoV-2
JPB RNase P POP7 F3




Control RT-
TTGATGAGCTGGAGCCA




LAMP
PRIMER -
313
  0.26 μM


Master Mix
JPB RNase P POP7 B3




3 - RNase
CACCCTCAATGCAGAGTC




POP P 7
PRIMER -
314
  2.12 μM



JPB RNase P POP7 FIP





GTGTGACCCTGAAGACTCGGTTTTA





GCCACTGACTCGGATC





PRIMER -
315
  2.12 μM



JPB RNase P POP7 BIP





CCTCCGTGATATGGCTCTTCGTTTTT





TTCTTACATGGCTCTGGTC





PRIMER -
316
  1.06 μM



JPB RNase P POP7 LF





ATGTGGATGGCTGAGTTGTT





PRIMER -
317
  1.06 μM



JPB RNase P POP7 LB





CATGCTGAGTACTGGACCTC





dNTPs

  1.85 mM



Bst 2.0 Polymerase

  1.06 units/μl



Warmstart RTx

  0.75 units/μl



RNase Inhibitor

  1.06 units/μl



Tris HCl pH 8.8

 26.49 mM



(NH4)2SO4

 13.25 mM



KC1

 66.23 mM



MgSO4

  2.65 mM



Tween 20

  0.13% (v/v)





SARS-CoV-
Bulk 100 mM MgSO4

100 mM


2 Amp





Activator









In some embodiments, a kit for detecting a target nucleic acid (e.g., a SARS-COV-2 nucleic acid) comprises a support medium (e.g., a 384-well block), optionally a reverse transcriptase (e.g., in cases where the target nucleic acid is an RNA molecule), a polymerase (e.g., Bst 2.0 polymerase), a primer (e.g., a primer that targets or is upstream from the target nucleic acid sequence), a guide nucleic acid targeting a sequence from the target nucleic acid, a programmable nuclease (e.g., a programmable nuclease with SEQ ID NO: 18), and a reporter (e.g., a reporter nucleic acid comprising a fluorophore-quencher pair and SEQ ID NO: 9). In some embodiments, the primer may comprise a sequence selected from among the group consisting of SEQ ID NO: 300-317. The guide nucleic acid may comprise a sequence selected from among the group consisting of SEQ ID NO: 318-SEQ ID NO: 327 presented in TABLE 15. The guide nucleic acid may comprise a sequence comprising at least 80% identity to any one of SEQ ID NO: 318-SEQ ID NO: 327. In some embodiments, the target gene for detection of coronaviruses may comprise a sequence of any one of SEQ ID NOs: 179-184 listed in TABLE 16.


For detection, the kit contains DETECTR reagents split between three vials. Each vial contains SEQ ID NO: 18 and a reporter containing the dye-quencher pair Alexa Fluor 594-3′ Iowa Black® RQ. The three vials contain separate guide nucleic acids targeting the SARS-COV-2 N-gene, the SARS-COV-2 E-gene, or the human RNase gene POP P 7. The constituents in each vial are dissolved in pH 7.5 HEPES buffer containing potassium acetate, magnesium acetate, glycerol and the detergent Triton-X. The compositions of the DETECTR vials are summarized in TABLE 14.









TABLE 14







DETECTR Vial Constituents












SEQ ID
Concentration


Vial
Constituent
NO
in Vial





Vial 5,
R1763 CDC-N2-Wuhan Guide RNA
318
 44.4 nM


SARS-CoV-
UAAUUUCUACUAAGUGUAGAUCCCCC




2 DETECTR
AGCGCUUCAGCGUUC




Master Mix 1 -
Bulk, SEQ ID NO: 18

 44.4 nM


N Gene
HEPES pH 7.5

  0.5 M



Potassium Acetate

  2.2 mM



Magnesium Acetate

  5.6 mM



Glycerol

  1.1%



Triton-X-100

  0.00018%



Reporter Nucleic Acid - rep33

111.11 nM



/AF594/SEQ ID NO: 9/3IAbRQSp/







Vial 6,
Vial, SARS-CoV-2 DETECTR Master Mix 2 -
319
 44.4 nM


SARS-CoV-
E Gene




2 DETECTR
UAAUUUCUACUAAGUGUAGAUGUGG




Master Mix 2 -
UAUUCUUGCUAGUUAC




E-Gene
Bulk, SEQ ID NO: 18

 44.4 nM



HEPES pH 7.5

  0.5 M



Potassium Acetate

  2.2 mM



Magnesium Acetate

  5.6 mM



Glycerol

  1.1%



Triton-X-100

  0.00018%



Reporter - rep33

111.11 nM



/AF594/TTATTATT/3IAbRQSp/







Vial 7,
Vial, SARS-CoV-2 DETECTR Master Mix 2 -
320
 44.4 nM


SARS-CoV-
RNAse P




2 DETECTR
UAAUUUCUACUAAGUGUAGAUAAUU




Master Mix -
ACUUGGGUGUGACCCU




RNase POP
Bulk, SEQ ID NO: 18

 44.4 nM


P 7
HEPES pH 7.5

  0.5 M



Potassium Acetate

  2.2 mM



Magnesium Acetate

  5.6 mM



Glycerol

  1.1%



Triton-X-100

  0.00018%



Reporter - rep33

111.11 nM



/AF594/TTATTATT/3IAbRQSp/









In some embodiments, the kit also contains the equipment needed to perform the SARS-COV-2 detection assay. In some embodiments, the kit provides a 96 well plate for the RT-LAMP reactions and a 384 well plate for the DETECTR reactions, as well as films for sealing the well plates (ultra-Clear Polyester Heat-Resistant Films). In some embodiments, the kit further provides a set of pipettes and pipette tips for reagent transfer during the assay.


In some embodiments, as an internal control, the kit provides a vial containing genomic DNA comprising the human RNAse POP P 7 gene. In some embodiments, a control can be run in parallel using this DNA sample the kit is used to assay for SARS-COV-2. In some embodiments, the kit thus allows a single user to perform a large number of parallel assays for SARS-COV-2 genomic markers.


Example 20
Assay for Detecting SARS-COV-2

In some embodiments, the methods and compositions herein outline an assay for using the kit from example 18 to detect SARS-COV-2 from a sample. In some embodiments, the assay involves an initial RT-LAMP reaction that produces DNA amplicons from SARS genomic RNA, followed by a DETECTR reaction to determine whether SARS-Cov-2 was present in the assayed sample, to overcome the challenge of a low concentration of the SARS virus. In some embodiments, the assay also involves RT-LAMP and DETECTR on a control sample to enable easier visualization of the SARS DETECTR reaction results.


In some embodiments, the kit can be stably stored by −25° C. and −15° C., and may need to be thawed at room temperature for 30 minutes prior to use. In some embodiments, once the reagents have thawed, the amplification master mixes are prepared by mixing 13 μl aliquots of Amp-A (vial 4) into 13 μl aliquots of Amp-N (vial 1), Amp-E (vial 2), and Amp-RP (vial 3). In some embodiments, the amplification master mixes are vortexed and spun, and then dispensed in 8 μl portions into individual wells in the 96- or 384-well plates. In some embodiments, RNAs from the biological samples are extracted using spin columns, beads or just crude extractions and 2 μl of the extracted sample is added to each well containing amplification reagents, and the well plates are sealed with film, spun down, and then heated to 62° C. for 30 minutes to enable the RT-LAMP reactions.


In some embodiments, the products of the RT-LAMP reactions are then transferred to a separate well plate pre-loaded with DETECTR mix. Each well is loaded with 18 μl DETECTR reagents from one of vials 5, 6 or 7 (targeting the SARS-COV-2 N-gene, the SARS-Cov-2 E-gene, and RNase POP P 7, respectively) and 2 μl amplification products. In some embodiments, the plate is then sealed with film, spun down for 30 s at 2000 rcf, and heated to 37° C. to enable the DETECTR reaction. In some embodiments, the progress of the DETECTR reactions can be monitored with a commercial fluorimeter (e.g., with a ThermoFisher QS5).









TABLE 15







Exemplary gRNA Sequences for Detection of Coronaviruses










SEQ ID NO:
gRNA
Target
Sequence





SEQ ID NO: 318
R1763
CDC-N2-Wuhan
UAAUUUCUACUAAGUGUAGAUCCCC





CAGCGCUUCAGCGUUC





SEQ ID NO: 321
R1764
E-Sarbeco-1
UAAUUUCUACUAAGUGUAGAUUUGC





UUUCGUGGUAUUCUUG





SEQ ID NO: 319
R1765
E-Sarbeco-2
UAAUUUCUACUAAGUGUAGAUGUGG





UAUUCUUGCUAGUUAC





SEQ ID NO: 322
R1766
CDC-N2-SARS
UAAUUUCUACUAAGUGUAGAUCUCC





AAGUGCCUCUGCAUUC





SEQ ID NO: 323
R1767
N-Sarbeco-1
UAAUUUCUACUAAGUGUAGAUGCAA





UGUUGUUCCUUGAGGA





SEQ ID NO: 324
R1768
ORF1ab-Wuhan
UAAUUUCUACUAAGUGUAGAUCACA





UACCGCAGACGGUACA





SEQ ID NO: 325
R1769
CDC-RNaseP
UAAUUUCUACUAAGUGUAGAUGACC





UGCGAGCGGGUUCUGA





SEQ ID NO: 320
R779
RNaseP POP7
UAAUUUCUACUAAGUGUAGAUAAUU





ACUUGGGUGUGACCCU





SEQ ID NO: 326
R1965
RNaseP POP7 v2
UAAUUUCUACUAAGUGUAGAUUUAC





AUGGCUCUGGUCCGAG





SEQ ID NO: 327
R780
RNaseP POP7 v3
UAAUUUCUACUAAGUGUAGAUGGCT





TCCAGGGAACAGGCCT
















TABLE 16







Exemplary Coronavirus N-Gene and E-Gene Gene Fragments









SEQ ID NO:
Target
Sequence





SEQ ID NO:
2019-
CCAAATTGGCTACTACCGAAGAGCTACCAGACGAATTCGTGGTGG


179
nCoV
TGACGGTAAAATGAAAGATCTCAGTCCAAGATGGTATTTCTACTA



N-gene
CCTAGGAACTGGGCCAGAAGCTGGACTTCCCTATGGTGCTAACAA




AGACGGCATCATATGGGTTGCAACTGAGGGAGCCTTGAATACAC




CAAAAGATCACATTGGCACCCGCAATCCTGCTAACAATGCTGCAA




TCGTGCTACAACTTCCTCAAGGAACAACATTGCCAAAAGGCTTCT




ACGCAGAAGGGAGCAGAGGCGGCAGTCAAGCCTCTTCTCGTTCCT




CATCACGTAGTCGCAACAGTTCAAGAAATTCAACTCCAGGCAGCA




GTAGGGGAACTTCTCCTGCTAGAATGGCTGGCAATGGCGGTGATG




CTGCTCTTGCTTTGCTGCTGCTTGACAGATTGAACCAGCTTGAGA




GCAAAATGTCTGGTAAAGGCCAACAACAACAAGGCCAAACTGTC




ACTAAGAAATCTGCTGCTGAGGCTTCTAAGAAGCCTCGGCAAAA




ACGTACTGCCACTAAAGCATACAATGTAACACAAGCTTTCGGCAG




ACGTGGTCCAGAACAAACCCAAGGAAATTTTGGGGACCAGGAAC




TAATCAGACAAGGAACTGATTACAAACATTGGCCGCAAATTGCA




CAATTTGCCCCCAGCGCTTCAGCGTTCTTCGGAATGTCGCGCATT




GGCATGGAAGTCACACCTTCGGGAACGTGGTTGACCTACACAGGT




GCCATCAAATTGGATGACAAAGATCCAAATTTCAAAGATCAAGTC




ATTTTGCTGAATAAGCATATTGACGCATACAAAACATTCCCACCA




ACAGAGCCTAAAAAGGACAAAAAGAAGAAGGCTGATGAAACTC




AAGCCTTACCGCAGAGACAGAAGAAACAGCAAACTGTG





SEQ ID NO:
SARS-
CCAAATTGGCTACTACCGAAGAGCTACCCGACGAGTTCGTGGTGG


180
CoV N-
TGACGGCAAAATGAAAGAGCTCAGCCCCAGATGGTACTTCTATTA



gene
CCTAGGAACTGGCCCAGAAGCTTCACTTCCCTACGGCGCTAACAA




AGAAGGCATCGTATGGGTTGCAACTGAGGGAGCCTTGAATACAC




CCAAAGACCACATTGGCACCCGCAATCCTAATAACAATGCTGCCA




CCGTGCTACAACTTCCTCAAGGAACAACATTGCCAAAAGGCTTCT




ACGCAGAGGGAAGCAGAGGCGGCAGTCAAGCCTCTTCTCGCTCC




TCATCACGTAGTCGCGGTAATTCAAGAAATTCAACTCCTGGCAGC




AGTAGGGGAAATTCTCCTGCTCGAATGGCTAGCGGAGGTGGTGA




AACTGCCCTCGCGCTATTGCTGCTAGACAGATTGAACCAGCTTGA




GAGCAAAGTTTCTGGTAAAGGCCAACAACAACAAGGCCAAACTG




TCACTAAGAAATCTGCTGCTGAGGCATCTAAAAAGCCTCGCCAAA




AACGTACTGCCACAAAACAGTACAACGTCACTCAAGCATTTGGG




AGACGTGGTCCAGAACAAACCCAAGGAAATTTCGGGGACCAAGA




CCTAATCAGACAAGGAACTGATTACAAACATTGGCCGCAAATTGC




ACAATTTGCTCCAAGTGCCTCTGCATTCTTTGGAATGTCACGCATT




GGCATGGAAGTCACACCTTCGGGAACATGGCTGACTTATCATGGA




GCCATTAAATTGGATGACAAAGATCCACAATTCAAAGACAACGT




CATACTGCTGAACAAGCACATTGACGCATACAAAACATTCCCACC




AACAGAGCCTAAAAAGGACAAAAAGAAAAAGACTGATGAAGCT




CAGCCTTTGCCGCAGAGACAAAAGAAGCAGCCCACTGTG





SEQ ID NO:
bat-SL-
CCAAATTGGCTACTACCGTAGAGCTACCAGACGAATTCGTGGTGG


181
CoVZC
TGACGGTAAAATGAAAGAGCTCAGCCCCAGATGGTATTTTTACTA



45 N-
TCTAGGAACTGGACCAGAAGCTGGACTTCCCTATGGTGCTAACAA



gene
AGAAGGCATCATATGGGTTGCAACTGAGGGAGCCTTAAACACAC




CGAAAGACCACATTGGCACCCGCAATCCTGCTAACAATGCTGCAA




TCGTGCTACAACTTCCTCAAGGAACAACATTGCCAAAAGGCTTCT




ACGCAGAAGGGAGCAGAGGCGGCAGTCAAGCTTCTTCACGCTCC




TCATCACGTAGTCGCAACAGTTCAAGAAACTCAACTCCAGGCAGC




AGTAGGGGAACTTCTCCTGCTAGAATGGCTGGCAATGGCGGTGAC




ACTGCTCTTGCTTTGCTGCTGCTAGATAGGTTGAACCAGCTTGAG




AACAAAGTATCTGGCAAAGGCCAACAACAACAGGGCCAAACTGT




CACTAAGAAATCTGCTGCTGAGGCATCTAAAAAGCCTCGCCAAA




AACGTACTGCTACAAAACAGTACAACGTCACTCAAGCATTTGGGA




GACGTGGTCCAGAACAAACCCAAGGAAATTTTGGGGACCAAGAA




TTAATCAGACAAGGAACTGATTACAAACATTGGCCGCAAATTGCA




CAATTTGCTCCAAGTGCCTCTGCATTCTTTGGAATGTCACGCATTG




GCATGGAAGTCACACCTTCGGGAACATGGCTGACTTATCATGGAG




CCATTAAATTGGATGACAAAGATCCACAATTCAAAGATAACGTCA




TACTGCTGAATAAGCACATTGACGCATACAAAACATTCCCACCAA




CAGAGCCTAAAAAGGACAAAAAGAAAAAGGCTGATGAACTTCAG




GCTTTACCGCAGAGACAGAAGAAACAACAAACTGTG





SEQ ID NO:
2019-
ACTATTACCAGCTGTACTCAACTCAATTGAGTACAGACACTGGTG


182
nCoV
TTGAACATGTTACCTTCTTCATCTACAATAAAATTGTTGATGAGCC



E-gene
TGAAGAACATGTCCAAATTCACACAATCGACGGTTCATCCGGAGT




TGTTAATCCAGTAATGGAACCAATTTATGATGAACCGACGACGAC




TACTAGCGTGCCTTTGTAAGCACAAGCTGATGAGTACGAACTTAT




GTACTCATTCGTTTCGGAAGAGACAGGTACGTTAATAGTTAATAG




CGTACTTCTTTTTCTTGCTTTCGTGGTATTCTTGCTAGTTACACTAG




CCATCCTTACTGCGCTTCGATTGTGTGCGTACTGCTGCAATATTGT




TAACGTGAGTCTTGTAAAACCTTCTTTTTACGTTTACTCTCGTGTT




AAAAATCTGAATTCTTCTAGAGTTCCTGATCTTCTGGTCTAAACG




AACTAAATATTATATTAGTTTTTCTGTTTGGAACTTTAATTTTAGC




CATGGCAGATTCCAACGGTACTATTACCGTTGAAGAGCTTAAAAA




GCTCCTTGAACAATGGAACCTAGTAATAGGTTTCCTATTCCTTAC




ATGGATT





SEQ ID NO:
SARS-
TTTACTACCAGCTTGAGTCTACACAAATTACTACAGACACTGGTA


183
CoV E-
TTGAAAATGCTACATTCTTCATCTTTAACAAGCTTGTTAAAGACCC



gene
ACCGAATGTGCAAATACACACAATCGACGGCTCTTCAGGAGTTGC




TAATCCAGCAATGGATCCAATTTATGATGAGCCGACGACGACTAC




TAGCGTGCCTTTGTAAGCACAAGAAAGTGAGTACGAACTTATGTA




CTCATTCGTTTCGGAAGAAACAGGTACGTTAATAGTTAATAGCGT




ACTTCTTTTTCTTGCTTTCGTGGTATTCTTGCTAGTCACACTAGCC




ATCCTTACTGCGCTTCGATTGTGTGCGTACTGCTGCAATATTGTTA




ACGTGAGTTTAGTAAAACCAACGGTTTACGTCTACTCGCGTGTTA




AAAATCTGAACTCTTCTGAAGGAGTTCCTGATCTTCTGGTCTAAA




CGAACTAACTATTATTATTATTCTGTTTGGAACTTTAACATTGCTT




ATCATGGCAGACAACGGTACTATTACCGTTGAGGAGCTTAAACAA




CTCCTGGAACAATGGAACCTAGTAATAGGTTTCCTATTCCTAGCC




TGGATT





SEQ ID NO:
bat-SL-
ATTACTACCAGCTGTACTCAACACAAGTGAGTACAGACACTGGTG


184
CoVZC
TTGAACATGTTACTTTCTTCATCTACAATAAAATTGTTGATGAGCC



45 E-
TGAAGAACATGTTCAAATTCACACAATCGACGGTACATCTGGAGT



gene
TGTTAATCCAGCAATGGAACCAATTTATGATGAACCGACGACGAC




TACTAGCGTGCCTTTGTAAGCACAAGCTGATGAGTACGAACTTAT




GTACTCATTCGTTTCGGAAGAGACAGGTACGTTAATAGTTAATAG




CGTACTTCTTTTTCTTGCTTTTGTGGTATTCTTGCTAGTCACACTAG




CCATCCTTACTGCGCTTCGATTGTGTGCGTACTGCTGCAATATTGT




TAACGTGAGTCTTGTAAAACCTTCTTTTTACGTTTACTCTCGTGTT




AAAAATCTGAATTCTTCTAGAGTTCCTGATCTTTTGGTCTAAACGA




ACTAAATATTATATTAGTCTTTCTGTTTGGAACTTTAATTTTAGCC




ATGTCAGGTGACAACGGTACCATTACCGTTGAAGAGCTTAAAAA




GCTCTTAGAACAATGGAACCTAGTAATAGGATTCTTGTTTCTTAC




ATGGATT









Example 21
Assay Testing Using Bead Immobilized Reporters Following HotPot Protocol


FIG. 67 shows a drawing illustrating the manual HotPot experimental protocol used to test bead-immobilized reporter cleavage. A sample containing target nucleic acids was added to a tube containing lysis buffer (15201). After lysing 1-2 minutes at ambient temperature, the solution was transferred to a reaction tube (15202) containing lyophilized reagents (i.e., the base bead, the master mix bead, and the reporter bead). Contents of the reaction tube were rehydrated and reconstituted with lysis buffer, and the HotPot reaction was started and maintained at 55° C. for 30 minutes (15203). During the reaction, programmable nucleases in the solution cleaved reporter molecules from the beads at the same time as an RT-LAMP reaction proceeded to amplify the target nucleic acids (15204). The reaction medium was then filtered through a membrane to trap the beads and a first portion of the filtered product was used to measure fluorescence thereof on a fluorescence reader. A second portion of the filtered product was applied to a sample pad of a lateral flow strip. The lateral flow strip included a target capture area (T) comprising streptavidin and a control area (C) comprising IgG. The lateral flow strip assay was allowed to run for 3 minutes at ambient temperature (15205) before pictures were taken of the resulting bands at the target capture area T and the control area C.



FIG. 68 shows fluorescence DETECTR results with reporters immobilized onto glass beads. Experiments with both DNase and CasM.21526 (SEQ ID NO. 406)/R1763 showed larger fluorescence signal in the presence of target nucleic acids (2 nM, GF703) compared to the no target control experiments (NTC), thus the HotPot DETECTR reaction successfully cleaved the immobilized reporters from the glass beads. Experiments with CasM.21526 (SEQ ID NO. 406) were carried out at 55° C. with H2.B buffer. Experiments with DNase were carried out at 37° C. with 1× Turbo DNase buffer. FIG. 69 shows photographs of the lateral flow strips to which the DNase and CasM.21526 (SEQ ID NO. 406) samples from FIG. 68 were applied.



FIG. 70 shows results with maleimide-coated magnetic beads immobilized with thiol-FAM reporter. Experiments with each protein (a Cas14 variant (SEQ ID NO: 63), CasM.21526 (SEQ ID NO. 406), a Cas12 variant (SEQ ID NO: 28)) resulted in larger signals with target nucleic acids (GF703) compared to the no target control experiments (NTC), thus the HotPot DETECTR reaction successfully cleaved the immobilized reporters from the maleimide-coated magnetic beads.


Example 22
HotPot DETECTR-Based Assay for Respiratory Virus Targets


FIG. 71 shows the results HotPot DETECTR-based assays using the protocol shown in FIG. 67 with Cas14a. 1 (SEQ ID NO: 63) for multiple different respiratory virus target nucleic acids including SARS-COV-2, MS2, FluB, RSV-A, and RSV-B and fluorescence-based readout. Plots in the top row and the plots in the bottom row show the same data, with different y-axis scale. Each plot shows the raw fluorescence measured during the assay of unique nucleic acid sequences as a function of time. Depending on the target, the saturation signal strength varied from about 70000 AUs to 1000000 AUs, and the saturation time was reached in between about 20 to about 40 minutes (SARS-COV-2=25 min, MS2=35 min, FluB=20 min, RSV-A=25 min, RSV-B=40 min). Depending on the target, the signal strength from the HotPot DETECTR assay ranged from about 1× to about 25× the signal strength of the SARS-COV-2 reaction (SARS-COV-2=1×, MS2=1×, FluB=25×, RSV-A=0.9×, RSV-B=7×).


Example 23
HotPot Limit of Detection (LOD) Assay for SARS-COV-2 Target

The following describes experiments carried out to determine limit of detection for HotPot reactions using CasM.21526 (SEQ ID NO. 406). The HotPot DETECTR-based assays were run using the protocol shown in FIG. 67 and a fluorescence-based readout. FIG. 72A shows results of experiments carried out using IB15 buffer with different target concentrations ranging from 200 copies to 0 copies. Multiple replicates were conducted at each concentration, and the results of each replicate are shown as individual lines in the plots. FIG. 72B shows an aggregated plot of the data in FIG. 72A where the average and the error from the multiple replicates are plotted in a single plot. FIG. 72C shows the saturation fluorescence heatmap of the multiple replicates (from 1 to 7) of FIG. 72A at different target concentrations. Strong signal was produced at all target concentrations, with about 5 copies being the LOD for the experimental conditions tested.


Example 24
Additive Screening for SARS-COV-2 Target HotPot

These experiments were conducted to identify potential additives for HotPot assays using Cas14a.1 (SEQ ID NO: 63). 96 potential additives were identified and individually screened for their influence on the output fluorescence. FIG. 73 shows the fluorescence detected from the assays when using a target concentration of 200 input copies/reaction (left column) and when using a target concentration of 0 copies/reaction (i.e., no target, right column). The color intensity of the squares in the heatmap indicate the strength of the fluorescence detected.


Additives that were identified to increase the speed of the reaction under the conditions tested included: betaine monohydrate, acetamide, GABA, L-proline, beta-alanine, 6-aminohexanoic acid, urea, methylurea, ethylurea, hypotaurine, NDSB-256, and ammonium acetate.


Additives that were identified to increase the signal strength from the reaction under the conditions tested included: trehalose, xylitol, D-sorbitol, sucrose, and trimethylamine N-oxide dihydrate.



FIG. 74A shows the influence of some additives that increased the speed of the reaction under the conditions tested. The influence of each additive (e.g., betaine monohydrate, NDSB-256, and beta-alanine) is shown against control experiments carried out with added water (i.e., without additive). Experiments carried out with no target (i.e., 0 copies/reaction) showed negligible fluorescence signals. In each scenario, the presence of the additives reduced the amount of time it took for fluorescence signals to reach saturation point (i.e., a plateau in the plot).



FIG. 74B shows the influence of some additives that increased the signal strength of the reaction under the conditions tested. The influence of each additive (e.g., sucrose and xylitol) is shown against control experiments carried out with added water (i.e., without additive). Experiments carried out with no target (i.e., 0 copies/reaction) showed negligible fluorescence signal. In each scenario, the presence of the additives increased the overall fluorescence signal strength generated in the presence of targets.



FIG. 75 shows HotPot DETECTR results for various combinations of additives with 300 copies/reaction of Twist SC2 RNA or with no target controls (NTC). Experiments were conducted in the presence of different amounts of trimethylamine N-oxide dihydrate (TMAO, concentrations shown on top of columns), in combination with IB1 buffer (top row), IB13 (comparing IB1 buffer plus 1 mg/ml bovine serum albumin, middle row), or IB14 (comparing IB1 buffer plus 1 mg/ml bovine serum albumin and no Tween, bottom row). All conditions tested exhibited strong DETECTR signals, with 250 mM TMAO appearing to provide the strongest signal in all buffer conditions.


Further HotPot experiments were carried out to determine the effects of using glycerol free (GF) Bsm DNA polymerase and glycerol (G) containing Bsm DNA polymerase with or without 250 mM TMAO added. FIG. 76 shows that both G and GF Bsm DNA polymerases were able to output satisfactory fluorescence signals with each formulation.


Example 25

DETECTR-Based OnePot and HotPot Reactions Using Reporter Immobilization within Hydrogels


These experiments were carried out to synthesize hydrogels containing immobilized reporters co-polymerized with a mixture of oligomers as described in FIG. 78 and FIGS. 79A-79B and determine their applicability for OnePot and HotPot DETECTR assays. FIG. 78 illustrates the hydrogel structure with a covalently incorporated reporter that was generated via co-polymerization with the reporter.


Reporter was covalently incorporated into PEG hydrogels during polymerization. A 2:1 ratio mixture of unfunctionalized PEG (MW-600 monomers) and PEG-diacrylate (MW=700 monomers) were mixed together with a photoinitiator (2-Hydroxy-2-methylpropiophenone (Darocur 1173)) and 100 μM of Acrydite-modified Reporter 172 (/5Acryd/TTT TTT TTT TTT TTT TTT TT/16-FAMK//3Bio/(SEQ ID NO: 428)). The mixture was exposed to UV light (365 nm, 200 ms) under a photomask. The mask was configured to polymerize the mix into circular cross-sectional rods of hydrogel 400 μm in diameter. Excess material was washed off hydrogels after polymerization. The acrydite group on the 5′ end of the reporter was covalently reacted with the acrylate groups of PEG-diacrylate oligomers during co-polymerization in order to incorporate the reporter into the hydrogel.


OnePot (using a Cas12 enzyme, SEQ ID NO: 28) and HotPot (using Cas14a. 1, SEQ ID NO: 63) DETECTR reactions were run as described herein by applying the programmable nuclease complexes and target nucleic acids to a tube containing the hydrogels. 6 hydrogels/reaction were added for OnePot DETECTR and 10 hydrogels/reaction for Cas14a.1 HotPot DETECTR assays. DETECTR reactions were run for 60 min at 37° C. with mixing for Cas12 OnePot or 60 min at 55° C. with mixing for Cas14a.1 HotPot. Duplicate reactions were run for each of a target RNA and the NTC for both Cas12 OnePot and Cas14a. 1 HotPot.


The tubes were then spun down and the supernatant was applied to lateral flow strips. The sample pad of lateral flow strip contained anti-FITC conjugate particles (colloidal gold). If target was present, the supernatant contained cleaved FAM-biotin-labeled reporter molecules which bound to an anti-biotin (e.g., streptavidin) target line on the lateral flow strip. The anti-FITC conjugate particles bound the FAM moiety on the reporter molecules and a target band appeared on lateral flow strips at the anti-biotin target line. If target was not present (as in NTC DETECTR reactions), the supernatant did not contain any FAM-biotin-labeled molecules and nothing bound to the anti-biotin target line. The lateral flow assay strip also contained an anti-IgG flow control line, downstream of the anti-biotin target line, which bound to the anti-FITC moiety of the conjugate particles to confirm that the lateral flow assay functioned properly. FIG. 77A shows the results of the Cas12 OnePot DETECTR assays. FIG. 77B shows the results of the Cas14a.1 HotPot DETECTR assays. Strong signals were seen in both positive sample replicates while minimal background appeared in NTC replicate strips at the target line.


Example 26
Isothermal Nicking Enzyme Amplification for DETECTR Reactions

This example describes isothermal nicking enzyme pre-amplification for DETECTR reactions. CRISPR-based diagnostic DETECTR reactions using loop-mediated isothermal amplification (LAMP) can be completed in as little as 30 minutes. In some cases, longer LAMP amplification may be required to achieve single copy sensitivity. A DETECTR reaction utilizing nicking enzyme amplification (NEAR) was developed to reduce amplification time relative to methods utilizing LAMP amplification. The DETECTR reaction with NEAR also showed improved sensitivity over NEAR reactions performed with a beacon. In the DETECTR reaction with NEAR, a 30 to 40 nucleotide region of a target nucleic acid was amplified using NEAR and detected using a Cas12 variant (SEQ ID NO: 28).


NEAR amplification was performed by contacting the target nucleic acid with a forward primer, a reverse primer, dNTPs, a DNA polymerase, and a nicking endonuclease. The reaction was performed at 60° C. for 10 minutes then cooled to 4° C. The forward primer contained a 16 to 20 nucleotide region reverse complementary to a first strand of the target nucleic acid and a 16 to 20 nucleotide nicking enzyme stabilization, binding, and recognition site 5′ of the region reverse complementary to the target nucleic acid. The reverse primer contained a 16 to 20 nucleotide region reverse complementary to a second strand of the target nucleic acid and a 16 to 20 nucleotide nicking enzyme stabilization, binding, and recognition site 5′ of the region reverse complementary to the target nucleic acid. A NEAR duplex incorporating a nicking enzyme stabilization, binding, and recognition site into an amplicon also containing the region of the target nucleic acid was then generated, as illustrated in FIG. 80. The duplex was then nicked by the nicking endonuclease and amplified using the DNA polymerase. The amplicons were detected using a DETECTR reaction with the Cas12 variant as described in at least Examples 30-33.


Example 27

Detection of a Strep-A Target Nucleic Acid Using DETECTR with NEAR


This example describes detection of Streptococcus pyogenes using NECTR (NEAR combined with DETECTR) via amplification of the Strep-A target. The Strep-A target was amplified using the process described in Example 26. Specific conditions were optimized for Strep-A detection.


A 10 μL preamplification reaction was prepared at a final concentration of 1× Isothermal Amplification Buffer (NEB), 1×NEBuffer 3.1 (NEB), 300 nM dNTPs (NEB), 4 U of Bst 2.0, 3 U of Nt BstNBI, 500 nM of the forward primer (SEQ ID NO: 359), and 100 nM of the reverse primer (SEQ ID NO: 360). In order to prevent non-specific amplification, the primers were added last.


8 μL of the prepared pre-amplification mixture was dispensed into a 96-well or 384-well plate. 2 μL of the target nucleic acid was added to each well in order to prevent early initiation of the reaction. Sample mixtures are mixed thoroughly and centrifuged to ensure that mixture is at the bottom of the wells. The pre-amplification reactions were incubated at 60° C. for 10 minutes and immediately placed at 4° C. or on ice to stop the reaction. Detection of the Strep-A was achieved by our DETECTR technology via Cas12a. A Cas12a complexing reaction was prepared at a final concentration of 1× MB3, 160 nM of Cas12 variant (SEQ ID NO: 28) and 160 nM of R1107 crRNA (SEQ ID NO: 358) and incubated at 37° C. for 30 minutes. After incubation, the Beacon-AlexaFluor 594 (SEQ ID NO: 357) was added to the complexing reaction at a final concentration of 400 nM.


In a 384-well black assay plate on ice, 13 μL of the 1× MB3 and 5 μL of the Cas12a complexing mixture was added. Using aseptic technique to prevent any nucleic acid contamination, 2 L of the pre-amplification reaction was added to the assay plate. The plate was sealed with an optically clear adhesive and spun at 2000 rcf. The plate was read at AF594 setting with extended gain. FIGS. 81A-81B, 84-85, and 91 show results obtained using variations of the Strep-A method described here.









TABLE 17







Nucleic acid sequences for NEAR reaction for


Strep-A and SARS-CoV-2 Target Nucleic Acid.









SEQ ID




NO
Name
Sequence





SEQ ID
Beacon
ACAAGTATGTGAGGAGAGGCCATACTTGT


NO: 357







SEQ ID

UAAUUUCUACUAAGUGUAGAUUAGGUCAACAUGCA


NO: 358

GCUACA





SEQ ID

GCCACCCCAAAAAUGAAGGGGACUAAAACAGGUCA


NO: 359

ACAUGCAGCUACAGG





SEQ ID

GACGAAUGAAGGAAUGCAACUAGGUCAACAUGCAG


NO: 360

CUACA





SEQ ID

GACGAAUGAAGGAAUGCAACGUAGGUCAACAUGCA


NO: 361

GCUAC





SEQ ID

TTGCTTTCGTGGTATT


NO: 362







SEQ ID

GGATGGCTAGTGTAA


NO: 363









Example 28

Detection of a SARS-COV-2 Target Nucleic Acid Using DETECTR with NEAR


This example describes detection of SARS-COV-2 using NECTR (NEAR combined with DETECTR) via amplification of the E-gene target. The Strep-A target was amplified using the process described in Example 26. Specific conditions were optimized for E-gene detection.


A 10 μL preamplification reaction was prepared at a final concentration of 1× Isothermal Amplification Buffer (NEB), 1×NEBuffer 3.1 (NEB), 300 nM dNTPs (NEB), 4 U of Bst 2.0, 3 U of Nt BstNBI, 500 nM of the Forward primer (SEQ ID NO: 362), and 100 nM of the reverse primer (SEQ ID NO: 363). In order to prevent non-specific amplification, the primers were added last.


8 μL of the prepared pre-amplification mixture was dispensed into a 96-well or 384-well plate. 2 μL of the target nucleic acid was added to each well in order to prevent early initiation of the reaction. Sample mixtures are mixed thoroughly and centrifuged to ensure that mixture is at the bottom of the wells. The pre-amplification reactions were incubated at 60° C. for 10 minutes and immediately placed at 4° C. or on ice to stop the reaction.


Detection of the Strep-A was achieved by our DETECTR technology via Cas12a. A Cas12a complexing reaction was prepared at a final concentration of 1× MB3, 160 nM of a Cas12 variant (SEQ ID NO: 28), and 160 nM of R1107 crRNA (SEQ ID NO: 358) and incubated at 37° C. for 30 minutes. After incubation, the Beacon-AlexaFluor 594 (SEQ ID NO: 361) was added to the complexing reaction at a final concentration of 400 nM.


In a 384-well black assay plate on ice, 13 μL of the 1× MBuffer 3 and 5 μL of the Cas12a complexing mixture was added. Using aseptic technique to prevent any nucleic acid contamination, 2 μL of the pre-amplification reaction was added to the assay plate. The plate was sealed with an optically clear adhesive and spun at 2000×g rcf. The plate was read at AF594 setting with extended gain. FIGS. 82A-82B and 86A-90 show results obtained using variations of the SARS-COV-2 E-gene method described here.


Example 29
NEAR+DETECTR Reaction Optimization
Determining the Mg2+ Concentration for a NEAR Reaction

This example describes optimization of Mg2+ concentration for the NEAR reaction. In addition, the performance of Bst 2.0 and Bst 3.0 were assessed. In this example, the protocol used was identical to that used in Example 26.


After amplification of Strep-A, the samples were tested in different buffers, Thrmopol, IsoAmp I, and IsoAmp II at different added Mg2+ concentrations—0 mM, 2.5 mM, 5 mM, 7.5 mM, and 10 mM. The elapsed time at which it took to achieve a fluorescent readout from the Beacon plate reader was measured, with a lower value indicating that there were more copies of the amplicon present in the solution.


It was found that no additional magnesium was required and that Bst 2.0 was preferred for the NEAR reactions under the conditions tested. As shown in FIG. 85, Bst 2.0 was able to generate an observable fluorescent signal (˜20,000 copies) in less than 5 minutes. In FIG. 85, Bst 3.0 performed slightly slower in IsoAmp I and IsoAmp II buffers. Therefore, Bst 2.0 and a concentration of 12 mM Mg2+ were selected for future NEAR reaction conditions.


Determining the Preferred Guide Sequence

This example describes how to select a guide RNA for detection of Strep-A, but it will be understood that the teachings described here may be used to select guides for other targets of interest as desired. The same primers and probe used in Example 27 were used. The following experiment determined if ssDNA could be detected using DETECTR from a NEAR reaction. Due to the biased reaction, gRNAs were generated in the reverse direction to detect any ssDNA produced from the nicking occurring on the forward primer nicking site.


A panel of 19 gRNAs as shown in TABLE 18 was synthesized in order to determine which gRNA worked best in the DETECTR reaction under the conditions tested. As shown in FIGS. 81A-81B, all gRNAs successfully detected the positive control which comprised an oligonucleotide that was a complement of the gRNA. Guide RNAs 14-19 successfully detected the NEAR reaction products containing the target and positive control, but did not detect the NEAR negative control and negative (no input) controls. It was observed that the maximum overlap with the guide RNA allowed was 3 nucleotides, otherwise signal detection in the negative controls were detected under the conditions tested.


Characterizing NEAR RNA Targets Using DETECTR

This example describes quantification of NEAR RNA targets derived from SARS COV-2 using the NECTR system described in Example 28. Two NEAR primers were used to amplify the SARS-COV-2 target sequence and a guide design as shown in FIG. 82A. As shown in FIGS. 82A-82B, the results of the SARS-COV-2 E-gene DETECTR reaction are shown for samples including 20,000 NEAR copies or 0 NEAR copies. The DETECTR reaction successfully detected SARS-COV-2 E-gene in the 20,000 target copy system and did not produce a signal in the 0 copy negative control conditions.


Next, the minimal incubation time of a NEAR reaction of Strep-A targets prior to detection via DETECTR was determined. The NEAR protocol was substantially similar to that described in Example 27. Incubation of the target with NEAR reaction components proceeded for 30 seconds, 1 minute, 2 minutes, 3 minutes, 4 minutes, 5 minutes, or 6 minutes prior to DETECTR. Under the conditions tested, a minimum of 2 minutes was observed to be required to achieve a detectable DETECTR signal, however, 6 minutes was found to be more optimal as it achieved a signal much higher than the limit of detection, as shown in FIG. 83.


Detecting NEAR RNA Targets Using Cas Variants

This example describes a comparison of NEAR amplicon detection using orthogonal Cas system to detect the target. The protocol of Example 27 was used for this example. Here, a Cas12 variant (SEQ ID NO: 28), a Cas13 variant (SEQ ID NO: 154), and a Cas14 variant (SEQ ID NO: 63) were utilized. They were chosen because they have different cleavage preferences for the reporter molecule and can be multiplexed together. In FIG. 84, the Cas12 variant showed superior signal after NEAR DETECTR reaction under the conditions tested. These results showed that it is possible to multiplex different NEAR-DETECTR reactions with orthogonal Cas systems.


We also optimized the stability of the hairpin loop in the nickase stabilization region present in the NEAR primers. Without being bound to any particular theory, it is thought that the hairpin loop may impact the nickase activity of the enzyme. To test this, alternative primer pairs to destroy, maintain, or enhance the hairpin loop formation (FIG. 87A) were used. The performance of these new hairpin loops were tested on detection of SARS-COV-2 E-gene as described in Example 28. In FIG. 87B, the destroyed hairpin loops showed reduced signal compared to both the normal and optimized hairpins, however, the enhanced hairpin showed strong signal even in the negative control under the conditions tested. It was determined that based on the results of this study that the normal primers would be used for future NEAR reactions.


Determining the Optimal Reverse Transcriptase

This example describes comparison of different reverse transcriptases for the RT-NECTR reaction described in Example 28. In this study, Wartmstart RTx (NEB), Bst 3.0, and Omniscript RT (Qiagen) were tested and they were compared by using the limit of detection as a metric. FIG. 88 shows that Omniscript achieved a better limit of detection compared to the other two reverse transcriptases under the conditions tested.


Optimizing Primer Concentrations

Primer concentrations were optimized in order to determine which concentrations would result in a better signal using the RT-NEAR reaction described in Example 28. In FIG. 89, primer concentrations of the forward and reverse NEAR primers were prepared at a concentration of 1000 nM, 750 nM, 500 nM, 250 nM nM, 100 nM, and 50 nM and mixed in a pairwise fashion in order to determine which concentration of forward and reverse primer will result in the best detectable signal. The forward primer used was M2805 (GAC TCC ACA CGG AGT Ctt gct ttc gtg gta tt (SEQ ID NO: 429)) and the reverse primer used was M2811 (GAC TCC ACA CGG AGT Cgg atg gct agt gta ac (SEQ ID NO: 430)). For this study, NEAR reactions were incubated and amplified targets for 5 minutes at 60° C. A combination of 1000 nM of the reverse primer and 500 nM of the forward primer performed best and resulted in a raw fluorescence greater than all other primer combinations tested. It was determined that by increasing the concentration of either the reverse or the forward primer resulted in increased efficiency of the reaction.


Optimizing Amplification Times

Due to the need to reverse transcribe the SARS-COV2 prior to detection, different amplification times need to be tested in order to determine the optimal resulting signal from RT-NECTR protocol of Example 28. In FIG. 90, different amplification times of 5 minutes, 10 minutes, and 20 minutes were tested on different cDNA (SARS-COV2 E-gene) inputs (500, 250, 100, 75, 10, and 0 cDNA copies). In this experiment, higher amplification times resulted in increased DETECTR signal, however, even after 20 minutes of incubation, DETECTR only resulted in a slight signal in 10 copies. At 75 copies, 10 minutes of amplification was sufficient to detect 75 copies of SAR-COV2 via DETECTR within 5 minutes.









TABLE 18







Exemplary Guides











GUIDE
ALTERNATIVE



SEQ ID NO:
NAME
NAME
Sequence





SEQ ID NO: 328
R1763
CoV2-G1
TTTGCCCCCAGCGCTTCAGC





GTTC





SEQ ID NO: 329
R1765
CoV2-G2
TTTCGTGGTATTGCTAGTTAC





SEQ ID NO: 330
EXAMPLE
EXAMPLE
CAATCTGAGGAGAGGCCATA



GUIDE 19
GUIDE 19






SEQ ID NO: 331
EXAMPLE
EXAMPLE
AATCTGAGGAGAGGCCATAC



GUIDE 18
GUIDE 18






SEQ ID NO: 332
EXAMPLE
EXAMPLE
ATCTGAGGAGAGGCCATACT



GUIDE 17
GUIDE 17






SEQ ID NO: 333
EXAMPLE
EXAMPLE
TCTGAGGAGAGGCCATACTT



GUIDE16
GUIDE16






SEQ ID NO: 334
EXAMPLE
EXAMPLE
CTGAGGAGAGGCCATACTTG



GUIDE 15
GUIDE 15






SEQ ID NO: 335
EXAMPLE
EXAMPLE
TGAGGAGAGGCCATACTTGT



GUIDE 14
GUIDE 14






SEQ ID NO: 336
EXAMPLE
EXAMPLE
GAGGAGAGGCCATACTTGTT



GUIDE 13
GUIDE 13






SEQ ID NO: 337
EXAMPLE
EXAMPLE
AGGAGAGGCCATACTTGTTC



GUIDE 12
GUIDE 12






SEQ ID NO: 338
EXAMPLE
EXAMPLE
GGAGAGGCCATACTTGTTCC



GUIDE 11
GUIDE 11






SEQ ID NO: 339
EXAMPLE
EXAMPLE
GAGAGGCCATACTTGTTCCT



GUIDE 10
GUIDE 10






SEQ ID NO: 340
EXAMPLE
EXAMPLE
AGAGGCCATACTTGTTCCTG



GUIDE 9
GUIDE 9






SEQ ID NO: 341
EXAMPLE
EXAMPLE
GAGGCCATACTTGTTCCTGT



GUIDE 8
GUIDE 8






SEQ ID NO: 342
EXAMPLE
EXAMPLE
AGGCCATACTTGTTCCTGTT



GUIDE 7
GUIDE 7






SEQ ID NO: 343
EXAMPLE
EXAMPLE
GGCCATACTTGTTCCTGTTT



GUIDE 6
GUIDE 6






SEQ ID NO: 344
EXAMPLE
EXAMPLE
GCCATACTTGTTCCTGTTTG



GUIDE 5
GUIDE 5






SEQ ID NO: 345
EXAMPLE
EXAMPLE
CCATACTTGTTCCTGTTTGG



GUIDE 4
GUIDE 4






SEQ ID NO: 346
EXAMPLE
EXAMPLE
CATACTTGTTCCTGTTTGGC



GUIDE 3
GUIDE 3






SEQ ID NO: 347
EXAMPLE
EXAMPLE
ATACTTGTTCCTGTTTGGCT



GUIDE 2
GUIDE 2






SEQ ID NO: 348
EXAMPLE
EXAMPLE
TACTTGTTCCTGTTTGGCTA



GUIDE 1
GUIDE 1









Example 30
Testing Cas System Orthogonality

This example describes testing of different Cas systems for orthogonality of the NEAR-DETECTR reaction described in Example 27. Cas14 variant (SEQ ID NO: 63) has been previously reported to detect ssDNA, whereas Cas13 variant (SEQ ID NO: 154) has been determined to be capable of detecting ssDNA. It was determined whether it was possible for the Cas14 variant or Cas13 variant to be used in NEAR-DETECTR to detect ssDNA. As shown in FIG. 91, Strep-A was detected using the primers M2048 and M2049, using the Cas12 variant (SEQ ID NO: 28), Cas13 variant (SEQ ID NO: 154), and Cas14 variant (SEQ ID NO: 63) systems. Detection was shown to be possible with Cas13 and Cas14 systems, suggesting the possibility of multiplexing NEAR amplicons using Cas12/13 or Cas14/13 reaction combinations.


While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. It is not intended that the invention be limited by the specific examples provided within the specification. While the invention has been described with reference to the aforementioned specification, the descriptions and illustrations of the embodiments herein are not meant to be construed in a limiting sense. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. Furthermore, it shall be understood that all aspects of the invention are not limited to the specific depictions, configurations or relative proportions set forth herein which depend upon a variety of conditions and variables. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is therefore contemplated that the invention shall also cover any such alternatives, modifications, variations or equivalents. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims
  • 1. A system for detecting a target nucleic acid, comprising a buffer comprising: i. amplification reagents for an amplification reaction targeting the target nucleic acid; andii. detection reagents for a detection reaction targeting the target nucleic acid; wherein the amplification reagents comprise one or more oligonucleotide primers, and a DNA polymerase;wherein the detection reagents comprise a programmable nuclease, a non-naturally occurring guide nucleic acid, and reporters;wherein the non-naturally occurring guide nucleic acid comprises a sequence that hybridizes to a segment of the target nucleic acid or DNA amplicons thereof; wherein the amplification reagents are present in amounts effective to amplify the target nucleic acid in a test sample to produce DNA amplicons of the target nucleic acid; andwherein the programmable nuclease and non-naturally occurring guide nucleic acid form a complex in the buffer that is activated upon binding one of the DNA amplicons to induce detectable transcollateral cleavage of the reporters.
  • 2. The system of claim 1, wherein (a) at least 1 nM of the reporters undergo transcollateral cleavage within one hour of addition of at least 5000 copies of the target nucleic acid to the system; (b) at least 5 nM of the reporters undergo transcollateral cleavage within one hour of addition of at least 5000 copies of the target nucleic acid to the system; (c) at least 10 nM of the reporters undergo transcollateral cleavage within one hour of addition of at least 5000 copies of the target nucleic acid to the system; (d) at least 1 nM of the reporters undergo transcollateral cleavage within one hour of addition of at least 1000 copies of the target nucleic acid to the system; or (e) at least 1 nM of the reporters undergo transcollateral cleavage within one hour of addition of at least 1000 copies of the target nucleic acid to the system.
  • 3-5. (canceled)
  • 6. The system of claim 1, wherein the amplification reagents comprise reagents for isothermal amplification.
  • 7-12. (canceled)
  • 13. The system of claim 1, wherein the programmable nuclease is a type V CRISPR/Cas effector protein.
  • 14-19. (canceled)
  • 20. The system of claim 1, further comprising a reverse transcriptase, an oligonucleotide primer, and dNTPs for reverse transcribing the target nucleic acid.
  • 21. The system of claim 1, wherein the buffer is a lysis buffer.
  • 22. (canceled)
  • 23. The system of claim 1, wherein the buffer comprises a) a pH of 7.5 to 8.5, at least 10 mM of a buffering agent, at least 1 mM ammonium acetate, at least 10 mM potassium acetate, at least 2.5 mM magnesium acetate, and at least 0.5% glycerol; or b) a pH of 7.5 to 8.5, at least 5 mM of a buffering agent, at least 20 mM potassium acetate, at least 2.5 mM magnesium acetate, and at least 0.5% glycerol; or c) a pH of 7.25 to 8.75, at least 5 mM of a buffering agent, at least 7.5 mM potassium acetate, at least 1 mM magnesium acetate, and at least 0.5% glycerol.
  • 24-28. (canceled)
  • 29. The system of claim 1, further comprising a circular template with internal complementarity formed from a single polynucleotide strand, wherein: (a) the circular template comprises a first portion with complementarity to one of the one or more oligonucleotide primers and a second portion with complementarity to a portion of the target nucleic acid;(b) the internal complementarity comprises part of the first portion and part of the second portion;(c) the second portion has a total length that is longer than a combined length of the first portion and second portion that are within the internal complementarity; and(d) the circular template undergoes a conformational change upon hybridization to the target nucleic acid to expose the first portion to hybridization to the oligonucleotide primer.
  • 30. The system of claim 1, further comprising a circular template, wherein: (a) the circular template comprises a first portion with complementarity to one of the one or more oligonucleotide primers and a second portion with complementarity to the target nucleic acid;(b) the oligonucleotide primer complementary to the first portion comprises a blocking motif at its 3′ end; and(c) the oligonucleotide primer complementary to the first portion undergoes cleavage to remove the blocking motif by the programmable nuclease in the presence of the target nucleic acid.
  • 31. The system of claim 1, further comprising a polymer matrix, wherein the polymer matrix is complexed with the reporters.
  • 32-34. (canceled)
  • 35. A method of assaying for a target nucleic acid in a sample, the method comprising: a. amplifying a portion of the target nucleic acid with a DNA polymerase to produce DNA amplicons of the target nucleic acid;b. forming a complex comprising one of the DNA amplicons, a programmable nuclease, and a non-naturally occurring guide nucleic acid that hybridizes to a segment of the DNA amplicon, thereby activating the programmable nuclease;c. cleaving reporters with the activated programmable nuclease; andd. detecting a change in a signal, wherein the change in the signal is produced by cleavage of the reporters;wherein the target nucleic acid and reagents for the amplifying and cleaving are present in the same reaction volume.
  • 36. The method of claim 35, wherein the amplifying and the cleaving occur simultaneously.
  • 37. The method of claim 35, further comprising measuring the rate of the change in the signal.
  • 38. The method of claim 35, further comprising measuring a concentration of the target nucleic acid in the sample based on the change in the signal.
  • 39. The method of claim 35, wherein the amplifying comprises isothermal amplification.
  • 40-43. (canceled)
  • 44. The method of claim 35, wherein the amplifying comprises reverse transcribing the target nucleic acid.
  • 45-52. (canceled)
  • 53. The method of claim 35, further comprising lysing a cell or virus comprising the target nucleic acid.
  • 54. The method of claim 53, wherein the lysing is performed in the same reaction volume as the amplifying and the cleaving.
  • 55-60. (canceled)
  • 61. The system of claim 1, wherein the system further comprises Thermostable inorganic pyrophosphatase (TIPP).
  • 62-74. (canceled)
  • 75. A system for detecting a target nucleic acid, comprising reagents in a buffer, wherein: (a) the reagents comprise hairpin polynucleotides, programmable nucleases, non-naturally occurring guide nucleic acids, and reporters;(b) each hairpin polynucleotide comprises one or more RNA loops, a first portion comprising DNA, and a second portion joined to the first portion by one of the one or more RNA loops;(c) each non-naturally occurring guide nucleic acid comprises a sequence that hybridizes to a segment of the target nucleic acid;(d) in each hairpin polynucleotide, the second portion of the hairpin polynucleotide hybridizes to a segment of the first portion;(e) the programmable nucleases and non-naturally occurring guide nucleic acids form complexes in the buffer that are activated upon binding the target nucleic acid;(f) an activated programmable nuclease is effective to induce (i) transcollateral cleavage of the one or more RNA loops, and (ii) detectable transcollateral cleavage of the reporters; and(g) cleavage of the one or more RNA loops of one of the hairpin polynucleotides is effective to release the first portion of the hairpin polynucleotide to hybridize with one of the non-naturally occurring guide nucleic acids and form a further activated programmable nuclease.
  • 76-77. (canceled)
  • 78. A method of assaying for a target nucleic acid in a sample, the method comprising the following steps in a single reaction volume: (a) forming a complex comprising the target nucleic acid, a first programmable nuclease, and a first non-naturally occurring guide nucleic acid that hybridizes to a segment of the target nucleic acid, thereby activating the programmable nuclease;(b) cleaving a hairpin polynucleotide of a plurality of hairpin polynucleotides with the activated programmable nuclease, wherein each hairpin polynucleotide comprises (i) one or more RNA loops that are cleaved, (i) a first portion comprising DNA, and (iii) a second portion joined to the first portion by one of the one or more RNA loops, wherein the second portion is hybridized to a segment of the first portion;(c) forming a second complex comprising the first portion of the cleaved hairpin polynucleotide, a second programmable nuclease, and a second non-naturally occurring guide nucleic acid that hybridizes to the first portion of the cleaved hairpin, thereby activating the second programmable nuclease;(d) cleaving reporters with the activated first or second programmable nuclease; and(e) detecting a change in a signal, wherein the change in the signal is produced by cleavage of the reporters.
  • 79-83. (canceled)
CROSS-REFERENCE

This application claims the benefit of U.S. Provisional Application No. 63/125,384, filed on Dec. 14, 2020; U.S. Provisional Application No. 63/166,923, filed on Mar. 26, 2021; U.S. Provisional Application No. 63/239,884 filed on Sep. 1, 2021; U.S. Provisional Application No. 63/125,387 filed on Dec. 14, 2020; U.S. Provisional Application No. 63/222,377 filed on Jul. 15, 2021; U.S. Provisional Application No. 63/239,917 filed on Sep. 1, 2021; and U.S. Provisional Application No. 63/151,592 filed on Feb. 19, 2021, each of which is incorporated herein by reference in its entirety.

STATEMENT AS TO FEDERALLY SPONSORED RESEARCH

This invention was made with government support under Contract No. N66001-21-C-4048 awarded by the Department of Defense, Defense Advanced Research Projects Agency (DARPA). The US government has certain rights in the invention.

Provisional Applications (7)
Number Date Country
63125384 Dec 2020 US
63125387 Dec 2020 US
63151592 Feb 2021 US
63166923 Mar 2021 US
63222377 Jul 2021 US
63239884 Sep 2021 US
63239917 Sep 2021 US
Continuations (1)
Number Date Country
Parent PCT/US21/63405 Dec 2021 WO
Child 18334320 US