The present disclosure provides, inter alia, specially designed DNA adaptors and various methods and kits for carrying out and detecting marker-free precision genome editing and genetic variation using such adaptors.
This application contains references to amino acids and/or nucleic acid sequences that have been filed as sequence listing text file “1035795-000704-seq.txt”, file size of 63 KB, created on Apr. 8, 2021. The aforementioned sequence listing is hereby incorporated by reference in its entirety pursuant to 37 C.F.R. § 1.52(e)(5).
Precision genome editing allows the modeling and correction of desired genomic variants containing insertions or deletions of specific nucleotide sequences or changes in single DNA bases (Anzalone et al., 2019; Barbieri et al., 2017; Cong et al., 2013; Dow, 2015; Guo et al., 2018; Liu et al., 2018; Mali et al., 2013; Roy et al., 2018). Precision genome editing can be obtained by CRISPR-dependent homology-directed repair (HDR) of Cas9-induced DNA double-strand breaks (DSBs) (Jasin and Haber, 2016) or result from the use of alternative DSB-free methods, such as CRISPR-dependent base editing, which utilizes cytidine or adenosine deaminases fused to a nickase Cas9 (nCas9) mutant to generate base transitions (Gaudelli et al., 2017; Komor et al., 2016), and prime editing, which employs a reverse transcriptase-nCas9 fusion and a template prime editing guide RNA (pegRNA) to install into the genome a large variety of genomic changes, including transversions, transitions, small insertions and deletions (Anzalone et al., 2019).
Genome editing has been facilitated by the development of accessible and cost-effective methods for the detection of small insertions and deletions (indels) resulting from the repair of Cas9-induced DSBs, such as the T7E1 and Surveyor nuclease assays (Mashal et al., 1995; Qiu et al., 2004; Ran et al., 2013). However, since these methods do not determine the identity of DNA bases, they are ill-suited for the detection of genomic changes introduced by precision genome editing (Germini et al., 2018). Precision genome editing events can be detected by the addition of genomic markers by CRISPR-dependent HDR or prime editing, such as silent mutations that create or disrupt restriction sites, or selectable reporters encoding for antibiotic resistance or fluorescent proteins. However, the use of genomic markers entails an elaborate experimental design that is unique for each targeted site, thus complicating the insertion of the desired genetic modifications. In addition, genomic markers can cause unintended perturbations of coding or non-coding genomic elements. Moreover, marker-based detection methods are not compatible with CRISPR-dependent base editing strategies, which induce single DNA base changes (Rees and Liu, 2018). Alternatively, methods that employ Sanger sequencing or next-generation sequencing (NGS) enable the detection of precise genomic changes without the use of genomic markers (Brinkman et al., 2014; Pinello et al., 2016). However, Sanger sequencing-based approaches suffer from low sensitivity and precision due to variable quality of the sequencing reactions and background signals that often affect the sequencing reads (Brinkman et al., 2014; Brinkman et al., 2018). While NGS-based detection strategies are highly sensitive (Clement et al., 2019; Lindsay et al., 2016; Pinello et al., 2016), they remain expensive and time-consuming, which limits their value for the development of mutant cell lines and animal models and for applications that require a rapid turnaround time, such as the identification of pathogenic variants in certain clinical settings. Therefore, a simple, efficient, inexpensive and rapid method that enables quantitative detection of genetic variants in complex biological systems is needed. This disclosure is directed to meeting these and other needs.
Genome editing technologies have transformed our ability to engineer desired genomic changes within living systems. However, detecting precise genomic modifications often requires sophisticated, expensive and time-consuming experimental approaches. The present disclosure provides DTECT (Dinucleotide signaTurE CapTure), a rapid and versatile detection method that relies on the capture of targeted dinucleotide signatures resulting from the digestion of genomic DNA amplicons by the type IIS restriction enzyme AcuI. DTECT enables the accurate quantification of marker-free precision genome editing events introduced by CRISPR-dependent homology-directed repair, base editing or prime editing in various biological systems, such as mammalian cell lines, organoids and tissues. Furthermore, DTECT allows the identification of oncogenic mutations in cancer mouse models, patient-derived xenografts and human cancer patient samples; it also allows the identification of genetic modifications incurred in various infectious diseases. Ultimately, DTECT enables the capture of signatures in nucleic acids from any organism including, e.g., viruses such as SARS-CoV-2. The ease, speed and cost efficiency by which DTECT identifies genomic signatures should facilitate the generation of marker-free cellular and animal models of human disease and expedite the detection of human pathogenic variants.
Accordingly, one embodiment of the present disclosure is a DNA adaptor comprising: (a) one strand with sequence of 5′-CTGGGGCACGGGTAAGAAGCATTCTGTCTCTCTTCTAAGAATTCGAGCTCGGTACC CG-3′ (SEQ ID NO: 230); and (b) one complementary strand with sequence of 5′-CGGGTACCGAGCTCGAATTCTTAGAAGAGAGACAGAATGCTTCTTACCCGTGCCC CAGNN-3′ with “N” corresponding to A, T, G or C (SEQ ID NOs: 231-246).
Another embodiment of the present disclosure is a method of preparing a DNA adaptor disclosed herein, comprising: (a) synthesizing one constant oligonucleotide with sequence of 5′-CTGGGGCACGGGTAAGAAGCATTCTGTCTCTCTTCTAAGAATTCGAGCTCGGTACC CG-3′ (SEQ ID NO: 230); (b) synthesizing one complementary oligonucleotide with sequence of 5′-CGGGTACCGAGCTCGAATTCTTAGAAGAGAGACAGAATGCTTCTTACCCGTGCCC CAGNN-3′ with “N” corresponding to A, T, G or C (SEQ ID NOs: 231-246); (c) mixing the constant and complementary oligonucleotides; and (d) annealing the mixture to obtain the DNA adaptor.
Another embodiment of the present disclosure is a library of DNA adaptors prepared by methods disclosed herein, the library comprises 16 DNA adaptors, wherein each DNA adaptor has a different “NN”.
Another embodiment of the present disclosure is a method for detecting a genetic modification, comprising the steps of: (a) amplifying a genomic locus of interest using a specially designed Type IIS restriction enzyme-tagging primer, comprising: (i) extracting genomic DNA from a biological sample of interest; (ii) synthesizing the Type IIS restriction enzyme-tagging primer based on the genomic locus of interest; (iii) amplifying the genomic locus of interest using the Type IIS restriction enzyme-tagging primer and a reverse primer; and (iv) purifying a Type IIS restriction enzyme-tagged genomic amplicon; (b) digesting the Type IIS restriction enzyme-tagged genomic amplicon with the Type IIS restriction enzyme; (c) isolating the smaller DNA fragment containing a genomic signature of interest exposed in a 3′ single-stranded overhang; (d) capturing the genomic signature of interest, comprising: (i) preparing the library of DNA adaptors disclosed herein; (ii) incubating the isolated smaller DNA fragment containing the 3′ overhang signature with the library of DNA adaptors and performing a ligation; and (iii) obtaining a ligated product; and (e) amplifying the ligated product to detect the presence of the genetic modification.
A further embodiment of the present disclosure is a kit for detecting a genetic modification of interest, comprising a specially designed Type IIS restriction enzyme-tagging primer disclosed herein, and a library of DNA adaptors disclosed herein, packaged together with instructions for its use.
Another embodiment of the present disclosure is a method for detecting a genetic modification, comprising the steps of: (a) amplifying a genomic locus of interest using a specially designed AcuI-tagging primer, comprising: (i) extracting DNA of interest; (ii) synthesizing the AcuI-tagging primer based on the genomic locus of interest; (iii) amplifying the genomic locus of interest using the AcuI-tagging primer and a reverse primer; and (iv) purifying an AcuI-tagged genomic amplicon; (b) digesting the AcuI-tagged genomic amplicon with restriction enzyme AcuI; (c) isolating the smaller DNA fragment containing a genomic signature of interest produced by AcuI-digestion; (d) capturing the genomic signature of interest, comprising: (i) preparing the library of DNA adaptors disclosed herein; (ii) incubating the isolated smaller DNA fragment with the library of DNA adaptors and performing a ligation; and (iii) obtaining a ligated product; and (e) amplifying the ligated product to detect the presence of the genetic modification.
An additional embodiment of the present disclosure is a kit for detecting a genetic modification, comprising a specially designed AcuI-tagging primer and a library of DNA adaptors disclosed herein, packaged together with instructions for its use.
Another embodiment of the present disclosure is a method for quantifying a genomic variant in a biological system, comprising the steps of: (a) obtaining a sample from the biological system; (b) amplifying a genomic locus of interest using a specially designed AcuI-tagging primer, comprising: (i) extracting DNA of interest; (ii) synthesizing the AcuI-tagging primer based on the genomic locus of interest; (iii) amplifying the genomic locus of interest using the AcuI-tagging primer and a reverse primer; and (iv) purifying an AcuI-tagged genomic amplicon; (c) digesting the AcuI-tagged genomic amplicon with restriction enzyme AcuI; (d) isolating the smaller DNA fragment containing a genomic signature of interest produced by the AcuI-digestion; (e) capturing the genomic signature of interest, comprising: (i) preparing the library of DNA adaptors disclosed herein; (ii) incubating the isolated smaller DNA fragment with the library of DNA adaptors and performing a ligation; and (iii) obtaining a ligated product; and (f) quantifying the genomic variant and determining its relative abundance.
Still another embodiment of the present disclosure is a method for identifying and quantifying an oncogenic mutation of interest in a biological sample, comprising the steps of: (a) obtaining a biological sample; (b) amplifying a genomic locus of interest using a specially designed AcuI-tagging primer, comprising: (i) extracting DNA of interest; (ii) synthesizing the AcuI-tagging primer based on the genomic locus of interest; (iii) amplifying the genomic locus of interest using the AcuI-tagging primer and a reverse primer; and (iv) purifying an AcuI-tagged genomic amplicon; (c) digesting the AcuI-tagged genomic amplicon with restriction enzyme AcuI; (d) isolating the smaller DNA fragment containing a genomic signature of interest produced by the AcuI-digestion; (e) capturing the genomic signature of interest, comprising: (i) preparing the library of DNA adaptors disclosed herein; (ii) incubating the isolated smaller DNA fragment with the library of DNA adaptors and performing a ligation; and (iii) obtaining a ligated product; (f) amplifying the ligated product to identify the presence of the oncogenic mutation of interest; and (g) quantifying the oncogenic mutation of interest, if present, and determining its frequency.
A further embodiment of the present disclosure is a process for marker-free detection of a precision genome editing event comprising carrying out Dinucleotide signaTurE CapTure (DTECT) on a nucleic acid sequence of interest.
Still another embodiment of the present disclosure is a method for detecting a virus variant of interest, comprising the steps of: (a) obtaining a nucleic acid of the virus variant of interest from a biological sample; and (b) if the nucleic acid is DNA, carrying out Dinucleotide signaTurE CapTure (DTECT) to detect the variant of interest; or (c) if the nucleic acid is RNA, coverting it to DNA by reverse transcription PCR (RT-PCR) and then carrying out DTECT to detect the variant of interest.
The present disclosure provides a versatile method that uses standard molecular biology techniques to detect variants introduced by precision genome editing or resulting from genetic variation. This detection method, designated Dinucleotide signaTurE CapTure (DTECT), enables accurate and sensitive quantification of marker-free precision genome editing events induced by CRISPR-dependent HDR, base editing and prime editing. In addition, we show that DTECT can readily identify oncogenic mutations in cancer mouse models, patient-derived xenograft models and cancer patient samples. These studies establish a cost-effective method for the rapid detection of genetic variants, which will aid the generation of marker-free cellular and animal models of human disease and expedite the detection of pathogenic variants for clinical applications.
Accordingly, one embodiment of the present disclosure is a DNA adaptor comprising: (a) one strand with sequence of 5′-CTGGGGCACGGGTAAGAAGCATTCTGTCTCTCTTCTAAGAATTCGAGCTCGGTACC CG-3′ (SEQ ID NO: 230); and (b) one complementary strand with sequence of 5′-CGGGTACCGAGCTCGAATTCTTAGAAGAGAGACAGAATGCTTCTTACCCGTGCCC CAGNN-3′ with “N” corresponding to A, T, G or C (SEQ ID NOs: 231-246).
In some embodiments, the DNA adaptor is labeled with a detection molecule. Non-limiting examples of the detection molecule include a radiolabel, a fluorescent label, a biotinylated label, a non-fluorescent label, an enzyme, a hapten, a phosphorescent molecule, a chemiluminescent molecule, a chromophore, a luminescent molecule, a photoaffinity molecule, a color particle or a ligand.
Another embodiment of the present disclosure is a method of preparing a DNA adaptor disclosed herein, comprising: (a) synthesizing one constant oligonucleotide with sequence of 5′-CTGGGGCACGGGTAAGAAGCATTCTGTCTCTCTTCTAAGAATTCGAGCTCGGTACC CG-3′ (SEQ ID NO: 230); (b) synthesizing one complementary oligonucleotide with sequence of 5′-CGGGTACCGAGCTCGAATTCTTAGAAGAGAGACAGAATGCTTCTTACCCGTGCCC CAGNN-3′ with “N” corresponding to A, T, G or C (SEQ ID NOs: 231-246); (c) mixing the constant and complementary oligonucleotides; and (d) annealing the mixture to obtain the DNA adaptor.
Another embodiment of the present disclosure is a library of DNA adaptors prepared by methods disclosed herein, the library comprises 16 DNA adaptors, wherein each DNA adaptor has a different “NN”.
Another embodiment of the present disclosure is a method for detecting a genetic modification, comprising the steps of: (a) amplifying a genomic locus of interest using a specially designed Type IIS restriction enzyme-tagging primer, comprising: (i) extracting genomic DNA from a biological sample of interest; (ii) synthesizing the Type IIS restriction enzyme-tagging primer based on the genomic locus of interest; (iii) amplifying the genomic locus of interest using the Type IIS restriction enzyme-tagging primer and a reverse primer; and (iv) purifying a Type IIS restriction enzyme-tagged genomic amplicon; (b) digesting the Type IIS restriction enzyme-tagged genomic amplicon with the Type IIS restriction enzyme; (c) isolating the smaller DNA fragment containing a genomic signature of interest exposed in a 3′ single-stranded overhang; (d) capturing the genomic signature of interest, comprising: (i) preparing the library of DNA adaptors disclosed herein; (ii) incubating the isolated smaller DNA fragment containing the 3′ overhang signature with the library of DNA adaptors and performing a ligation; and (iii) obtaining a ligated product; and (e) amplifying the ligated product to detect the presence of the genetic modification.
In some embodiments, the genetic modification is selected from a base change, a deletion, or an insertion. In some embodiments, the genetic modification is selected from a single genomic change or multiple genomic changes. In some embodiments, the multiple genomic changes can occur within a single locus or distinct loci.
In some embodiments, the Type IIS restriction enzyme is selected from AcuI, BpmI, BpuEI, BsgI, MmeI and NmeAIII. In some embodiments, the Type IIS restriction enzyme is selected from AcuI and BpuEI. In some embodiments, the Type IIS restriction enzyme is AcuI.
In some embodiments, the Type IIS restriction enzyme-tagging primer is an oligonucleotide comprising: (a) a non-complementary handle sequence positioned on the 5′ side; (b) a complementary sequence of the genomic locus of interest on the 5′ side; (c) a recognition motif of the Type IIS restriction enzyme that is positioned at a predicted distance from its cleavage site to generate the genomic signature of interest; and (d) a complementary sequence of the genomic locus of interest on the 3′ side.
In some embodiments, the reverse primer is positioned at more than 100 bp downstream of the genomic locus of interest.
In some embodiments, the non-complementary handle sequence can have any suitable length. In some embodiments, the non-complementary handle sequence is 25 bp. In some embodiments, the non-complementary handle sequence can have any suitable sequence. In some embodiments, the non-complementary handle sequence is 5′-GCAATTCCTCACGAGACCCGTCCTG-3′ (SEQ ID NO: 3).
In some embodiments, the ligation in step (d)(ii) of the methods disclosed above is carried out by T4 DNA ligase.
A further embodiment of the present disclosure is a kit for detecting a genetic modification of interest, comprising a specially designed Type IIS restriction enzyme-tagging primer disclosed herein, and a library of DNA adaptors disclosed herein, packaged together with instructions for its use. In some embodiments, the Type IIS restriction enzyme is AcuI.
Another embodiment of the present disclosure is a method for detecting a genetic modification, comprising the steps of: (a) amplifying a genomic locus of interest using a specially designed AcuI-tagging primer, comprising: (i) extracting DNA of interest; (ii) synthesizing the AcuI-tagging primer based on the genomic locus of interest; (iii) amplifying the genomic locus of interest using the AcuI-tagging primer and a reverse primer; and (iv) purifying an AcuI-tagged genomic amplicon; (b) digesting the AcuI-tagged genomic amplicon with restriction enzyme AcuI; (c) isolating the smaller DNA fragment containing a genomic signature of interest produced by AcuI-digestion; (d) capturing the genomic signature of interest, comprising: (i) preparing the library of DNA adaptors disclosed herein; (ii) incubating the isolated smaller DNA fragment with the library of DNA adaptors and performing a ligation; and (iii) obtaining a ligated product; and (e) amplifying the ligated product to detect the presence of the genetic modification.
In some embodiments, the AcuI-tagging primer is an oligonucleotide comprising: (a) a non-complementary handle sequence positioned on the 5′ side; and (b) a complementary sequence of the genomic locus of interest containing an AcuI motif (5′-CTGAAG-3′) positioned 14 bp upstream from the genomic locus of interest.
In some embodiments, the AcuI-tagging primer can have any suitable length. In some embodiments, the AcuI-tagging primer is 60 bp.
In some embodiments, the reverse primer is positioned at more than 100 bp downstream of the genomic locus of interest.
In some embodiments, the non-complementary handle sequence can have any suitable length. In some embodiments, the non-complementary handle sequence is 25 bp.
In some embodiments, the complementary sequence has the structure of: 5′-N(20)CTGAAGN(14)-3′ or 5′-N(15)CTGAAGN(14)-3′, with “N” corresponding to A, T, G or C, depending on the DNA sequence of the genomic locus of interest.
In some embodiments, the non-complementary handle sequence is 5′-GCAATTCCTCACGAGACCCGTCCTG-3′ (SEQ ID NO: 3) and the complementary sequence is 5′-N(15)CTGAAGN(14)-3′, with “N” corresponding to A, T, G or C.
In some embodiments, the ligation in step (d)(ii) of the methods disclosed above is carried out by T4 DNA ligase.
An additional embodiment of the present disclosure is a kit for detecting a genetic modification, comprising a specially designed AcuI-tagging primer and a library of DNA adaptors disclosed herein, packaged together with instructions for its use.
Another embodiment of the present disclosure is a method for quantifying a genomic variant in a biological system, comprising the steps of: (a) obtaining a sample from the biological system; (b) amplifying a genomic locus of interest using a specially designed AcuI-tagging primer, comprising: (i) extracting DNA of interest; (ii) synthesizing the AcuI-tagging primer based on the genomic locus of interest; (iii) amplifying the genomic locus of interest using the AcuI-tagging primer and a reverse primer; and (iv) purifying an AcuI-tagged genomic amplicon; (c) digesting the AcuI-tagged genomic amplicon with restriction enzyme AcuI; (d) isolating the smaller DNA fragment containing a genomic signature of interest produced by the AcuI-digestion; (e) capturing the genomic signature of interest, comprising: (i) preparing the library of DNA adaptors disclosed herein; (ii) incubating the isolated smaller DNA fragment with the library of DNA adaptors and performing a ligation; and (iii) obtaining a ligated product; and (f) quantifying the genomic variant and determining its relative abundance.
In some embodiments, the genomic variant is generated by precision genome editing. In some embodiments, the precision genome editing is CRISPER-dependent homology-directed repair, base editing or prime editing.
In some embodiments, the biological system is a mammalian cell line, an organoid, or a tissue.
In some embodiments, the quantification in step (f) of the methods disclosed above is carried out by quantitative PCR (qPCR).
Still another embodiment of the present disclosure is a method for identifying and quantifying an oncogenic mutation of interest in a biological sample, comprising the steps of: (a) obtaining a biological sample; (b) amplifying a genomic locus of interest using a specially designed AcuI-tagging primer, comprising: (i) extracting DNA of interest; (ii) synthesizing the AcuI-tagging primer based on the genomic locus of interest; (iii) amplifying the genomic locus of interest using the AcuI-tagging primer and a reverse primer; and (iv) purifying an AcuI-tagged genomic amplicon; (c) digesting the AcuI-tagged genomic amplicon with restriction enzyme AcuI; (d) isolating the smaller DNA fragment containing a genomic signature of interest produced by the AcuI-digestion; (e) capturing the genomic signature of interest, comprising: (i) preparing the library of DNA adaptors disclosed herein; (ii) incubating the isolated smaller DNA fragment with the library of DNA adaptors and performing a ligation; and (iii) obtaining a ligated product; (f) amplifying the ligated product to identify the presence of the oncogenic mutation of interest; and (g) quantifying the oncogenic mutation of interest, if present, and determining its frequency.
In some embodiments, the biological sample is obtained from a cancer animal model, a patient-derived xenograft (PDX), or a human cancer patient sample.
In some embodiments, the quantification in step (g) of the methods disclosed above is carried out by quantitative PCR (qPCR).
A further embodiment of the present disclosure is a process for marker-free detection of a precision genome editing event comprising carrying out Dinucleotide signaTurE CapTure (DTECT) on a nucleic acid sequence of interest.
DTECT can also be used to detect genetic signatures in any organism, for example, a virus. Thus, still another embodiment of the present disclosure is a method for detecting a virus variant of interest, comprising the steps of: (a) obtaining a nucleic acid of the virus variant of interest from a biological sample; and (b) if the nucleic acid is DNA, carrying out Dinucleotide signaTurE CapTure (DTECT) to detect the variant of interest; or (c) if the nucleic acid is RNA, coverting it to DNA by reverse transcription PCR (RT-PCR) and then carrying out DTECT to detect the variant of interest. This detection method is applicable to any type of virus including but not limited to a DNA virus, an RNA virus, a retrovirus, etc. In some embodiments, the virus is an RNA virus. In some embodiments, the virus is SARS-CoV-2.
The following examples are provided to further illustrate the methods of the present disclosure. These examples are illustrative only and are not intended to limit the scope of the disclosure in any way.
Material Availability
Plasmids for DTECT quantification and expression of base editing sgRNAs targeting BRCA1, BRCA2 and FANCD2 have been deposited to Addgene (#139321-139333, and 139511).
Cell Line Generation and Single Clone Isolation
HEK293T and DLD1 cell lines were obtained from ATCC. Cells were cultured in DMEM (ThermoFisher Scientific) supplemented with 10% Fetalgro bovine growth serum (BGS, RMBIO) and 1% penicillin-streptomycin (ThermoFisher Scientific). Cells were grown at 37° C. with 5% CO2 and tested regularly for mycoplasma. NIH/3T3 were maintained in DMEM supplemented with 10% bovine calf serum. Organoids were isolated and cultured as previously described (Zafra et al., 2018). To generate cells constitutively expressing FNLS-BE3-P2A-BlastR, HEK293T cells were infected with a lentivirus expressing the above construct. Viruses were produced in HEK293T in 6-well plates by transfecting 2 μg of FNLS-BE3-P2A-BlastR, 0.2 μg of Tat, 0.2 μg of Gag/Pol, 0.2 μg of Rev, 0.4 μg of VSV-G expressing plasm ids in 250 μl of DMEM without serum. 9 μl of TransIT-293 (Mirus) were added to the DNA, mixed and incubated for 15 min at room temperature. The DNA transfection reagent mix was added dropwise to the cells and incubated at 37° C. with 5% CO2. The next day the cell medium was replaced and cells were incubated for 48 hours. The medium containing lentiviruses was then collected and utilized to infect new HEK293T cells. 48 hours after infection, blasticidin was added to the medium until the uninfected control cells were killed. FNLS-BE3 expression was determined by western blot and the base editing activity of the construct was tested using previously validated sgRNAs. Single HEK293T clones were selected for high base editing efficiency. Clones were isolated by trypsinization of the initial cell population into individual cells. Cell density was evaluated by counting the cells with a hemocytometer and cells were diluted to approximately 0.13 cells/μl, equivalent to 20 cells per 150 μl. Serial dilutions were prepared and 150 μl of the diluted cell mixture were seeded into 96-well plates. Single clones were expanded and further examined for FNLS-BE3 expression and activity.
Editing of Cell Lines, Organoids and Mice
To induce CRISPR-mediated HDR editing, HEK293T cells were seeded at 50%-70% confluency into 24-well plates and reverse transfected with 0.25 μg of sgRNA and 0.25 μg of Cas9 expressing plasmid (Addgene #42230) with or without 0.5 μl of ssODN (40 μM) into 100 μl of DMEM without Fetalgro BGS and antibiotics. 3 μl of TransIT-293 (Mirus) were added to the DNA, mixed and incubated for 15 min at room temperature. Experiments involving i53 were done by adding 0.25 μg of i53 (Addgene #77939) to the transfection mixture. The gDNAs of cell populations and individual clones were recovered by resuspending the cell pellets in the Quick Extract DNA Extraction Solution (Epicentre), followed by incubation at 65° C. for 10 min and 95° C. for 5 min. The isolated gDNAs were diluted in H2O, quantified using Nanodrop and stored at −20° C. or directly used in PCR reactions. In base editing experiments, we used cells constitutively expressing FNLS-BE3 or transfected with pCMV-BE3 (Addgene #73021) and sgRNAs, as described above. Empty plasmids (Addgene #100708) with no sgRNAs were used as controls. To determine the accuracy of the quantification of variant frequency by DTECT (
In order to introduce multiple variants into the BRCA1 and BRCA2 genes, HEK293T cells expressing FNLS-BE3 were seeded at 50%-70% confluency into 24-well plates and reverse transfected with 1 μg of sgRNA into 100 μl of DMEM without Fetalgro BGS and antibiotics. 3 μl of TransIT-293 (Mirus) were added to the DNA, mixed and incubated for 15 min at room temperature. The DNA transfection mix was added dropwise to the cells and incubated at 37° C. with 5% CO2 for 4 days. Single clones were generated and the gDNAs of cell populations and individual clones were recovered as describe above. Genomic loci were Sanger sequenced by Eton Bioscience or Genewiz. Sanger sequencing data were analyzed using Serial cloner and viewed by Snapgene Viewer. The sequencing profiles shown in this manuscript were generated by SnapGene Viewer. Quantitative detection of the editing level using the AcuI-tagged amplicon was done blindly.
In vivo mouse editing was performed as previously described (Zafra et al., 2018). Briefly, eight week-old C57BL/6N mice (Charles River) were injected with 0.9% sterile sodium chloride solution containing 20 μg of pLenti-FNLS-P2A-Puro and 10 μg of sgRNA vector. The total injection volume corresponded to 20% of the individual mouse body weight and was injected into the lateral tail vein in 5-7 seconds. All animal experiments were authorized by the regional board of Karlsruhe, Germany.
Mouse Genotyping and Bone Marrow Transplantation
The generation of genetically engineered mice harboring the Brca1 S1598F and Bard1 S563F alleles was previously described (Billing et al., 2018; Shakya et al., 2011). Mouse genotyping was performed using DTECT on genomic DNA extracted from mouse tails. AcuI-tagging of the targeted loci was performed using 50 ng of gDNA (see DTECT protocol above). All primer sequences are listed in Table S1. Genotyping experiments were conducted blindly.
Competitive transplantation experiments were performed to assess chimerism of Jak2 V617F mutant cells in relation to wild-type support. Specifically, Mx1-Cre+; CD45.2 Jak2V617F/+ and Mx1Cre+; CD45.1 wild-type mice were dosed with polyinosine-polycytosine (PIPC) 8 weeks prior to sacrifice to induce MPN in mutant mice. On day of sacrifice, dissected femurs and tibias were isolated and bone marrow flushed with a syringe into PBS. Red blood cells (RBCs) were lysed in ammonium chloride-potassium bicarbonate lysis buffer for 10 min on ice. 1.5×106 filtered whole donor Mx1-Cre+; Jak2V617F/+ bone marrow cells (CD45.2) were then mixed with wild-type 1.5×106 competitor bone marrow cells (CD45.1) and transplanted via tail vein injection into lethally irradiated (2×550 Rad) CD45.1 host mice. Mice were then monitored serially for the development of MPN based on blood counts and donor chimerism by retroorbital bleed draws using heparinized microhematocrit capillary tubes (ThermoFisher Scientific). After 3 consecutive hematocrits of >65%, mice were then sacrificed for peripheral blood fluorescence-activated cell sorting (FACS) analysis and DNA extraction. All animal procedures were conducted in accordance with the Guidelines for the Care and Use of Laboratory Animals and were approved by the Institutional Animal Care and Use Committees at Memorial Sloan Kettering Cancer Center. The conditional Mx1-Cre+; Jak2V617F/+ mice are all C57BL/6 background and have been previously described (Mullally et al., 2010). Automated peripheral blood counts were obtained using a ProCyte Dx (IDEXX Laboratories) according to the manufacturer's protocol. For surface flow cytometry of mouse peripheral blood, bone marrow, and spleen, RBCs were lysed and stained with monoclonal antibodies in PBS plus 1% BSA for 1 hour on ice. For flow cytometry of erythroid lineage, bone marrow or splenic cells were stained without RBC lysis. DAPI was used for live/dead cell analysis. Cell populations were analyzed using an LSR Fortessa (Becton Dickinson), and data were analyzed with FlowJo software (Tree Star). DNA extraction was performed using the QIAamp DNA Micro Kit (Qiagen) per manufacturer's protocol.
Analysis of ALL Patient Samples and PDXs
DNA samples from leukemic ALL blasts obtained at diagnosis and after relapse were provided by multiple institutions, as previously described (Oshima et al., 2016). Informed consent was obtained at study entry and samples were collected under the supervision of local Institutional Review Boards for participating institutions and analyzed under the supervision of the Columbia University Irving Medical Center Institutional Review Board. Research was conducted in compliance with ethical regulations. ALL patients received standard combination chemotherapy at diagnosis. Diagnosis and relapse samples were harvested from bone marrow. High molecular weight genomic DNA from matched diagnosis and relapse samples of ALL patients was extracted from patient leukemic blasts or from xenografts using the DNeasy Blood & Tissue Kit (Qiagen) or the AllPrep DNA/RNA Mini Kit (Qiagen). Primary human xenograft ALL cells were passaged and harvested from the spleens of NRG (NOD.Cg-ag1tm1MomII2rgtm1WjI/SzJ, The Jackson Laboratory) mice. Whole exome sequencing was performed and analyzed as previously described (Oshima et al., 2016).
Vector Construction and Cloning
sgRNAs were synthesized as complementary oligonucleotides (IDT) compatible with BbsI restriction sites located into the B52 plasmid (Addgene #100708). Oligonucleotides were designed as previously described (Billon et al., 2017). Cloned sgRNAs were verified by Sanger sequencing. Sequences of the sgRNAs are available in Table S1. ssODNs used in HDR experiments were synthesized as ultramer oligos (IDT) and their sequences are available in Table S1. To generate the FNLS-BE3-P2A-BlastR plasmid, the pLenti-FNLS-P2A-Puro plasmid (Addgene #110841) (Zafra et al., 2018) was modified by replacing the puromycin resistance gene with the blasticidin resistance gene. Briefly, the blasticidin resistance gene coding sequence was amplified by PCR and recombined using Gibson assembly into FNLS-BE3-P2A. The FNLS-BE3-P2A-BlastR sequence was verified by Sanger sequencing.
AcuI-Tagging Primer Design
The AcuI-tagging oligonucleotide enables the insertion of an AcuI motif (5′-CTGAAG-3′) 14 bp away from a targeted dinucleotide. This motif is inserted as a hairpin in the middle of a sequence complementary to the targeted genomic locus. The AcuI-tagging oligonucleotide is 60 bp-long and contains a non-complementary handle sequence of 20-25 bp. Common handle sequences used are PB547 (5′-GATCCTCTAGAGTCGACCTG-3′) (SEQ ID NO: 1) or PB1072 (5′-GCAATTCCTCACGAGACCCGTCCTG-3′) (SEQ ID NO: 3) (Table S1). The oligonucleotide sequence complementary to the targeted genomic locus plus the AcuI motif has the following sequence: 5′-N(20)CTGAAGN(14)-3′ or 5′-N(15)CTGAAGN(14)-3′, with “N” corresponding to A, T, G or C bases complementary to the targeted locus. Reverse primers used in AcuI-tagging reactions were designed by Primer 3 (http://bioinfo.ut.ee/primer3-0.4.0/) using the default parameters with the following changes: Mispriming library=“HUMAN” for amplifying from human genomic DNA or Mispriming library=“RODENT” for amplifying from mouse genomic DNA, Primer size “min=25, Opt=27, Max=30”, Primer Tm “Min=57.0° C., Opt=60.0° C., Max=63.0° C.”. Reverse primers are located >100 bp away from the targeted dinucleotides. All sequences of the primers used in this study are available in Table S1.
Adaptor Library Generation and Characterization
A set of 17 individual oligonucleotides constitutes the full adaptor library. This library contains: a) One constant oligonucleotide with the following sequence: 5′-CTGGGGCACGGGTAAGAAGCATTCTGTCTCTcttctaagaattcgagctcggtacccg-3′ (SEQ ID NO: 230). The lowercase nucleotide sequence located at the 3′-end of the constant oligonucleotide (5′-cttctaagaattcgagctcggtacccg-3′) (SEQ ID NO: 319) corresponds to the handle sequence used to detect the ligated products with either PB548 (5′-cgggtaccgagctcgaattc-3′) (SEQ ID NO: 2) or PB1073 (5′-cgggtaccgagctcgaattcttagaag-3′) (SEQ ID NO: 4); b) 16 variable oligonucleotides that contain a sequence complementary to the constant oligonucleotide plus one of 16 different dinucleotides at their 3′-end. The variable oligonucleotides have the following sequence: 5′-cgggtaccgagctcgaattcttagaagAGAGACAGAATGCTTCTTACCCGTGCCCCAGNN-3′. NN, with N=A, C, G or T (SEQ ID NOs: 231-246), corresponds to the dinucleotide that is different for each of the 16 oligos. The adaptor sequences are available in Table S1. The constant oligonucleotide and each variable oligonucleotide were resuspended at a concentration of 100 μM in H2O. 2.5 μl of constant oligonucleotide and 2.5 μl of each variable oligonucleotide were mixed with 1× ligase buffer (ThermoFisher Scientific) and water in a 20 μl reaction. The reactions were placed in a thermocycler and oligonucleotides were annealed by incubating them for 5 min at 95° C., followed by a gradual temperature decrease from 95° C. to 15° C. After annealing was completed, 100 μl of water were added to dilute the adaptors in a 120 μl final volume. Adaptors were frozen and stored at −20° C.
The adaptor library was tested at two independent loci, as shown in
The measurement of the dinucleotide capture efficiency of each adaptor (
The assay performed to measure the efficiency of DNA ligation (
To calculate the frequency of non-specific dinucleotide capture shown in
DTECT Protocol
The DTECT protocol consists of 6 steps (I-VI,
Next-Generation Sequencing
Samples for NGS were prepared by amplifying the edited regions of interest by PCR. Samples were sequenced by the Genome Sciences Facility at The Pennsylvania State College of Medicine or by Genewiz and the results were analyzed by Genewiz, or by using an R-based script of the Ciccia laboratory or CRISPResso2 (Clement et al., 2019). To ensure that no biases were introduced during DTECT assays, the AcuI-tagging amplicons for the BRCA1 and BRCA2 mutant samples were sequenced by NGS and analyzed using an R-based script. In this analysis, 7 sequences with >6000 reads were filtered out from the analysis due to incorrect sequence. The editing frequency from the NGS results were determined using the formula: ((Number of reads for the edited dinucleotide)/(total number of reads))×100. Oligonucleotides used for PCR amplifications, Illumina sequencing adaptors and indexes are listed in Table S1.
Quantification and Statistical Analysis
Technical duplicates of each sample were performed in each qPCR reaction. A standard curve to determine the concentration of the captured material was generated using predefined concentrations of a DTECT ligation product (
Data and Code Availability
R-based scripts of the Ciccia laboratory for analysis of NGS reads and ClinVar datasets are available upon request. Raw NGS reads of edited DLD1 and NIH/3T3 cells, organoids and liver samples are available under accession SRP151111 in the Sequence Read Archive. NGS reads have been deposited into the NCBI database and are and are accessible as BioProject #PRJNA603357. All uncropped gels, raw qPCR data and Sanger sequencing reads are available in Mendeley (https://data.mendeley.com/datasets/gtkk6sthtw/draft?a=ca72630e-56eb-4e29-bcdb-158b2c7d4123).
In our detection method, we take advantage of the property of type IIS restriction enzymes to generate single-stranded DNA overhangs at a specific distance from their recognition motif. Based on the above property, we hypothesized that single-stranded DNA overhangs generated by digestion of genomic DNA sequences with type IIS restriction enzymes could be captured and identified using DNA adaptors containing overhangs complementary to the exposed DNA signatures (
In our approach, the genomic locus of interest is PCR-amplified using a locus-specific DNA primer (red) and a DNA oligonucleotide (AcuI-tagging primer) containing two regions of complementarity to the genomic locus (purple) interrupted by an AcuI recognition site (AcuI hairpin, green) positioned 14 bp upstream of a dinucleotide of interest (
To demonstrate the feasibility of DTECT, we designed two AcuI-tagging DNA primers flanking four adjacent bases (5′-TTGG-3′) on opposite DNA strands (TT and CC signatures, blue) (
Next, we examined whether DTECT can determine the relative abundance of DNA variants with distinct DNA signatures, including low abundance DNA variants. To this end, we transfected HEK293T cells with sgRNAs that introduce nonsense mutations into the SPRTN, PIK3R1 and SMARCAL1 genes using iSTOP, a CRISPR-mediated base editing approach that creates STOP codons within genes of interest (Billon et al., 2017) (
To examine the ability of DTECT to identify precise genomic changes introduced into mammalian cell populations, we utilized CRISPR-mediated HDR for generating various types of disease-related mutations using single-stranded oligodeoxynucleotides (ssODNs), including a cancer-associated frameshift mutation in TP53 (i.e., R209fs*6), a missense mutation in HBB (i.e., G6V) that causes sickle cell anemia, a small tandem duplication in BRCA2 (dupAGAAGAT) identified in breast cancer, and small insertions into JAK2 and EMX1 (Paulsen et al., 2017), two genes associated with myeloproliferative disorders and Kallmann syndrome, respectively. Three days after co-transfection of Cas9 with site-specific sgRNAs and ssODNs into HEK293T cells, we harvested the cellular genomic DNA and utilized DTECT to determine by analytical and quantitative PCR whether the desired changes were incorporated into the targeted chromosomal loci (
To further confirm the accuracy of DTECT in quantifying precision genome editing, we compared the frequency of editing events determined by either DTECT or NGS across 62 samples derived from human cells, mouse cells and intestinal organoids, which were modified using CRISPR-mediated HDR or base editing (Zafra et al., 2018). As shown in
Recent studies led to the development of Sanger sequencing-based methods, such as ICE (Synthego; https://ice.synthego.com/#/) or EditR (Kluesner et al., 2018), that enable the detection of genomic variants based on the deconvolution of chromatogram peaks. To compare DTECT with the above methods, we subjected to Sanger sequencing the genomic amplicons of 23 samples edited by precision genome editing. In these experiments, we used two primers annealing to opposite DNA strands to obtain independent sequencing duplicates of the same amplicons, and analyzed the Sanger sequencing reads using either ICE or EditR. Notably, ˜10% of the sequencing reactions failed to generate high quality reads required for ICE or EditR, despite using high quality amplicons for sequencing (Mendeley dataset, Data availability section). Independent repeats using new genomic amplicons did not improve the sequencing outcome (Mendeley dataset, Data availability section). In addition, we noted that technical duplicates of Sanger sequencing reactions analyzed by ICE or EditR displayed lower levels of consistency relative to technical replicates of DTECT assays (
The modeling and correction of pathogenic mutations in adult mice is critical for the development of novel approaches to therapeutic intervention against cancer and other diseases (Chadwick et al., 2017; Gao et al., 2018; Levy et al., 2020; Ryu et al., 2018; Song et al., 2020; Villiger et al., 2018; Yin et al., 2016; Yin et al., 2014). To determine whether DTECT can determine editing levels in adult mouse tissue, we hydrodynamically delivered into the mouse liver (Tschaharganeh et al., 2014) a cytidine base editor and an sgRNA introducing the oncogenic Pik3ca E545K mutation (Zafra et al., 2018) (
The above studies indicate that DTECT can determine the identity of individual genomic changes. To examine whether DTECT can also identify complex sets of mutations, we employed CRISPR-dependent base editing to target two adjacent cytosines in the EMX1 locus that had previously been converted into four distinct dinucleotide combinations (i.e., CC, CT, TC or TT) by base editing (Komor et al., 2016) (
Precision genome editing allows the modeling of clinically relevant gene variants. Given that DTECT enables the identification of newly created DNA signatures without requiring the insertion of markers or elaborate experimental design specific for each edited site, we tested whether DTECT could facilitate the generation of multiple cell lines harboring clinically relevant mutations. In particular, we focused our attention on mutations in the BRCA1 and BRCA2 genes, which in heterozygosity can predispose women to the development of breast and/or ovarian cancer (Apostolou and Fostira, 2013), whereas in homozygosity can cause Fanconi anemia (Ceccaldi et al., 2016). More than 7,000 clinically associated SNVs have been identified in BRCA1/2, according to the ClinVar database, but efforts to characterize their functional impact and pathogenic potential have been limited in part due to the challenge of generating cell lines that carry such a large number of individual homozygous and heterozygous variants. To determine whether DTECT can facilitate the production of cell lines harboring clinically relevant BRCA1/2 SNVs, we expressed a cytidine base editor in HEK293T cells along with individual sgRNAs to generate 23 different BRCA1/2 mutations identified in patients with ovarian and breast cancers, as reported in ClinVar (
Given the ability of DTECT to correctly determine the genotype of cellular clones, we then tested whether DTECT could also be applied to mouse genotyping. To this end, we obtained tail DNA samples from genetically engineered mice carrying knock-in mutations in Brca1 (S1598F) and its partner protein Bard1 (S563F) (Billing et al., 2018). As shown in
Precise and rapid detection of pathogenic variants in patients is critical for accurate diagnosis and personalized therapy. Given the ability of DTECT to identify genetic variants rapidly and accurately, we tested whether DTECT could be utilized to expedite the identification of pathogenic variants in pre-clinical and clinical settings. In particular, we examined whether DTECT could identify the presence of oncogenic variants in various biological systems. In our studies we focused our attention on the JAK2 V617F variant, which is present in the majority of patients with myeloproliferative neoplasm (MPN) (Levine et al., 2005). Mice transplanted with Jak2 V617F mutant bone marrow cells develop MPN and recapitulate the human disease (Mullally et al., 2010). Therefore, we analyzed the Jak2 V617F variant in the peripheral blood of mice transplanted with a mixture of bone marrow cells that do or do not carry an inducible Jak2 V617F variant (Bhagwat et al., 2014) (
We next examined whether DTECT can identify the presence of specific oncogenic mutations in human samples from patients diagnosed with acute lymphoblastic leukemia (ALL), the most common form of childhood cancer (Inaba et al., 2013). Although most ALL patients respond to chemotherapy, ˜20% suffer a relapse as a result of resistance to chemotherapy (Bhojwani and Pui, 2013). Moreover, secondary genetic alterations that promote chemoresistance, including mutations in the NT5C2 gene (Tzoneva et al., 2018; Tzoneva et al., 2013), are found in a large fraction of ALL relapse cases (Dieck and Ferrando, 2019; Oshima et al., 2016). To test whether DTECT can identify these relapse-specific oncogenic signatures, we obtained matched DNA samples from the bone marrow of ALL patients at diagnosis and relapse and analyzed them for the presence of three common NT5C2 mutations (R238W, K359Q and R367Q) (
In this study, we established DTECT as a sensitive method for the identification of genomic DNA signatures. In particular, we show that DTECT readily identifies precision genome editing events induced by CRISPR-dependent HDR, base editing and prime editing, including low abundance and complex genomic changes. In addition, we show that DTECT can be employed to identify pathogenic lesions of interest, such as oncogenic mutations, in cancer mouse models, PDXs, and cancer patient specimens. DTECT is a rapid (˜4-5 hours) and easy-to-perform detection method that relies on standard molecular biology techniques (PCR, DNA digestion and ligation) and common laboratory reagents. This methodology is also not labor-intensive, given that it entails short periods (5-10 min) of sample processing followed by hands-free incubations. Importantly, DTECT assays utilize a unique and common set of adaptors that includes positive and negative controls to ensure specificity and accuracy. The ease, speed and cost efficiency by which DTECT identifies genetic variants in a wide variety of cellular and animal systems (e.g., cell lines, organoids, animal models, patient samples) should facilitate the generation and study of biological models of human diseases and expedite the detection of pathogenic variants for both pre-clinical and clinical applications.
Although highly robust, DTECT has three potential limitations. First, AcuI-induced dinucleotide byproducts can be generated if a genomic AcuI restriction site located in close proximity to the targeted dinucleotide is incorporated into the amplicon of the targeted locus. However, an analysis of the ClinVar database revealed that genomic AcuI sites occur relatively infrequently and 95% of clinically relevant variants (404,393 variants) are compatible with DTECT (
In addition to its ease of use, speed and cost efficiency, DTECT has several advantages compared to other detection methods. A major benefit of DTECT is its versatility, which allows the detection and quantification of nucleotide substitutions, precise base insertions and deletions using the same small set of 16 predefined adaptors (
The ability to model clinically relevant mutations in a marker-free manner is critical for assessing their potential pathogenicity, especially in the case of genes, such as BRCA1 and BRCA2, which have thousands of clinically-associated SNVs. Recent studies have led to the development of high-throughput saturation genome editing (SGE) to examine en masse the pathogenicity of BRCA1 variants (Findlay et al., 2018). Although highly useful for classifying BRCA1 SNVs, SGE requires the use of haploid cells and is therefore not compatible with the study of the functional impact of BRCA1 mutations in heterozygosity, as observed in BRCA1 mutation carriers (Apostolou and Fostira, 2013). BRCA1/2 heterozygous mutations have been recently shown to cause genome instability induced by DNA replication stress (Billing et al., 2018; Pathania et al., 2014; Tan et al., 2017). By facilitating the derivation of both heterozygous and homozygous BRCA1/2 mutant cells and animal models (
In addition to facilitating precision genome editing, we showed that DTECT can also be used to detect pathogenic variants in pre-clinical and clinical settings. In particular, DTECT can rapidly identify the presence of oncogenic variants in cancer mouse models (
Collectively, our work established DTECT as a facile, rapid and cost-effective method for identifying genomic variants in various biological systems, such as mammalian cell lines, organoids, mouse tissues, PDX models and human patient samples. Given the growing number of genetic variants identified in the human population (Lek et al., 2016) and in human genetic disorders (McClellan and King, 2010), this versatile method for the detection of genomic signatures should facilitate the study of human genetic variation and expedite the diagnosis and treatment of human disease.
All documents cited in this application are hereby incorporated by reference as if recited in full herein.
Although illustrative embodiments of the present disclosure have been described herein, it should be understood that the disclosure is not limited to those described, and that various other changes or modifications may be made by one skilled in the art without departing from the scope or spirit of the disclosure.
The present application claims benefit of U.S. Provisional Patent Application Ser. No. 62/985,746, filed on Mar. 5, 2020, which application is incorporated by reference herein in its entirety.
This invention was made with government support under grant no. GM117064, awarded by the National Institutes of Health. The government has certain rights in the invention.
Number | Name | Date | Kind |
---|---|---|---|
20160208319 | Berman et al. | Jul 2016 | A1 |
20160273023 | Jouvenot et al. | Sep 2016 | A1 |
Number | Date | Country |
---|---|---|
104894255 | Sep 2015 | CN |
2017091811 | Nov 2016 | WO |
Entry |
---|
Shen, et al. “Predictable and precise template-free CRISPR editing of pathogenic variants,” Nature 563, 646-651 (2018). |
Song, et al. “Adenine base editing in an adult mouse model of tyrosinaemia,” Nat Biomed Eng 4, 125-130 (2020). |
Tan, et al. “A Class of Environmental and Endogenous Toxins Induces BRCA2 Haploinsufficiency and Genome Instability,” Cell 169, 1105-1118 e1115 (2017). |
Tschaharganeh, et al. “p53-dependent Nestin regulation links tumor suppression to cellular plasticity in liver cancer,” Cell 158, 579-592 (2014). |
Tzoneva, et al. “Clonal evolution mechanisms in NT5C2 mutant-relapsed acute lymphoblastic leukaemia,” Nature 553, 511-514 (2018). |
Tzoneva, et al. “Activating mutations in the NT5C2 nucleotidase gene drive chemotherapy resistance in relapsed ALL,” Nat Med 19, 368-371 (2013). |
Van Overbeek, et al. “DNA Repair Profiling Reveals Nonrandom Outcomes at Cas9-Mediated Breaks,” Mol Cell 63, 633-646 (2016). |
Villiger, et al. “Treatment of a metabolic liver disease by in vivo genome base editing in adult mice,” Nat Med 24, 1519-1525 (2018). |
Wang, et al. “Enhanced base editing by co-expression of free uracil DNA glycosylase inhibitor,” Cell Res 27, 1289-1292 (2017). |
Yeh, et al. “In vivo base editing of post-mitotic sensory cells,” Nature communications 9, 2184 (2018). |
Yin, et al. “Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo,” Nature biotechnology 34, 328-333 (2016). |
Yin, et al. “Genome editing with Cas9 in adult mice corrects a disease mutation and phenotype,” Nature biotechnology 32, 551-553 (2014). |
Zafra, et al. “Optimized base editors enable efficient editing in cells, organoids and mice,” Nature biotechnology 36, 888-893 (2018). |
Zhang, et al. “Non-invasive prenatal sequencing for multiple Mendelian monogenic disorders using circulating cell-free fetal DNA,” Nat Med 25, 439-447(2019). |
Wang, et al. “CRISPR/Cas9 in Genome Editing and Beyond,” Annu Rev Biochem. Jun. 2016; 85: pp. 227-264. |
Falabella, et al. “Single-Step qPCR and dPCR Detection of Diverse CRISPR-Cas9 Gene Editing Events In Vivo,” G3 (Bethesda). Oct. 2017; 7(10): pp. 3533-3542. |
Hendel, et al. “Quantifying on- and off-target genome editing,” Trends Biotechnol. Feb. 2015; 33(2): pp. 132-140. |
Findlay, et al. “A Digital PCR-Based Method for Efficient and Highly Specific Screening of Genome Edited Cells,” PLoS One vol. 11 / Issue 4 e0153901 Apr. 2016. |
Miyaoka, et al. “Systematic quantification of HDR and NHEJ reveals effects of locus, nuclease, and cell type on genome-editing,” Scientific Reports vol. 6 23549 Mar. 2016. |
Jayavaradhan, et al. “A Versatile Tool for the Quantification of CRISPR/Cas9-lnduced Genome Editing Events in Human Hematopoietic Cell Lines and Hematopoietic Stem/Progenitor Cells,” Journal of Molecular Biology doi: 10.1016/j.jmb.2018.05.005 May 2018. |
St Martin, et al. “A fluorescent reporter for quantification and enrichment of DNA editing by APOBEC-Cas9 or cleavage by Cas9 in living cells,” Nucleic Acids Research doi: 10.1093/nar/gky332 May 2018. |
Labun, et al. “Accurate analysis of genuine CRISPR editing events with ampliCan,” BioRxiv doi: 10.1101/249474 Feb. 2018. |
Hendel, et al. “Quantifying genome-editing outcomes at endogenous loci with SMRT sequencing,” Cell Reports vol. 7 / Issue 1 pp. 293-305 Apr. 2014. |
Miyaoka, et al. “Detection and Quantification of HDR and NHEJ Induced by Genome Editing at Endogenous Gene Loci Using Droplet Digital PCR,” Methods in Molecular Biology vol. 1768 pp. 349-362 May 2018. |
Giannoukos, et al. “UDiTaS™, a genome editing detection method for indels and genome rearrangements,” BMC Genomics vol. 19 / Issue 1 212 Mar. 2018. |
Harayama and Riezman “Detection of genome-edited mutant clones by a simple competition-based PCR method,” PLoS One vol. 12 / Issue 6 e0179165 Jun. 2017. |
Fujita, et al. “Detection of genome-edited cells by oligoribonucleotide interference-PCR,” DNA Research doi: 10.1093/dnares/dsy012 Apr. 2018. |
Editas Medicine Demonstrates First Achievement of In Vivo Editing in Non-human Primate Retinas, GlobeNewswire May 13, 2017. |
Allen, et al. “Predicting the mutations generated by repair of Cas9-induced double-strand breaks,” Nature biotechnology (2018). |
Anzalone, et al. “Search-and-replace genome editing without double-strand breaks or donor DNA,” Nature (2019). |
Apostolou and Fostira “Hereditary breast cancer: the era of new susceptibility genes,” Biomed Res Int 2013, 747318 (2013). |
Barbieri, et al. “Precise Editing at DNA Replication Forks Enables Multiplex Genome Engineering in Eukaryotes,” Cell 171, 1453-1467 e1413, (2017). |
Bath, et al. “Many type IIs restriction endonucleases interact with two recognition sites before cleaving DNA,” J Biol Chem 277, 4024-4033, (2002). |
Bhagwat, et al. “Improved targeting of JAK2 leads to increased therapeutic efficacy in myeloproliferative neoplasms,” Blood 123, 2075-2083 (2014). |
Bhojwani and Pui “Relapsed childhood acute lymphoblastic leukaemia,” Lancet Oncol 14, e205-217 (2013). |
Billing, et al. “The BRCT Domains of the BRCA1 and BARD1 Tumor Suppressors Differentially Regulate Homology-Directed Repairand Stalled Fork Protection,” Mol Cell 72, 127-139 e128 (2018). |
Billon, et al. “CRISPR-Mediated Base Editing Enables Efficient Disruption of Eukaryotic Genes through Induction of STOP Codons,” Mol Cell 67, 1068-1079 e1064 (2017). |
Brinkman, et al. “Easy quantitative assessment of genome editing by sequence trace decomposition,” Nucleic Acids Res 42, e168 (2014). |
Brinkman, et al. “Easy quantification of template-directed CRISPR/Cas9 editing,” Nucleic Acids Res 46, e58 (2018). |
Canny, et al. “Inhibition of 53BP1 favors homology-dependent DNA repair and increases CRISPR-Cas9 genomeediting efficiency,” Nat Biotechnol 36, 95-102 (2018). |
Ceccaldi, et al. “The Fanconi anaemia pathway: new players and new functions,” Nat Rev Mol Cell Biol 17, 337-349 (2016). |
Chadwick, et al. “In Vivo Base Editing of PCSK9 (Proprotein Convertase Subtilisin/Kexin Type 9) as a Therapeutic Alternative to Genome Editing,” Arterioscler Thromb Vase Biol 37, 1741-1747 (2017). |
Clement, et al. “CRISPResso2 provides accurate and rapid genome editing sequence analysis,” Nature biotechnology 37, 224-226 (2019). |
Cong, et al. “Multiplex genome engineering using CRISPR/Cas systems,” Science 339, 819-823 (2013). |
Dieck and Ferrando “Genetics and mechanisms of NT5C2-driven chemotherapy resistance in relapsed ALL,” Blood (2019). |
Dow, L.E. “Modeling Disease In Vivo With CRISPR/Cas9,” Trends Mol Med 21, 609-621 (2015). |
Farzadfard and Lu “Emerging applications for DNA writers and molecular recorders,” Science 361, 870-875 (2018). |
Findlay, et al. “Accurate classification of BRCA1 variants with saturation genome editing,” Nature (2018). |
Gao, et al. “Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents,” Nature 553, 217-221 (2018). |
Gaudelli, et al. “Programmable base editing of A*T to G*C in genomic DNA without DNA cleavage,” Nature (2017). |
Germini, et al. “A Comparison of Techniques to Evaluate the Effectiveness of Genome Editing,” Trends Biotechnol 36, 147-159 (2018). |
Guo, et al. “High-throughput creation and functional profiling of DNA sequence variant libraries using CRISPR-Cas9 in yeast,” Nat Biotechnol 36, 540-546 (2018). |
Inaba, et al. “Acute lymphoblastic leukaemia,” Lancet 381, 1943-1955. |
Jasin and Haber “The democratization of gene editing: Insights from site-specific cleavage and double-strand break repair,” DNA Repair (Amst) 44, 6-16 (2016). |
Kluesner, et al. “EditR: A Method to Quantify Base Editing from Sanger Sequencing,” CRISPR J 1, 239-250 (2018). |
Komor, et al. “Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage,” Nature 533, 420-424 (2016). |
Komor, et al. “Improved base excision repair inhibition and bacteriophage Mu Gam protein yields C:G-to-T:A base editors with higher efficiency and product purity,” Sci Adv 3, eaao4774 (2017). |
Leenay, et al. “Large dataset enables prediction of repair after CRISPR-Cas9 editing in primary T cells,” Nature biotechnology 37, 1034-1037 (2019). |
Lek, et al. “Analysis of protein-coding genetic variation in 60,706 humans,” Nature 536, 285-291 (2016). |
Levine, et al. “Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis,” Cancer Cell 7, 387-397 (2005). |
Levy, et al. “Cytosine and adenine base editing of the brain, liver, retina, heart and skeletal muscle of mice via adeno-associated viruses,” Nat Biomed Eng 4, 97-110 (2020). |
Lindsay, et al. “CrispRVariants charts the mutation spectrum of genome engineering experiments,” Nat Biotechnol 34, 701-702 (2016). |
Liu, et al. “Efficient generation of mouse models of human diseases via ABE- and BE-mediated base editing,” Nat Commun 9, 2338 (2018). |
Lundin, et al. “Endonuclease specificity and sequence dependence of type IIS restriction enzymes,” PLoS One 10, e0117059 (2015). |
Mali, et al. “RNA-guided human genome engineering via Cas9,” Science 339, 823-826 (2013). |
Mashal, et al. “Detection of mutations by cleavage of DNA heteroduplexes with bacteriophage resolvases,” Nat Genet 9, 177-183 (1995). |
McClellan and King “Genetic heterogeneity in human disease,” Cell 141, 210-217 (2010). |
Mullally, et al. “Physiological Jak2V617F expression causes a lethal myeloproliferative neoplasm with differential effects on hematopoietic stem and progenitor cells,” Cancer Cell 17, 584-596 (2010). |
Oshima, et al. “Mutational landscape, clonal evolution patterns, and role of RAS mutations in relapsed acute Tymphoblastic leukemia,” Proc Natl Acad Sci USA 113, 11306-11311 (2016). |
Pathania, et al. “BRCA1 haploinsufficiency for replication stress suppression in primary cells,” Nature communications 5, 5496 (2014). |
Paulsen, et al. “Ectopic expression of RAD52 and dn53BP1 improves homology-directed repair during CRISPR-Cas9 genome editing,” Nat Biomed Eng 1, 878-888 (2017). |
Pinello, et al. “Analyzing CRISPR genome-editing experiments with CRISPResso,” Nature biotechnology 34, 695-697 (2016). |
Qiu, et al. “Mutation detection using Surveyor nuclease,” Biotechniques 36, 702-707 (2004). |
Ran, et al. “Genome engineering using the CRISPR-Cas9 system,” Nature protocols 8, 2281-2308 (2013). |
Rees and Liu “Base editing: precision chemistry on the genome and transcriptome of living cells,” Nat Rev Genet 19, 770-788 (2018). |
Roy, et al. “Multiplexed precision genome editing with trackable genomic barcodes in yeast,” Nat Biotechnol 36, 512-520 (2018). |
Ryu, et al. “Adenine base editing in mouse embryos and an adult mouse model of Duchenne muscular dystrophy,” Nature biotechnology 36, 536-539 (2018). |
Shakya, et al. “BRCA1 tumor suppression depends on BRCT phosphoprotein binding, but not its E3 ligase activity,” Science 334, 525-528 (2011). |
Number | Date | Country | |
---|---|---|---|
20210283567 A1 | Sep 2021 | US |
Number | Date | Country | |
---|---|---|---|
62985746 | Mar 2020 | US |