CELLS FOR THE PRODUCTION OF VIRUSES AND METHODS OF USING THE SAME

Information

  • Patent Application
  • 20220010285
  • Publication Number
    20220010285
  • Date Filed
    November 20, 2019
    4 years ago
  • Date Published
    January 13, 2022
    2 years ago
Abstract
The present disclosure provides compositions and methods for increasing the production of virus in vitro. Modified cells which are modified to eliminate or reduce as compared to a control cell the activity or expression of a Sterile a motif-domain containing protein 9 (SAMD9) polypeptide are provided. Methods of using these cells are also provided.
Description
SEQUENCE LISTING

A Sequence Listing accompanies this application and is submitted as a ASCII text file of the sequence listing named “169852_00056_ST25.txt” which is 25688 kb in size and was created on Nov. 18, 2019. The sequence listing is electronically submitted via EFS-Web with the application and is incorporated herein by reference in its entirety.


INTRODUCTION

Viruses are used in biotechnology applications as research tools, diagnostic agents, and therapeutic agents. As therapeutic agents, viruses may be used as vaccines, vectors for gene therapy, anti-cancer oncolytic agents, or immunotherapeutics. A viruses' usefulness in these applications, however, is partly dependent on having efficient means of producing the virus at titers that are sufficient for a particular application. There thus remains a need in the art for new viral production methods that improve viral titer.


As an exemplary virus with therapeutic potential, myxoma virus (MYXV) is a poxvirus with a narrow host range in nature, infecting only rabbits. However, wild-type MYXV and many mutant MYXVs possess oncolytic potential and have recently been shown to be an excellent immunotherapeutic agent. Their safety feature, outstanding therapeutic potential shown in many cancer types, and versatility of application approaches make them attractive for human disease therapy. To generate large quantities of MYXV has been challenging. It is largely because MYXV relies on cell-to-cell contact for the transmission of the progeny viruses. Thus conventional suspension culture cannot not provide optimal yield for MYXV production. For the same reason, monolayer cell lines that can support optimal production of other viruses supply limited viral production. Thus improving MYXV yield is a critical task for future therapeutic applications using MYXV. The present invention, for example, defines a way to significantly improve the yield of replicating MYXV in an FDA approved human cell line, A549, permitting a significantly improved viral production.


In addition to replicating MYXV, the engineered cell line permits production of a non-replicating MYXV that has a promising therapeutic potential as well. We previously found that a mutant MYXV deleted for the viral immunoregulatory gene, M062R (M062R-null MYXV) had a beneficial therapeutic effect despite an abortive infection. In human primary ovarian cancer cells M062R-null MYXV induces a potent inflammatory response that includes type I interferon (IFN-I) responses. Moreover, it effectively improved survival when it is administrated after cisplatin, the first line chemotherapy for ovarian cancer.


Although M062R-null MYXV shows therapeutic promise, it is difficult to produce in high titers that will be sufficient to conduct, for example, clinical trials because the virus produces an abortive infection. We sought to develop new compositions and methods that could be used to improve the titer of both replicating and non-replicating MYXV (e.g., M062R-null MYXV) as well as other viruses. Such compositions and methods should help realize the potential of viruses as important research tools, diagnostic agents, and therapeutic agents.


SUMMARY

In one aspect of the present invention, cells are provided. The cells may be modified to eliminate or reduce as compared to a control cell the activity or expression of a Sterile a motif-domain containing protein 9 (SAMD9) polypeptide.


In another aspect, the present invention relates to methods of producing a virus. The methods may include introducing the virus into any one of the cells described herein. Optionally, the methods may also further include purifying the virus from the cell.





BRIEF DESCRIPTION OF DRAWINGS


FIG. 1 shows a sequence alignment. The sequence alignment was created using a sequencing result from a representative clone (E1) of SAMD9-null cells that was engineered using CRISPR/CAS9 technology in human cell line A549 cells (ATCC® CCL-185™). SAMD9 mRNA: numbering is according to NCBI Reference Sequence of NM_001193307.1. SAMD9 CDS was obtained from NCBI CCDS34680.1. The reference polynucleotide sequence displayed is from NCBI Reference Sequence NM_001193307.1 (SEQ ID NO: 6). The reference amino acid (AA) sequence shown (SEQ ID NO: 7) is the amino acid sequence translated from SAMD9 mRNA. Sequencing result: The genomic DNA sequence of SAMD9 from a representative clone (E1) of SAMD9-null cells (SEQ ID NO: 8). Amino acid (AA) sequence: The amino acid sequence of SAMD9 that was converted from the sequencing result (SEQ ID NO: 9). The premature stop codon introduced by the CRISPR/CAS9 led to a production of truncated protein of 212 aa. The same deletion was found in HeLa SAMD9-null cell clones, e.g., B3.



FIG. 2 shows a western blot analysis of single cell clones that were engineered to delete SAMD9 expression. Normal expression of a neighboring gene SAMD9L is also shown along with internal loading control using β-actin. Lane 1: A549 cells expressing normal level of SAMD9; lane 2-8 are clone C1, E1, F1, G1, A2, B2, and C2, respectively.



FIG. 3 shows deleting SAMD9 from A549 cells restored M062R-null MYXV infection comparable to WT MYXV. A low multiplicity of infection (MOI) at 0.1 for either M062R-null or WT MYXV was used to infect either A549 SAMD9+/+ (parental) or SAMD9−/− (SAMD9-null) cells. At given time points, cell lysates were harvested for titration on BSC40 cells. Statistical significance was determined by multiple t tests (Holm-Sidak method) with α=5.000% defined as being statistically significant (**p<0.001, and ***p<0.0001).



FIG. 4 shows single cell clones of HeLa cells with SAMD9 deleted improved WT MYXV viral yield. In engineered SAMD9-null HeLa cell clones, WT MYXV infection is significantly improved. Using the multiple t test, we found at each given time point (7, 24, 48, and 72 hrs) other than 1 hr input both SAMD9-null cell lines support significantly increased viral yield (p<0.05).



FIGS. 5A-5D show SAMD9-null A549 cells permit significantly increased MYXV infection. We used a well-characterized entry assay7 to examine the mechanism of SAMD9-null A549 cells on how it can achieve such dramatic increase in MYXV viral infection. The WT MYXV used in this assay expressing firefly luciferase immediately after entering the cells and by examining the luciferase activity using an appropriate substrate we can measure and quantify the virus input. This is a very sensitive assay. We included many controls so that we can examine the process of infection that lead to an improved viral yield. (FIG. 5A) Inhibition of RNA synthesis after 1 hr post infection did not affect the difference in viral input. Actinomycin D (ActD) is an inhibitor for RNA synthesis. Although treating cells at 2 μg/ml during infection did reduce the overall luciferase activity compared with untreated corresponding infection, Act D treatment did not eliminate the elevated luciferase activity resulting from infecting SAMD9-null cells. (FIG. 5B) Inhibition of viral DNA replication did not reduce the improved viral infection in SAMD9-null cells. Cytosine arabinoside (AraC) is a specific inhibitor of poxvirus DNA replication. Pre-treating cells 30 minutes before infection followed by its presence during and after infection at 50 μM did not affect luciferase activity emitted from infection in both cell lines. AraC treatment at this dose inhibits post-replicative gene expression and thus reduce overall viral yield. (FIG. 5C) Inhibition of virion uncoating did not reduce the improvement in viral infection in SAMD9-null cells. MG132 is known to inhibit poxvirus virion uncoating, which occurs after virion entry. We used a dose of 10 μM to pretreat cells 30 minutes before infection, and during and throughout the rest course of infection. We found that inhibition of virion uncoating also did not affect improved infection occurred in SAMD9-null cells. (FIG. 5D) SAMD9 deletion leads to significant improvement of MYXV infection in A549 cells. A separate group of cells were infected with the same amount of virus as in A, B, and C without drug treatment and at 24 h post-infection viral yield was evaluated by titration as previously described. Using multiple t tests we found in SAMD9-null cell WT MYXV infection can be significantly improved by a log.



FIGS. 6A-6B show knocking-down SAMD9 expression in another human cell line, THP1, leads to dramatic increase of HSV-1 infection. (FIG. 6A) Knocking down SAMD9 improved HSV-1 infection. HSV-1 infection was conducted in PMA-differentiated THP1 cells (ATCC© TIB-202™) and at given time points cell lysates were collected for titration on RS cells as previously described.8 (FIG. 6B) Characterization of SAMD9 knockdown THP1 cells. The SAMD9 knockdown cells were engineered using lentivirus expressing shRNAs targeting SAMD9 mRNA and the control cells were engineered using lentivirus expressing control shRNAs; to engineer these cell lines we utilized a method similar to what has been reported.2 In the western blot, a reduced SAMD9 protein level is seen in cells stably expressing SAMD9 targeting shRNAs, while introduction of non-targeting shRNAs (control) did not affect SAMD9 expression.



FIG. 7 CRISPR-Cas9 knockout of SAMD9 with 2 different guide RNAs (gRNAs) causing inversion at the targeted region. SAMD9 sequence is a representation of the human SAMD9 CDS. Cleavage at right after both the nucleotides 526 and 1030 guided by gRNAs (red arrows) led to addition of nucleotides (red squares) during DNA repair and DNA inversion (black solid and dotted lines) in the genome. This reconfiguration of the SAMD9 sequence generated a stop codon (blue square) at the current location of 591 nucleotides, which caused SAMD9 expression knockout. The resulting truncated and mutant SAMD9 was not detectable using the SAMD9 antibody (SigmaAldrich Catalog# HPA021319) that recognizes the N-terminal of the protein (1-82 amino acids or aa). Red-underline labeled nucleotides are the PAM sequence. To illustrate the inversion, SAMD9 reverse complementary sequence between the gRNA targets was shown in comparison to SAMD9-null (B2 clone). Red squares specify inserted nucleotides with codon frameshift. The blue square indicates stop codon at nucleotide position 591.





DETAILED DESCRIPTION

Here, in the non-limiting Examples, the present inventors have created cells and methods that improve the production of myxoma viruses, including wild-type replication-competent myxoma viruses and replication-defective mutant myxoma viruses. In addition to myxoma viruses, the present inventors show that the disclosed cells and methods also improve the production of other viruses such as, without limitation, herpes simplex viruses, poxviruses, reoviruses and rota viruses.


Cells

In one aspect of the present invention, cells are provided. The cells may be modified to eliminate or reduce as compared to a control cell the activity or expression of a Sterile a motif-domain containing protein 9 (SAMD9) polypeptide.


As used herein, the terms “polypeptide” or “protein” or “peptide” may be used interchangeably to refer to a polymer of amino acids. A “polypeptide” as contemplated herein typically comprises a polymer of naturally occurring amino acids (e.g., alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine).


Sterile a motif-domain containing protein 9 (SAMD9) polypeptides play critical roles in human health and may play a role in the inflammatory response to tissue injury and the control of extra-osseous calcification. The present inventors previously found that SAMD9 has antiviral function against poxviruses and poxviruses have evolved many viral proteins to inhibit SAMD9 function.1, 2


SEQ ID NO: 1 is an exemplary SAMD9 polypeptide from humans that may be used as a reference sequence. SAMD9 polypeptides from other species may also be used as reference sequences and can be found by a person of ordinary skill in the art by using SEQ ID NO: 1 to identify similar sequences from other species in databases such as at the National Center for Biotechnology Information (NCBI). The polypeptides disclosed herein (i.e., SAMD9 polypeptides and M062R polypeptides) may include “homologs” of the polypeptides disclosed herein. A “homolog” may be a protein related to a second protein by descent from a common ancestral protein.


The polypeptides disclosed herein (i.e., SAMD9 polypeptides and M062R polypeptides) may include “variants” of the polypeptides described herein. As used herein, a “variant” refers to a protein having an amino acid sequence that differs from a reference sequence. A variant may have one or more insertions, deletions, or substitutions of an amino acid residue relative to a reference sequence. For example, an SAMD9 polypeptide variant may have one or more insertion(s), deletion(s), or substitution(s) of at least one amino acid residue relative to the reference SAMD9 polypeptide (SEQ ID NO: 1) disclosed herein. In some embodiments, the SAMD9 polypeptide may include a polypeptide having at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 1 (human SAMD9 polypeptide sequence).


Regarding the polypeptides disclosed herein, the phrases “% sequence identity,” “percent identity,” or “% identity” refer to the percentage of residue matches between at least two amino acid sequences aligned using a standardized algorithm. Methods of amino acid sequence alignment are well-known in the art. A suite of commonly used and freely available sequence comparison algorithms is provided by the National Center for Biotechnology Information (NCBI) Basic Local Alignment Search Tool (BLAST), which is available from several sources, including the NCBI, Bethesda, Md., at its website. The BLAST software suite includes various sequence analysis programs including “blastp,” that may be used to align a known amino acid sequence with other amino acids sequences from a variety of databases.


Polypeptide sequence identity may be measured over the length of an entire defined polypeptide sequence, for example, as defined by a particular SEQ ID number, or may be measured over a shorter length, for example, over the length of a fragment taken from a larger, defined polypeptide sequence, for instance, a fragment of at least 15, at least 20, at least 30, at least 40, at least 50, at least 70 or at least 150 contiguous residues. Such lengths are exemplary only, and it is understood that any fragment length supported by the sequences shown herein, in the tables, figures or Sequence Listing, may be used to describe a length over which percentage identity may be measured.


A “deletion” in a polypeptide described herein refers to a change in the amino acid sequence resulting in the absence of one or more amino acid residues. A deletion may remove at least 1, 2, 3, 4, 5, 10, 20, 50, 100, 200, or more amino acids residues. A deletion may include an internal deletion and/or a terminal deletion (e.g., an N-terminal truncation, a C-terminal truncation or both of a reference polypeptide).


“Insertions” and “additions” in a polypeptide described herein refers to changes in an amino acid sequence resulting in the addition of one or more amino acid residues. An insertion or addition may refer to 1, 2, 3, 4, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, or more amino acid residues. A variant or homolog of a polypeptide described herein may have N-terminal insertions, C-terminal insertions, internal insertions, or any combination of N-terminal insertions, C-terminal insertions, and internal insertions.


Suitable “cells” in accordance with the present invention may include eukaryotic cells. Suitable eukaryotic cells include, without limitation, animal cells. In some embodiments, the cell is a mammalian cell such as, without limitation, a mouse cell, a rat cell, a hamster cell, or a human cell. The cell may be a cell line used to produce viral particles or viruses including, without limitation, mammalian cell lines such as A549 cells, THP1 cells, HeLa cells, CHO cells, HEK293 cells, HEK293T cells, COS cells, BK cells, MDCK cells, PER C6 cells, HKB-11 cells, CAP (CEVEC's Amniocyte Production) cells, Huh 7 cells, human diploid cells, HT-1080 cells, and Vero cells.


The eliminated or reduced activity or expression of the SAMD9 polypeptide is relative to a control cell. A “control cell” is a wild-type cell that has not been modified as described herein and may include a wild-type SAMD9 gene encoding a wild-type SAMD9 polypeptide. Exemplary control cells may include wild-type A549 cells, THP1 cells, or HeLa cells. The control cell may be the same type of cell as the cell modified to eliminate or reduce the activity or expression of a SAMD9 polypeptide.


As used herein, the “activity” of a SAMD9 polypeptide refers to any of the biological functions of a SAMD9 polypeptide including, without limitation, inflammatory or anti-viral functions. In some embodiments, the activity of the SAMD9 polypeptide is reduced by at least 30%, 40%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99%, or more as compared to a control cell.


As used herein, the term “expression” may refer either to the levels of an RNA encoding a protein in a cell or the levels of the protein in a cell.


In some embodiments, the expression of the SAMD9 polypeptide is reduced by at least 30%, 40%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99%, or more as compared to a control cell. In a preferred example, the SAMD9 is reduced at least 60% or more, preferably 80% or more.


The cells may be modified to eliminate or reduce as compared to a control cell the activity or expression of a SAMD9 polypeptide described herein. As used herein, the terms “modified” or “modifying” refer to using any laboratory methods available to those of skill in the art including, without limitation, genetic engineering techniques (i.e. CRISPR/Cas techniques or gene silencing technologies) or forward genetic techniques to affect the activity or expression of a SAMD9 polypeptide. It will be readily apparent to one of ordinary skill in the art that there are multiple potential ways to eliminate or reduce the activity or expression of a SAMD9 polypeptide in a cell by modifying the gene encoding the SAMD9 polypeptide by, for example, introducing targeted mutations, by modifying a mRNA (or levels thereof) encoding the SAMD9 polypeptide, for example, by using gene silencing techniques, or by inhibiting the SAMD9 polypeptide at the protein level.


In some embodiments, the cell may include a nucleic acid agent capable of downregulating an RNA transcript encoding the SAMD9 polypeptide. Suitable nucleic acid agents may include, without limitation, a short hairpin RNA (shRNA), microRNA, siRNA, or anti sense RNA.


As exemplarily, nucleic acid agents capable of downregulating an RNA transcript encoding the SAMD9 polypeptide, in the Examples, the inventors use a short hairpin RNA (shRNA) targeting SAMD9 mRNAs in THP1 cells.


The cell may also be modified to introduce a hypomorphic mutation or a null mutation in a polynucleotide (i.e., gene) encoding the SAMD9 polypeptide. A “null mutation” is an alteration in a gene that results in a gene that completely lacks its normal function. The complete lack of function may be the result of the complete absence of a gene product (i.e., protein or RNA) being produced in a cell or may result from the expression of a non-functional polypeptide. Similarly, a “hypomorphic mutation” is an alteration in a gene that results in a gene that has reduced activity. The reduced activity may be from a reduced level of expression of gene products (i.e., protein or RNA) from the gene or may result from the expression of a gene product (i.e. protein or RNA) that has reduced functional activity. For example, the gene product (i.e., protein or RNA) may result in a truncated form of the SAMD9 protein that has reduced or eliminated all functionality within a cell, as demonstrated in the examples.


It will be readily apparent to those of skill in the art that a variety of null or hypomorphic mutations may be introduced (using, for example, CRISPR/Cas or other genetic engineering techniques) into a polynucleotide encoding the SAMD9 polypeptides described herein to arrive at embodiments of the present invention. For example, early stop codons may be introduced into the open reading frame of the gene encoding the SAMD9 polypeptide, which would result in the expression of a shorter protein sequence completely lacking or having reduced activity. Alternatively or additionally, a person of ordinary skill may introduce alterations (i.e., substitutions or deletions) into the promoter of a gene encoding the SAMD9 polypeptide described herein that result in little or no expression of the SAMD9 polypeptide.


As exemplarily null or hypomorphic mutations that may be introduced into a gene encoding a SAMD9 polypeptide described herein, in the Examples, the inventors use CRISPR/Cas9 molecular tools to introduce a premature stop codon into the SAMD9 gene, which led to the production of a truncated protein (see, e.g., FIG. 1 wherein the introduction of a single nucleotide caused a frame shift and premature stop codon). CRISPR/Cas9 molecular tools was also used to knockout of SAMD9 with 2 different guide RNAs (gRNAs) causing inversion at the targeted region as demonstrated in the Examples and FIG. 7. Thus, suitable cells may produce a truncated form of the SAMD9 protein that has reduced or eliminated its activity.


Still further modifications contemplated herein include mutations that impact one or more of the activities of the SAMD9 polypeptide. The SAMD9 polypeptide has inflammatory and anti-viral functional activities. It will be understood by those of skill in the art that alterations (i.e., mutations and/or deletions) could be made to the SAMD9 polypeptide that would be expected to eliminate or reduce an activity of the SAMD9 polypeptide.


Methods

In another aspect, the present invention relates to methods of producing a virus. The methods may include introducing the virus into any of the cells described herein.


The virus may be “introduced” into the cell by, for example, contacting the cell with viral particles or introducing a polynucleotide including a viral polynucleotide into the cell (i.e., DNA plasmid). Methods of introducing polynucleotides into a cell are known in the art and may include, without limitation, microinjection, transformation, and transfection methods. Transformation or transfection may occur under natural or artificial conditions according to various methods well known in the art, and may rely on any known method for the insertion of foreign nucleic acid sequences into a host cell. The method for transformation or transfection is selected based on the type of host cell being transformed and may include, but is not limited to, bacteriophage or viral infection, electroporation, heat shock, lipofection, and particle bombardment. Microinjection of polynucleotides may also be used to introduce polynucleotides into cells.


In the nonlimiting Examples, the present inventors introduce viruses (i.e., myxoma and herpes simplex viruses) into the cells described herein by incubating cells with viruses on ice, at room temperature, or at 37° C. in regular medium for an hour followed by removing inoculum and replishing with standard growth medium for infection development. For myxoma and herpes simplex viruses, the present inventors used natural infections in conventional cell culture.


The cells described herein, e.g., cells with a modified SAMD9 (e.g., SAM9D null cells or truncated non-active or reduced SAMD9 activity), are shown to produce an increase in virus infectivity and thus the ability to increase the production of the amount of virus made in vitro for laboratory and clinical purposes.


Optionally, the methods may also further include purifying the virus from the cell. The term “purifying” is used to refer to the process of ensuring that the virus or viral particles are substantially or essentially free from cellular components and/or other impurities. Purification of viruses or viral particles is typically performed using molecular biology and analytical chemistry techniques such as, without limitation, ultracentrifugation, density gradient (i.e., sucrose gradient), chromatography, precipitation and/or filtration. Methods of purifying viruses or viral particles are well known to those skilled in the art. A “purified” virus or viral particles means that the virus or viral particles is at least 85% pure, more preferably at least 95% pure, and most preferably at least 99% pure.


As used herein, a “virus” may be any virus having DNA or RNA genomes. Suitable viruses may include, without limitation, myxoma viruses, herpes simplex viruses, rotaviruses, reoviruses, influenza viruses or adenoviruses.


As shown in the examples, the altered SAMD9 cells are able to substantially increase the viral titer production for a number of viruses within the in vitro cultures. The present cells are able to increase the virus titer production by at least 1 log (10 fold), in some examples, at least 2 log (100 fold) as compared to the wildtype cells. For example, for rotaviruses, a greater than a log increase in virus titers (e.g., number of viral genomes produced by the cells) was seen. In some examples, the altered SAMD9 cells were able to produce a greater than 2 log increase in virus production with the altered SAMD9 cells (ΔSAMD9 HeLa cells) (about 100 fold increase) as compare to wildtype cells. In some examples, the virus titer was able to go from an undetectable level within wildtype cells to a detectable level (e.g. 103 genomes) when using the altered SAMD9 cells.


Altering the SAMD9 production in any cell was able to increase the ability of that altered cell to produce viruses. The inventors discovered that the greater the reduction of SAMD9 activity within the cells, the greater an increase in the ability to produce virus (i.e. increase virus production and titer). For example, in one exemplary cell type, a 60% reduction of SAM9 activity within a cell resulted, in some examples, in a log (10 fold) increase in virus titer.


Viral titer, viral particles, or infectious particles in a given volume (e.g., per mL) can be measured by any method known in the art. For example, the number of genomes can be quantitated within the supernatant from the altered cells using methods known in the art. For example, nucleic acid based tests can be used (i.e. RNA copies per mL), which include, but are not limited to, for example, PCR, RT-PCR, viral plaque assay, etc., tittering assay (e.g., serial dilution, etc.), and are well within one skilled in the art to determine.


In some embodiments, the virus may include a mutation affecting the growth of the virus (i.e., replication-incompetent). Exemplary viruses including a mutation affecting the growth of the viruses may include, without limitation, myxoma viruses including a hypomorphic or a null mutation in a M062R polynucleotide encoding a M062R polypeptide. See Liu et al., I Virol. 85(7):3270-3282 (2011) and International Application No. PCT/US19/33973 all of which are incorporated herein by reference in their entirety. Myxoma virus protein M062R is an immunoregulatory protein in myxoma viruses. M062R is a functional homolog of the C7L family of host range genes from orthpoxviruses and is required for MYVX replication in most human cancer cells.


Exemplary viruses including a mutation affecting the growth of the viruses may include, without limitation, mutant viruses that lose an inhibitor of SAMD9 activity.


The present disclosure is not limited to the specific details of construction, arrangement of components, or method steps set forth herein. The compositions and methods disclosed herein are capable of being made, practiced, used, carried out and/or formed in various ways that will be apparent to one of skill in the art in light of the disclosure that follows. The phraseology and terminology used herein is for the purpose of description only and should not be regarded as limiting to the scope of the claims. Ordinal indicators, such as first, second, and third, as used in the description and the claims to refer to various structures or method steps, are not meant to be construed to indicate any specific structures or steps, or any particular order or configuration to such structures or steps. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to facilitate the disclosure and does not imply any limitation on the scope of the disclosure unless otherwise claimed. No language in the specification, and no structures shown in the drawings, should be construed as indicating that any non-claimed element is essential to the practice of the disclosed subject matter. The use herein of the terms “including,” “comprising,” or “having,” and variations thereof, is meant to encompass the elements listed thereafter and equivalents thereof, as well as additional elements. Embodiments recited as “including,” “comprising,” or “having” certain elements are also contemplated as “consisting essentially of” and “consisting of” those certain elements.


Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. For example, if a concentration range is stated as 1% to 50%, it is intended that values such as 2% to 40%, 10% to 30%, or 1% to 3%, etc., are expressly enumerated in this specification. These are only examples of what is specifically intended, and all possible combinations of numerical values between and including the lowest value and the highest value enumerated are to be considered to be expressly stated in this disclosure. Use of the word “about” to describe a particular recited amount or range of amounts is meant to indicate that values very near to the recited amount are included in that amount, such as values that could or naturally would be accounted for due to manufacturing tolerances, instrument and human error in forming measurements, and the like. All percentages referring to amounts are by weight unless indicated otherwise.


No admission is made that any reference, including any non-patent or patent document cited in this specification, constitutes prior art. In particular, it will be understood that, unless otherwise stated, reference to any document herein does not constitute an admission that any of these documents forms part of the common general knowledge in the art in the United States or in any other country. Any discussion of the references states what their authors assert, and the applicant reserves the right to challenge the accuracy and pertinence of any of the documents cited herein. All references cited herein are fully incorporated by reference in their entirety, unless explicitly indicated otherwise. The present disclosure shall control in the event there are any disparities between any definitions and/or description found in the cited references.


Unless otherwise specified or indicated by context, the terms “a”, “an”, and “the” mean “one or more.” For example, “a protein” or “an RNA” should be interpreted to mean “one or more proteins” or “one or more RNAs,” respectively.


The following examples are meant only to be illustrative and are not meant as limitations on the scope of the invention or of the appended claims.


EXAMPLES
Example 1

Sterile α motif-domain containing protein 9 (SAMD9) is an important protein that plays critical roles in human health. We previously found that SAMD9 has antiviral function against poxviruses and poxviruses have evolved many viral proteins to inhibit SAMD9 function.1, 2 During infection by myxoma virus (MYXV) one viral protein M062 takes the role to neutralize SAMD9. Without M062, MYXV infection cannot proceed resulting in an abortive infection. This is a form of infection in which viral genome replication is not complete and expression of many viral genes cannot be accomplished. However, by knocking down the mRNA level of SAMD9, we can restore partially the infection by a M062R-null MYXV.1 Interestingly, we found that in human A549 cells by deleting SAMD9 expression, we not only can restore M062R-null MYXV infection to the same levels as the replicating MYXV but also can drastically improve the viral yield of replicating MYXV (e.g., WT virus). We also found that knocking down SAMD9 mRNA level led to improved viral yield by herpes simplex virus type 1 (HSV-1). Thus cells without SAMD9 expression can be very useful for the production of viruses (e.g., MYXV and HSV-1) for clinical applications.


We engineered SAMD9-null cells with CRISPR/CAS9 technology and one example on how to engineer the cell line is described below. We first designed the guide RNAs (gRNAs) and chose two options as following: gRNA1-Forward: 5′-CACCGtaatccatatcgttacaagt-3′ (SEQ ID NO: 2); gRNA1-Reverse: 5′-AAACacttgtaacgatatggattaC-3′ (SEQ ID NO: 3); gRNA2-Forward: 5′-CACCGtctcactatttgtgcgagat-3′ (SEQ ID NO: 4); gRNA2-Reverse: 5′-AAACatctcgcacaaatagtgagaC-3′ (SEQ ID NO: 5). We cloned gRNA1 and 2 into vector pX330-U6-Chimeric_BB-CBh-hSpCas9 (Addgene plasmid #42230)3 and pSpCas9(BB)-2A-GFP (PX458) (Addgene plasmid #48138)4, respectively. After cotransfection of both constructs, single cells that turned green were individually collected via cell sorting and cultured to expand. Western blot was conducted to confirm the absence of SAMD9 protein as previously described.5 Briefly, both plasmids expressing gRNA1 and 2 are transfected into cells and followed by cell sorting of GFP positive cells at 24 h post-transfection. GFP expression indicated that transfected cells expressed at least Cas9-GFP and gRNA 2. It is most likely that transfected cells received both sets of Cas9 and gRNA. Single clones are obtained in 96-well-plates for amplification and a pooled cell population was also obtained and frozen as previously described.6 The GFP expression was only transient and the plasmids expressing Cas9-gRNA were lost in the cells while cells continued to divide. Once the single cell clone is confirmed to no longer express SAMD9 protein, we sequenced the genomic DNA of SAMD9 to confirm the genetic manipulation was successful. We found that the most frequent cause of SAMD9 deletion was caused by cleavage guided by gRNA1 and the example is shown in FIG. 1, which is from a single cell clone of A549 SAMD9-null cells. The Western blot result is shown in FIG. 2.


We next tested M062R-null cell infection in these cells and found that viral replication was completely restored to comparable levels of WT MYXV infection (FIG. 3).


We also found that once SAMD9 is deleted from human cells, WT myxoma virus (MYXV) infection is also significantly improved. See, e.g., FIG. 4 and FIGS. 5A-5D. For example in SAMD9-null cells of HeLa and A549 cells, WT MYXV replication increased ranging from 5 fold to up to a log (FIG. 4 and FIG. 5D).


We also tested whether knocking-down SAMD9 expression in another human cell line, THP1, could increase HSV-1 infection. HSV-1 infection was conducted in PMA-differentiated THP1 cells (ATCC© TIB-202™) and at given time points cell lysates were collected for titration on RS cells as previously described.8 SAMD9 knockdown cells were engineered using lentivirus expressing shRNAs targeting SAMD9 mRNA and the control cells were engineered using lentivirus expressing control shRNAs; to engineer these cell lines we utilized a method similar to what has been reported.2 Reduced SAMD9 protein levels were observed in cells stably expressing SAMD9 targeting shRNAs, while introduction of non-targeting shRNAs (control) did not affect SAMD9 expression. See FIG. 6B. We found that knocking-down SAMD9 improved HSV-1 infection. See FIG. 6A.



FIG. 7 describes a second method of knocking down using CRISPR-Cas9 knockout of SAMD9 with 2 different guide RNAs (gRNAs) causing inversion at the targeted region. SAMD9 sequence is a representation of the human SAMD9 CDS. Cleavage at right after both the nucleotides 526 and 1030 guided by gRNAs (red arrows) led to addition of nucleotides (red squares) during DNA repair and DNA inversion (black solid and dotted lines) in the genome. This reconfiguration of the SAMD9 sequence generated a stop codon (blue square) at the current location of 591 nucleotides, which caused SAMD9 expression knockout. The resulting truncated and mutant SAMD9 was not detectable using the SAMD9 antibody (SigmaAldrich Catalog# HPA021319) that recognizes the N-terminal of the protein (1-82 amino acids or aa). Red-underline labeled nucleotides are the PAM sequence. To illustrate the inversion, SAMD9 reverse complementary sequence between the gRNA targets was shown in comparison to SAMD9-null (B2 clone). Red squares specify inserted nucleotides with codon frameshift. The blue square indicate stop codon at nucleotide position 591.


Materials and Methods
Cell Lines and Viruses

A549 (ATCC® CCL-185™), THP1 (ATCC© TIB-202™), BSC40 (ATCC® CRL-2761) and HeLa (ATCC® 2™) were obtained from ATCC. While THP1 cells have been cultured in RMPI1640 (Lonza, BioWhittaker) supplemented with 10% fetal bovine serum (FBS; Atlantic Biological) and 100 μg of penicillin-streptomycin (pen/strep; Invitrogen)/ml, A549, BSC-40 and HeLa are cultured in Dulbecco minimum essential medium (DMEM; Invitrogen) supplemented with 10% FBS, 2 mM glutamine (Invitrogen) and 100 μg of pen/Sterp per ml. RS cells and HSV-1 (17syn+) were kindly provided by Dr. David Bloom (University of Florida). HSV-1 was amplified in RS cells and the titer is determined on RS cells using plaque assay as previously described (Liu et al., 2008). Wildtype MYXV and M062R-null MYXV have been reported previously (Liu et al., 2011). These MYXVs were amplified on BSC-40 and titers were determined on BSC-40 cells as previous described (Liu et al., 2011).


Antibodies and Reagents

Antibodies for SAMD9 (SigmaAldrich), SAMD9L (SigmaAldrich), β-actin (SigmaAldrich), goat anti-rabbit IgG-HRP, and goat anti-mouse IgG-HRP were used for the Western Bloting. Actinomycin D, cytosine beta-D-arabinofuranoside, MG132 (Z-Leu-Leu-Leu-al) were purchased from SigmaAldrich and resuspended for storage according to Cold Spring Harbor Protocols. Luciferase activity was detected using Luciferase Assay System (Promega).


Sequencing to Confirm SAMD9 Deletion

Cell genomic DNA was extracted using PureLink Genomic DNA Mini Kit (Invitrogen). PCR amplification of SAMD9 intron free region using Phusion High-Fidelity DNA polymerase (New England Biolabs) and the primer set of SAMD9F78 (5′-AGTCggtaccGCCATTGAAGATTCGATT-3′) (SEQ ID NO: 10) and SAMD9R724 (5′-GACTctcgagttaAGAATCTGCACAGTTTTGAA-3′) (SEQ ID NO: 11) to generate a fragment of approximately 1938 nt. This DNA fragment was then cloned into a plasmid using Zero Blunt TOPO cloning kit (ThermoFisher Scientific). Sequencing was conducted using M13 forward and reverse primers.


REFERENCES



  • 1. Liu J, Wennier S, Zhang L, McFadden G. M062 is a host range factor essential for myxoma virus pathogenesis and functions as an antagonist of host SAMD9 in human cells. Journal of virology 2011, 85(7): 3270-3282.

  • 2. Liu J, McFadden G. SAMD9 is an innate antiviral host factor with stress response properties that can be antagonized by poxviruses. Journal of virology 2015, 89(3): 1925-1931.

  • 3. Cong L, Ran F A, Cox D, Lin S, Barretto R, Habib N, et al. Multiplex genome engineering using CRISPR/Cas systems. Science (New York, N.Y.) 2013, 339(6121): 819-823.

  • 4. Ran F A, Hsu P D, Wright J, Agarwala V, Scott D A, Zhang F. Genome engineering using the CRISPR-Cas9 system. Nature protocols 2013, 8(11): 2281-2308.

  • 5. Nounamo B, Li Y, O'Byrne P, Kearney A M, Khan A, Liu J. An interaction domain in human SAMD9 is essential for myxoma virus host-range determinant M062 antagonism of host anti-viral function. Virology 2017, 503: 94-102.

  • 6. Bauer D E, Canver M C, Orkin S H. Generation of genomic deletions in mammalian cell lines via CRISPR/Cas9. Journal of visualized experiments: JOVE 2015(95): e52118.

  • 7. Villa N Y, Bartee E, Mohamed M R, Rahman M M, Barrett J W, McFadden G. Myxoma and vaccinia viruses exploit different mechanisms to enter and infect human cancer cells. Virology 2010, 401(2): 266-279.

  • 8. Liu J, Lewin A S, Tuli S S, Ghivizzani S C, Schultz G S, Bloom D C. Reduction in severity of a herpes simplex virus type 1 murine infection by treatment with a ribozyme targeting the UL20 gene RNA. Journal of virology 2008, 82(15): 7467-7474.


Claims
  • 1. A cell modified to eliminate or reduce as compared to a control cell the activity or expression of a Sterile α motif-domain containing protein 9 (SAMD9) polypeptide.
  • 2. The cell of claim 1, wherein the SAMD9 polypeptide comprises a polypeptide having at least 80% sequence identity to SEQ ID NO: 1 (human SAMD9 polypeptide sequence)
  • 3. The cell of claim 1, wherein the activity or expression of the SAMD9 polypeptide is reduced by at least 30% as compared to a control cell.
  • 4. The cell of claim 1, wherein the activity or expression of the SAMD9 polypeptide is reduced by at least 60% as compared to a control cell.
  • 5. The cell of claim 1, wherein the cell comprises a hypomorphic or a null mutation in a SAMD9 polynucleotide encoding the SAMD9 polypeptide.
  • 6. The cell of claim 1, wherein the cell comprises a nucleic acid agent capable of downregulating an RNA transcript encoding the SAMD9 polypeptide.
  • 7. The cell of claim 6, wherein the nucleic acid agent is selected from the group consisting of a shRNA, microRNA, siRNA, and antisense RNA.
  • 8. The cell of claim 1, wherein the cell comprises a truncated SAMD9 which has reduced or eliminated SAMD9 activity within the cell.
  • 9. The cell of claim 8, wherein the cell comprises truncated SAMD9 with a nucleic acid sequence comprising a premature stop codon.
  • 10. The cell of claim 9, wherein the premature stop codon is at position 638-640 or position 589-591 of the nucleic acid coding sequence of SEQ ID NO:6 or at position 807-809 or position 758-760 of the mRNA of SEQ ID NO: 12, which results in a truncated SAMD9 protein.
  • 11. The cell of claim 1, wherein the cell is a mammalian cell.
  • 12. The cell of claim 11, wherein the cell is an A549 cell, a THP1 cell, or a HeLa cell.
  • 13. A method of producing a virus comprising introducing the virus into any one of the cells of claim 1.
  • 14. The method of claim 13, further comprising purifying the virus from the cells.
  • 15. The method of claim 12, wherein the virus is a myxoma virus, a herpes simplex virus, a rotavirus, a reovirus, influenza virus, or an adenovirus.
  • 16. The method of claim 13, wherein the virus comprises a mutation affecting the growth of the virus.
  • 17. The method of claim 16, wherein the virus is a myxoma virus comprising a hypomorphic or a null mutation in a M062 polynucleotide encoding a M062 polypeptide.
  • 18. The method of claim 17, wherein the myxoma viruses comprises a null mutation in the M062 polynucleotide encoding the M062 polypeptide.
  • 19. The method of claim 13, wherein the cells produce at least a log increase in virus titer as compared with a control cell.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims benefit to U.S. Provisional Application No. 62/769,834 filed on Nov. 20, 2018, the contents of which are incorporated by reference in its entirety.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH

This invention was made with government support under R01 AI139106 and K22-AI099184 awarded by the National Institutes of Health. The government has certain rights in the invention.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2019/062470 11/20/2019 WO 00
Provisional Applications (1)
Number Date Country
62769834 Nov 2018 US