CORRECTION OF DYSTROPHIN EXON 43, EXON 45, OR EXON 52 DELETIONS IN DUCHENNE MUSCULAR DYSTROPHY

Abstract
Duchenne muscular dystrophy (DMD), which affects 1 in 5,000 male births, is one of the most common genetic disorders of children. This disease is caused by an absence or deficiency of dystrophin protein in striated muscle. The major DMD deletion “hot spots” are found between exon 6 to 8, and exons 45 to 53. Here, three DMD mouse models are provided that can be used to test a variety of DMD exon skipping and refraining strategies. Among these are, CRISPR/Cas9 oligonucleotides, small molecules or other therapeutic modalities that promote exon skipping or exon refraining or micro dystrophin mini genes or cell based therapies. Methods for restoring the reading frame of exon 43, exon 45, and exon 52 deletion via CRISPR-mediated exon skipping and refraining in the humanized DMD mouse model, in patient-derived iPSCs and ultimately, in patients using various delivery systems are also contemplated. The impact of CRISPR technology on DMD is that gene editing can permanently correct mutations.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically is hereby incorporated by reference in its entirety. The sequence listing was created on Jun. 20, 2019, is named UTFD_P3391WO.txt and is ˜6,540,494 bytes in size.


FIELD

The present disclosure relates to the fields of molecular biology, medicine and genetics. More particularly, the disclosure relates to the use of genome editing to create humanized animal models for different forms of Duchenne muscular dystrophy (DMD), each containing distinct DMD mutations.


BACKGROUND

Muscular dystrophies (MD) are a group of more than 30 genetic diseases characterized by progressive weakness and degeneration of the skeletal muscles that control movement. Duchenne muscular dystrophy (DMD) is one of the most severe forms of MD that affects approximately 1 in 5000 boys and is characterized by progressive muscle weakness and premature death. Cardiomyopathy and heart failure are common, incurable and lethal features of DMD. The disease is caused by mutations in the gene encoding dystrophin (DMD), a large intracellular protein that links the dystroglycan complex at the cell surface with the underlying cytoskeleton, thereby maintaining integrity of the muscle cell membrane during contraction. Mutations in the dystrophin gene result in loss of expression of dystrophin causing muscle membrane fragility and progressive muscle wasting.


There remains a need in the art for treatments and cures for DMD, and for mouse models to be used for developing the same.


SUMMARY

In accordance with the present disclosure, there is provided a nucleic acid comprising a sequence encoding a single guide RNA (sgRNA) comprising a spacer sequence and a scaffold sequence; wherein the spacer sequence comprises the sequence of any one of SEQ ID NOs: 135-146, 170-260, 337-339, or 617. The scaffold sequence may comprise, for example, the sequence of any one of SEQ ID NO: 147-153. In some embodiments, the nucleic acid comprises one copy of the sequence encoding the sgRNA. In some embodiments, the nucleic acid comprises two, three, four, or five copies of the sequence encoding the sgRNA. The nucleic acid may comprise a sequence encoding a promoter, wherein the promoter drives expression of the sgRNA. In some embodiments, the nucleic acid comprises three copies of the sequence encoding the sgRNA, wherein the nucleic acid comprises a sequence encoding a first promoter and expression of the first copy of the sgRNA is driven by the first promoter, wherein the nucleic acid comprises a sequence encoding a second promoter and expression of the second copy of the sgRNA is driven by the second promoter, and wherein the nucleic acid comprises a sequence encoding a third promoter and expression of the third copy of the sgRNA is driven by the third promoter. In some embodiments, the nucleic acid further comprises a sequence encoding a nuclease. In some embodiments, the nuclease is a Type II, Type V-A, Type V-B, Type V-C, Type V-U, or Type VI-B nuclease. In some embodiments, the nuclease is a TAL nuclease, a meganuclease, or a zinc-finger nuclease. In some embodiments, the nuclease is a Cas9, Cas12a, Cas12b, Cas12c, Tnp-B like, Cas13a (C2c2), Cas13b, or Cas14 nuclease. The Cas9 nuclease may be, for example, a Streptococcus pyogenes or Streptococcus aureus Cas9. In some embodiments, the Cas9 nuclease is a modified Cas9 nuclease, such as a modified Streptococcus pyogenes Cas9 or a modified Streptococcus aureus Cas9.


Also provided is a recombinant vector comprising a nucleic acid of the disclosure. In some embodiments, the recombinant vector is an expression vector. In some embodiments, the recombinant vector is a viral vector. In some embodiments, the viral vector is a lentiviral vector, a retroviral vector, an adenoviral vector, or an adeno-associated virus (AAV) vector. In some embodiments, the viral vector is an adeno-associated virus (AAV) vector. The AAV vector may have a serotype of, for example, AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVRh74, AAV2i8, AAVRh10, AAV39, AAV43, AAVRh8, avian AAV, bovine AAV, canine AAV, equine AAV, or ovine AAV. In some embodiments, the serotype of the AAV vector is AAV9. In some embodiments, the AAV vector is replication-defective or conditionally replication defective.


Also provided is a non-viral vector comprising a nucleic acid of the disclosure. The non-viral vector may comprise calcium phosphate, liposomes, nanoparticles, and/or lipid emulsions.


Also provided is an AAV expression cassette comprising a first inverted terminal repeat (ITR); a first promoter; a nucleic acid of the disclosure; and a second ITR. In some embodiments, the AAV expression cassette further comprises a polyadenosine (polyA) sequence. In some embodiments, one or both of the first ITR and the second ITR are isolated or derived from any one of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVRh74, AAV2i8, AAVRh10, AAV39, AAV43, AAVRh8, avian AAV, bovine AAV, canine AAV, equine AAV, or ovine AAV.


Also provided is an AAV vector comprising a nucleic acid or an AAV expression cassette of the disclosure. In some embodiments, the AAV vector has the serotype of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVRh74, AAV2i8, AAVRh10, AAV39, AAV43, AAVRh8, avian AAV, bovine AAV, canine AAV, equine AAV, or ovine AAV. In some embodiments, the serotype of the AAV vector is AAV9. In some embodiments, the AAV vector is replication-defective or conditionally replication defective.


Also provided is a composition comprising a nucleic acid, an AAV expression cassette, or an AAV vector of the disclosure. In some embodiments, the composition further comprises a pharmaceutically acceptable carrier.


Also provided is a cell comprising a nucleic acid, an expression cassette, an AAV vector, or a composition of the disclosure. In some embodiments, the cell is a stem cell. In some embodiments, the cell is a mammalian cell such as a human cell.


Also provided is a method of correcting a gene defect in a cell, the method comprising contacting the cell with a nucleic acid, a recombinant vector, a non-viral vector, an AAV vector, or a composition of the disclosure. In some embodiments, the cell is a stem cell. In some embodiments, the cell is a mammalian cell such as a human cell.


Also provided is a method of treating a subject suffering from Duchenne muscular dystrophy, the method comprising administering to the subject a therapeutically effective amount of a nucleic acid, a recombinant vector, a non-viral vector, an AAV vector, or a composition of the disclosure.


Also provided is a method of treating a subject suffering from Duchenne muscular dystrophy, the method comprising administering to the subject a first vector (e.g., a recombinant vector or non-viral vector of the disclosure), and a second vector, wherein the second vector encodes a nuclease. In some embodiments, the nuclease is a Type II, Type V-A, Type V-B, Type V-C, Type V-U, or Type VI-B nuclease. In some embodiments, the nuclease is a TAL nuclease, a meganuclease, or a zinc-finger nuclease. In some embodiments, the nuclease is a Cas9, Cas12a, Cas12b, Cas12c, Tnp-B like, Cas13a (C2c2), Cas13b, or Cas14 nuclease. The Cas9 nuclease may be, for example, a Streptococcus pyogenes or Streptococcus aureus Cas9. In some embodiments, the Cas9 is a modified Cas9 nuclease, such as a modified Streptococcus pyogenes Cas9 or a modified Streptococcus aureus Cas9. In some embodiments, the second vector is a plasmid. In some embodiments, the second vector is an expression vector. In some embodiments, the second vector is a viral vector. In embodiments wherein the second vector is a viral vector, it may be a lentiviral vector, a retroviral vector, an adenoviral vector, or an adeno-associated virus (AAV) vector. In some embodiments, the viral vector is an adeno-associated virus (AAV) vector. The serotype of the AAV vector may be selected from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVRh74, AAV2i8, AAVRh10, AAV39, AAV43, AAVRh8, avian AAV, bovine AAV, canine AAV, equine AAV, and ovine AAV. In some embodiments, the second vector is a non-viral vector, wherein the non-viral vector comprises calcium phosphate, liposomes, nanoparticles, and/or lipid emulsions. In some embodiments, the administering induces a frameshift mutation in a target nucleic acid sequence in a cell of the patient. In some embodiments, the frameshift mutation comprises a deletion of at least one nucleotide, wherein the number of nucleotides deleted is not a multiple of 3 (e.g., a deletion of 1, 2, 4, 5, 7, 8, 10, 11, 13, 14, 16, 17, 19 or 20 nucleotides.) In some embodiments, the frameshift mutation comprises an insertion of at least one nucleotide, wherein the number of nucleotides inserted is not a multiple of 3 (e.g., an insertion of 1, 2, 4, 5, 7, 8, 10, 11, 13, 14, 16, 17, 19 or 20 nucleotides.) In some embodiments, the frameshift mutation comprises an insertion of 1 nucleotide. The first vector and the second vector may be administered simultaneously, or may be administered sequentially. In some embodiments, the first vector and the second vector may be administered locally (e.g., to a muscle tissue), or may be administered systemically. In some embodiments, the first vector and the second vector are administered by an oral, rectal, transmucosal, topical, transdermal, inhalation, intravenous, subcutaneous, intradermal, intramuscular, intra-articular, intrathecal, intraventricular, intravenous, intraperitoneal, intranasal, or intraocular route of administration. The subject suffering from DMD may be greater than or equal to 18 years old, less than 18 years old, or less than 2 years old. In some embodiments, the subject is a human. In some embodiments, the ratio of the first vector to the second vector is 1:1 to 1:100. In some embodiments, the ratio of the second vector to the first vector is 1:1 to 1:100.


Also provided is a combination therapy comprising a first composition comprising a first vector comprising a nucleic acid of the disclosure, and a second composition comprising a second vector comprising a nucleic acid that encodes a nuclease. The first and/or the second composition may comprise a pharmaceutically acceptable carrier. In some embodiments, the nuclease is a Type II, Type V-A, Type V-B, Type V-C, Type V-U, or Type VI-B nuclease. In some embodiments, the nuclease is a TAL nuclease, a meganuclease, or a zinc-finger nuclease. In some embodiments, the nuclease is a Cas9, Cas12a, Cas12b, Cas12c, Tnp-B like, Cas13a (C2c2), Cas13b, or Cas14 nuclease. In embodiments wherein the nuclease is a Cas9 nuclease it may be, for example, a Streptococcus pyogenes or Streptococcus aureus Cas9. In some embodiments, the nuclease is a modified Cas9 nuclease, such as a modified Streptococcus pyogenes Cas9 or a modified Streptococcus aureus Cas9.


Also provided is a composition, a recombinant vector, or a non-viral vector of the disclosure for use as a medicament.


Also provided is a composition, a recombinant vector, or a non-viral vector of the disclosure for use in the treatment of Duchenne muscular dystrophy.


Also provided are three different mouse models, wherein the genome of each mouse model comprises a deletion of exon 43, exon 45, or exon 52 of the dystrophin gene, resulting in an out of frame shift and a premature stop codon in exon 44, exon 46, and exon 53, respectively. These mutations are similar to mutations found in approximately 18% of human DMD patients, and correction of these deletions through exon skipping or reframing of surrounding exons can be used to treat DMD. The genome of these mice may further comprise a reporter gene located downstream of and in frame with exon 79 of the dystrophin gene, and upstream of a dystrophin 3′-UTR, wherein said reporter gene is expressed when exon 79 is translated in frame with exon 45, exon 47, or exon 54. The reporter gene may be luciferase. The genome of the mouse may further comprise a protease coding sequence upstream of and in frame with said reporter gene, and downstream of and in frame with exon 79. The protease may be autocatalytic, such as 2A protease. The mouse may be heterozygous for said deletion, or homozygous for said deletion. The mouse may exhibit increased creatine kinase levels, and/or may not exhibit detectable dystrophin protein in heart or skeletal muscle.


Also provided is a method of producing the mouse described above comprising (a) contacting a fertilized oocyte with CRISPR/Cas9 elements and two single guide RNA (sgRNA) targeting sequences flanking exon 43, exon 45, or exon 52, thereby creating a modified oocyte, wherein deletion of exon 43, exon 45, or exon 52 by CRISPR/Cas9 results in an out of frame shift and a premature stop codon in exon 44, exon 46, or exon 53; (b) transferring said modified oocyte into a recipient female. The oocyte genome may comprise a dystrophin gene having a reporter gene located downstream of and in frame with exon 79 of said dystrophin gene, and upstream of a dystrophin 3′-UTR, wherein said reporter gene is expressed when exon 79 is translated in frame with exon 45, exon 47, or exon 54. The reporter gene may be luciferase. The oocyte genome may further comprise a protease coding sequence upstream of and in frame with said reporter gene, and downstream of and in frame with exon 79. The protease may be autocatalytic, such as 2A protease. The mouse may be heterozygous for said deletion, or homozygous for said deletion. The mouse may exhibit increased creatine kinase levels and/or may not exhibit detectable dystrophin protein in heart or skeletal muscle.


In another embodiment, there is provided an isolated cell obtained from the mouse described above. The genome of the cell may further comprise a reporter gene located downstream of and in frame with exon 79 of the dystrophin gene, and upstream of a dystrophin 3′-UTR, wherein said reporter gene is expressed when exon 79 is translated in frame with exon 45, exon 47, or exon 54. The reporter gene may be luciferase. The genome of the cell may further comprise a protease coding sequence upstream of and in frame with said reporter gene, and downstream of and in frame with exon 79. The protease may be autocatalytic, such as 2A protease. The cell may be heterozygous for said deletion, or homozygous for said deletion.


In a further embodiment, there is provided a mouse produced by a method comprising the steps of (a) contacting a fertilized oocyte with CRISPR/Cas9 elements and two single guide RNA (sgRNA) targeting sequences flanking exon 43, exon 45, or exon 52, thereby creating a modified oocyte, wherein deletion of exon 43, exon 45, or exon 52 by CRISPR/Ca9 results in an out of frame shift and a premature stop codon in exon 44, exon 46, or exon 53; (b) transferring said modified oocyte into a recipient female.


These mice provide an important system for assessing the efficacy of a variety of therapeutic analogues for correction of DMD mutations. In one embodiment, CRISPR/Cas9 can be used to skip or reframe exon 44, exon 46 or exon 53, putting the dystrophin protein back in frame. The mice allow for rapid optimization of the method. In another embodiment, the mice can be used to test exon-skipping oligonucleotides or small molecules or other therapeutic modalities in a “humanized” system. In still a further embodiment, there is provided a method of screening a candidate substance for DMD exon-skipping activity comprising (a) treating a mouse (e.g., a mouse from one of the mouse models described herein) with a candidate substance; and (b) assessing in frame transcription and/or translation of exon 79, wherein the presence of in frame transcription and/or translation of exon 79 indicates said candidate substance exhibits exon-skipping activity.


A further embodiment comprises an isolated nucleic acid comprising a sequence of any one of SEQ ID NO: 1-72, 340-359, or 360-515. Also provided is a double-stranded nucleic acid formed by hybridization of SEQ ID NO: 1 and 2, SEQ ID NO: 3 and 4, SEQ ID NO: 5 and 6, SEQ ID NO: 7 and 8, SEQ ID NO: 9 and 10, SEQ ID NO: 11 and 12, SEQ ID NO: 13 and 14, SEQ ID NO: 15 and 16, SEQ ID NO: 17 and 18, SEQ ID NO: 19 and 20, SEQ ID NO: 21 and 22, SEQ ID NO: 23 and 24, SEQ ID NO: 25 and 26, SEQ ID NO: 27 and 28, SEQ ID NO: 29 and 30, SEQ ID NO: 31 and 32, SEQ ID NO: 33 and 34, SEQ ID NO: 35 and 36, SEQ ID NO: 37 and 38, SEQ ID NO: 39 and 40, SEQ ID NO: 41 and 42, SEQ ID NO: 43 and 44, SEQ ID NO: 45 and 46, SEQ ID NO: 47 and 48, SEQ ID NO: 49 and 50, SEQ ID NO: 51 and 52, SEQ ID NO: 53 and 54, SEQ ID NO: 55 and 56, SEQ ID NO: 57 and 58, SEQ ID NO: 59 and 60, SEQ ID NO: 61 and 62, SEQ ID NO: 63 and 64, SEQ ID NO: 65 and 66, SEQ ID NO: 67 and 68, SEQ ID NO: 69 and 70, and SEQ ID NO: 71 and 72, and an expression construct comprising a nucleic acid formed by hybridization of SEQ ID NO: 1 and 2, SEQ ID NO: 3 and 4, SEQ ID NO: 5 and 6, SEQ ID NO: 7 and 8, SEQ ID NO: 9 and 10, SEQ ID NO: 11 and 12, SEQ ID NO: 13 and 14, SEQ ID NO: 15 and 16, SEQ ID NO: 17 and 18, SEQ ID NO: 19 and 20, SEQ ID NO: 21 and 22, SEQ ID NO: 23 and 24, SEQ ID NO: 25 and 26, SEQ ID NO: 27 and 28, SEQ ID NO: 29 and 30, SEQ ID NO: 31 and 32, SEQ ID NO: 33 and 34, SEQ ID NO: 35 and 36, SEQ ID NO: 37 and 38, SEQ ID NO: 39 and 40, SEQ ID NO: 41 and 42, SEQ ID NO: 43 and 44, SEQ ID NO: 45 and 46, SEQ ID NO: 47 and 48, SEQ ID NO: 49 and 50, SEQ ID NO: 51 and 52, SEQ ID NO: 53 and 54, SEQ ID NO: 55 and 56, SEQ ID NO: 57 and 58, SEQ ID NO: 59 and 60, SEQ ID NO: 61 and 62, SEQ ID NO: 63 and 64, SEQ ID NO: 65 and 66, SEQ ID NO: 67 and 68, SEQ ID NO: 69 and 70, and SEQ ID NO: 71 and 72, such as a viral or non-viral vector. Additionally, a kit comprising one or more isolated nucleic acids comprising the sequence of any one of SEQ ID NO: 1-72, 340-359, or 360-515 is provided.


Still a further embodiment comprises a method of correcting a dystrophin gene defect in exon 44, exon 46, or exon 53 of the DMD gene in a subject comprising contacting a cell in said subject with Cpf1 or Cas9 and a DMD guide RNA as defined above, resulting in selective skipping of a mutant DMD exon. The cell may be a muscle cell, a satellite cell, or an induced pluripotent stem cell (iPSC) or iPSC-derived cardiomyocyte (iPSC-CM). Cpf1 and/or DMD guide RNA may be provided to said cell through expression from one or more expression vectors coding therefore, such as a viral vector (e.g., adeno-associated viral vector) or as a non-viral vector. Cpf1 or Cas9 may be provided to said cell as naked plasmid DNA or chemically-modified mRNA.


The method of may further comprise contacting said cell with a single-stranded DMD oligonucleotide to effect homology directed repair or nonhomologous end joining (NHEJ). Cpf1 or Cas9, DMD guide RNA and/or single-stranded DMD oligonucleotide, or expression vectors coding therefor, may be provided to said cell in one or more nanoparticles. Cpf1 or Cas9, DMD guide RNA and/or single-stranded DMD oligonucleotide may be delivered directly to a muscle tissue, such as tibialis anterior, quadricep, soleus, diaphragm or heart. Cpf1 or Cas9, DMD guide RNA and/or single-stranded DMD oligonucleotide may be delivered systemically.


The subject may exhibit normal dystrophin-positive myofibers and/or mosaic dystrophin-positive myofibers containing centralized nuclei. The subject may exhibit a decreased serum CK level as compared to a serum CK level prior to contacting. The subject may exhibit improved grip strength as compared to a serum CK level prior to contacting. The correction may be permanent skipping of said mutant DMD exon, or more than one mutant DMD exon. The Cpf1 or Cas9 and/or DMD guide RNA may be delivered to a human iPSC with an adeno-associated viral vector.


It is contemplated that any method or composition described herein can be implemented with respect to any other method or composition described herein.


Other objects, features and advantages of the present disclosure will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments of the disclosure, are given by way of illustration only, since various changes and modifications within the spirit and scope of the disclosure will become apparent to those skilled in the art from this detailed description.





BRIEF DESCRIPTION OF THE DRAWINGS

The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present disclosure. The disclosure may be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein.



FIGS. 1A-1D. Generation of mice with DMD exon 43, exon 45, or exon 52 deletion. (FIG. 1A) Outline of the CRISPR/Cas9 strategy used for generation of the exon 43, exon 45, and exon 52 deleted mice. (FIG. 1B) Hematoxylin and eosin (H&E) immunostaining of TA, diaphragm and cardiac muscle in exon 43, exon 45, and exon 52 deleted mice. (FIG. 1C) Dystrophin staining of TA, diaphragm and cardiac muscle in exon 43, exon 45, and exon 52 deleted and wild type (WT) mice. Dystrophin stains in red. Nucleus marks by DAPI stains in blue. (FIG. 1D) Serum creatine kinase (CK), a marker of muscle dystrophy that reflects muscle damage and membrane leakage was measured in WT and Δ44, Δ43, Δ45, and Δ52 DMD mice.



FIGS. 2A-2C. Identification of optimal sgRNAs for CRISPR/Cas9 correction of DMD exon 43, exon 45, and exon 52 deletions. (FIG. 2A) Illustration of correction strategies for exon 43, exon 45, and exon 52 deletions. (FIG. 2B) T7E1 assay using 10T ½ mouse cells transfected with SpCas9 and exon 44, exon 46, or exon 53 targeting sgRNAs shows cleavage of the DMD locus. (FIG. 2C) T7E1 assay using 293 human cells transfected with SpCas9 and exon 44, exon 46, or exon 53 targeting sgRNAs shows cleavage of the DMD locus. gRNA spacer sequences used to perform these experiments are listed in Table 2 and Table 3, and the gRNA scaffold sequence used in combination with each spacer sequence corresponds to SEQ ID NO: 147.



FIGS. 3A-3C. DMD patient iPSC-derived cardiomyocytes express dystrophin after CRISPR/Cas9 mediated genome editing by exon skipping (FIG. 3A) Western blot analysis shows restoration of dystrophin expression in cardiomyocytes differentiated from exon 44-skipped iPSCs with exon 43 deletion. Vinculin is loading control. (FIG. 3B) Western blot analysis shows restoration of dystrophin expression in cardiomyocytes differentiated from exon 53-skipped iPSCs with exon 52 deletion. Vinculin is loading control. (FIG. 3C) Immunostaining shows restoration of dystrophin expression in exon 44-edited and exon 53-edited cells. Dystrophin stains in red. Cardiac troponin I stains in green. Nucleus marked by DAPI stains in blue. For ΔE43, the hDMD-E44g1 and hDMD-E44g4 spacers were used, and for ΔE52, the hDMD-E53g4 spacer was used. The gRNA scaffold sequence used in combination with each spacer sequence corresponds to SEQ ID NO: 147.



FIGS. 4A-4C. Identification of optimal sgRNAs for CRISPR/Cas9 correction of DMD exon 52 deletions. (FIG. 4A) Illustration of sgRNA selection strategies for targeting exon 53. (FIG. 4B) Location of sgRNAs targeting exon 53. (FIG. 4C) Western blot analysis shows restoration of dystrophin expression in cardiomyocytes differentiated from exon 53-skipped iPSCs with exon 52 deletion. Vinculin is loading control.



FIGS. 5A-5B. Identification of optimal sgRNAs for CRISPR/Cas9 correction of DMD exon 43 deletions. (FIG. 5A) Illustration of sgRNA selection strategies for targeting exon 44. (FIG. 5B) Location of sgRNAs targeting exon 44.



FIGS. 6A-6D. Identification of optimal sgRNAs for CRISPR/Cas9 correction of DMD exon 45 deletions. (FIG. 6A) Illustration of sgRNA selection strategies for targeting exon 44. (FIG. 6B) Location of sgRNAs targeting exon 44. (FIG. 6C) Western blot analysis shows restoration of dystrophin expression in cardiomyocytes differentiated from exon 44-skipped (SK) or reframed (RF) iPSCs with exon 45 deletion. The numbers #8, #44, #17, and #28 refer to single clones of the corrected iPSC line, with different indels. #8 and #44 were corrected by reframing (−1 nucleotide), and #17 and #28 were corrected by exon skipping. Vinculin is loading control. (FIG. 6D) Immunostaining shows restoration of dystrophin expression in exon 44-edited cells with exon 45 deletion. Dystrophin stains in red. Cardiac troponin I stains in green. Nucleus marked by DAPI stains in blue. For ΔE45, the hDMD-E44g4 spacer was used and the gRNA scaffold sequence corresponds to SEQ ID NO: 147.



FIGS. 7A-7B. Identification of optimal sgRNAs for targeting exon 46. (FIG. 7A) Illustration of sgRNA selection strategies for targeting exon 46. (FIG. 7B) Location of sgRNAs targeting exon 46.



FIGS. 8A-8B. Editing in DMD exons 44, 46, and 53. (FIG. 8A) Diagram of the exon editing strategy for DMD exon 43, exon 45 and exon 52 deletion (FIG. 8B) TIDE analysis using 293 human cells transfected with SpCas9 and exon 44, exon 45, exon 46, or exon 53 targeting sgRNAs. Sequences of the identified gRNA spacer sequences used to perform these experiments are listed in Table 2. For 4E43 and 4E45, the gRNA spacer sequences used were hDMD-E44g4, hDMD-E44g8, hDMD-E44g11, hDMD-E46g2, hDMD-E46g8, hDMD-E53g14, hDMD-E53g15, and hDMD-E53g23, and the gRNA scaffold sequence used in combination with each spacer sequence corresponds to SEQ ID NO: 147.





DETAILED DESCRIPTION

DMD is a new mutation syndrome, and more than 4,000 independent causative mutations that have been identified in humans (world-wide web at dmd.nl). The majority of patient mutations carry deletions that cluster in a hotspot, and thus a therapeutic approach for skipping certain exon applies to large group of patients. The rationale of the exon skipping approach is based on the genetic difference between DMD and Becker muscular dystrophy (BMD) patients. In DMD patients, the reading frame of dystrophin mRNA is disrupted resulting in prematurely truncated, non-functional dystrophin proteins. BMD patients have mutations in the DMD gene that maintain the reading frame allowing the production of internally deleted, but partially functional dystrophins leading to much milder disease symptoms compared to DMD patients.


One the most common mutational hot spots in DMD is the genetic region between exons 44 and 51. Therapeutic approaches involving skipping or reframing of exon 44, exon 46, and exon 53 would treat approximately 18% of the DMD population. Here, the efficiency of CRISPR/Cas9 mediated correction of DMD mutations in patient-derived iPSCs is shown. To further assess the efficiency and optimize CRISPR/Cas9-mediated exon skipping in vivo, a mimic of the human “hot spot” region was generated in mouse models by deleting the exon 43, exon 45, or exon 52 using CRISPR/Cas9 system directed by two single guide RNAs (sgRNAs). The Δ43, Δ45, and Δ52 mouse models exhibit dystrophic myofibers, increased serum creatine kinase level, and reduced muscle function, thus providing a new set of representative models of DMD. These and other aspects of the disclosure are reproduced below.


Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. The terminology used in the detailed description herein is for the purpose of describing particular embodiments only and is not intended to be limiting.


All publications, patent applications, patents, GenBank or other accession numbers and other references mentioned herein are each incorporated by reference herein in their entirety.


The singular forms “a,” “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise.


Furthermore, the terms “about” and “approximately” as used herein when referring to a measurable value such as an amount of the length of a polynucleotide or polypeptide sequence, dose, time, temperature, and the like, is meant to encompass variations of ±20%, ±10%, ±5%, ±1%, ±0.5%, or even ±0.1% of the specified amount.


Also as used herein, “and/or” refers to and encompasses any and all possible combinations of one or more of the associated listed items, as well as the lack of combinations when interpreted in the alternative (“or”).


Reference to a vector or other DNA sequences as “recombinant” merely acknowledges the operable linkage of DNA sequences which are not typically operably linked as isolated from or found in nature.


The terms “gRNA” and “sgRNA” are used interchangeably herein, and refer to a short synthetic RNA composed of a “spacer” (or “targeting”) sequence and a “scaffold” sequence. In some embodiments, the gRNA may further comprise a polyA tail.


In accordance with some embodiments of the disclosure, a “frameshift mutation” (or “frame-shift mutation” or “frameshift”) is caused by a deletion or insertion in a DNA sequence that shifts the reading frame of the DNA sequence.


In accordance with some embodiments of the disclosure, “exon skipping” (or “exon-skipping”) refers to a strategy which causes sections (e.g. mutated sections) of a gene to be “skipped” during RNA splicing, allowing the expression of a partially or fully functional protein.


Unless the context indicates otherwise, it is specifically intended that the various features described herein can be used in any combination.


I. GENE EDITING SYSTEMS, E.G., CRISPR SYSTEMS

Provided herein are gene editing systems which produce an insertion, deletion, or replacement of DNA at a specific site in the genome of an organism or cell. In some embodiments, the genome editing systems introduce a loss of function mutation or a gain of function mutation. In some embodiments, the genome editing systems of the disclosure are capable of modulating splicing or causing a frameshift in a target DNA sequence. In some embodiments, the genome editing systems correct DNA mutations in vitro and/or in vivo.


The genome editing systems of the disclosure may comprise at least one nuclease (or catalytic domain thereof) and at least one gRNA, or nucleic acids encoding the at least one nuclease (or catalytic domain thereof) and the at least one gRNA. A sequence encoding the at least one nuclease and a sequence encoding the at least one gRNA may be delivered using the same vector (e.g., an AAV vector), or using different vectors (e.g., a first AAV vector for delivering the sequence encoding the nuclease, and a second AAV vector for delivering the sequence encoding the at least one gRNA).


In some embodiments, the nuclease is a Type II, Type V-A, Type V-B, Type V-C, Type V-U, Type VI-B nuclease. In some embodiments, the nuclease is a transcription activator-like effector nuclease (TALEN), a meganuclease, or a zinc-finger nuclease. In some embodiments, the nuclease is a Cas9, Cas12a (Cpf1), Cas12b, Cas12c, Tnp-B like, Cas13a (C2c2), Cas13b, or Cas14 nuclease. For example, in some embodiments, the nuclease is a Cas9 nuclease or a Cpf1 nuclease.


In some embodiments, the nuclease is a modified form or variant of a Cas9, Cas12a (Cpf1), Cas12b, Cas12c, Tnp-B like, Cas13a (C2c2), Cas13b, or Cas14 nuclease. In some embodiments, the nuclease is a modified form or variant of a TAL nuclease, a meganuclease, or a zinc-finger nuclease. A “modified” or “variant” nuclease is one that is, for example, truncated, fused to another protein (such as another nuclease), catalytically inactivated, etc. In some embodiments, the nuclease may have at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% sequence identity to a naturally occurring Cas9, Cas12a (Cpf1), Cas12b, Cas12c, Tnp-B like, Cas13a (C2c2), Cas13b, Cas14 nuclease, or a TALEN, meganuclease, or zinc-finger nuclease.


In embodiments, the nuclease is a Cas9 nuclease derived from S. pyogenes (SpCas9). An exemplary SpCas9 sequence is provided in SEQ ID NO: 166. In some embodiments, the nuclease has at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to SEQ ID NO: 166, shown below:









(SEQ ID NO: 166)


MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGA





LLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHR





LEESFLVEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKAD





LRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENP





INASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTP





NFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAI





LLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEI





FFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLR





KQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPY





YVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDK





NLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVD





LLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKI





IKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQ





LKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDD





SLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKV





MGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHP





VENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFLKDD





SIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNL





TKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLI





REVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKK





YPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEI





TLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEV





QTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVE





KGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPK





YSLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKGSPE





DNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDK





PIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQ





SITGLYETRIDLSQLGGD






In embodiments, the nuclease is a Cas9 derived from S. aureus (SaCas9). An exemplary SaCas9 sequence is provided in SEQ ID NO: 167. In some embodiments, the nuclease has at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to SEQ ID NO: 167, shown below:









(SEQ ID NO: 167)


MKRNYILGLDIGITSVGYGIIDYETRDVIDAGVRLFKEANVENNEGRRSK





RGARRLKRRRRHRIQRVKKLLFDYNLLTDHSELSGINPYEARVKGLSQKL





SEEEFSAALLHLAKRRGVHNVNEVEEDTGNELSTKEQISRNSKALEEKYV





AELQLERLKKDGEVRGSINRFKTSDYVKEAKQLLKVQKAYHQLDQSFIDT





YIDLLETRRTYYEGPGEGSPFGWKDIKEWYEMLMGHCTYFPEELRSVKYA





YNADLYNALNDLNNLVITRDENEKLEYYEKFQIIENVFKQKKKPTLKQIA





KEILVNEEDIKGYRVTSTGKPEFTNLKVYHDIKDITARKEIIENAELLDQ





IAKILTIYQSSEDIQEELTNLNSELTQEEIEQISNLKGYTGTHNLSLKAI





NLILDELWHTNDNQIAIFNRLKLVPKKVDLSQQKEIPTTLVDDFILSPVV





KRSFIQSIKVINAIIKKYGLPNDIIIELAREKNSKDAQKMINEMQKRNRQ





TNERIEEIIRTTGKENAKYLIEKIKLHDMQEGKCLYSLEAIPLEDLLNNP





FNYEVDHIIPRSVSFDNSFNNKVLVKQEENSKKGNRTPFQYLSSSDSKIS





YETFKKHILNLAKGKGRISKTKKEYLLEERDINRFSVQKDFINRNLVDTR





YATRGLMNLLRSYFRVNNLDVKVKSINGGFTSFLRRKWKFKKERNKGYKH





HAEDALIIANADFIFKEWKKLDKAKKVMENQMFEEKQAESMPEIETEQEY





KEIFITPHQIKHIKDFKDYKYSHRVDKKPNRELINDTLYSTRKDDKGNTL





IVNNLNGLYDKDNDKLKKLINKSPEKLLMYHHDPQTYQKLKLIMEQYGDE





KNPLYKYYEETGNYLTKYSKKDNGPVIKKIKYYGNKLNAHLDITDDYPNS





RNKVVKLSLKPYRFDVYLDNGVYKFVTVKNLDVIKKENYYEVNSKCYEEA





KKLKKISNQAEFIASFYNNDLIKINGELYRVIGVNNDLLNRIEVNMIDIT





YREYLENMNDKRPPRIIKTIASKTQSIKKYSTDILGNLYEVKSKKHPQII






In embodiments, the nuclease is a Cpf1 enzyme from Acidaminococcus (species BV3L6, UniProt Accession No. U2UMQ6). For example, the Cpf1 enzyme may have the sequence set forth below (SEQ ID NO: 168), or a sequence with at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity thereto:









(SEQ ID NO: 168)


MTQFEGFTNLYQVSKTLRFELIPQGKTLKHIQEQGFIEEDKARNDHYKEL





KPIIDRIYKTYADQCLQLVQLDWENLSAAIDSYRKEKTEETRNALIEEQA





TYRNAIHDYFIGRTDNLTDAINKRHAEIYKGLFKAELFNGKVLKQLGTVT





TTEHENALLRSFDKFTTYFSGFYENRKNVFSAEDISTAIPHRIVQDNFPK





FKENCHIFTRLITAVPSLREHFENVKKAIGIFVSTSIEEVFSFPFYNQLL





TQTQIDLYNQLLGGISREAGTEKIKGLNEVLNLAIQKNDETAHIIASLPH





RFIPLFKQILSDRNTLSFILEEFKSDEEVIQSFCKYKTLLRNENVLETAE





ALFNELNSIDLTHIFISHKKLETISSALCDHWDTLRNALYERRISELTGK





ITKSAKEKVQRSLKHEDINLQEIISAAGKELSEAFKQKTSEILSHAHAAL





DQPLPTTLKKQEEKEILKSQLDSLLGLYHLLDWFAVDESNEVDPEFSARL





TGIKLEMEPSLSFYNKARNYATKKPYSVEKFKLNFQMPTLASGWDVNKEK





NNGAILFVKNGLYYLGIMPKQKGRYKALSFEPTEKTSEGFDKMYYDYFPD





AAKMIPKCSTQLKAVTAHFQTHTTPILLSNNFIEPLEITKEIYDLNNPEK





EPKKFQTAYAKKTGDQKGYREALCKWIDFTRDFLSKYTKTTSIDLSSLRP





SSQYKDLGEYYAELNPLLYHISFQRIAEKEIMDAVETGKLYLFQIYNKDF





AKGHHGKPNLHTLYWTGLFSPENLAKTSIKLNGQAELFYRPKSRMKRMAH





RLGEKMLNKKLKDQKTPIPDTLYQELYDYVNHRLSHDLSDEARALLPNVI





TKEVSHEIIKDRRFTSDKFFFHVPITLNYQAANSPSKFNQRVNAYLKEHP





ETPIIGIDRGERNLIYITVIDSTGKILEQRSLNTIQQFDYQKKLDNREKE





RVAARQAWSVVGTIKDLKQGYLSQVIHEIVDLMIHYQAVVVLENLNFGFK





SKRTGIAEKAVYQQFEKMLIDKLNCLVLKDYPAEKVGGVLNPYQLTDQFT





SFAKMGTQSGFLFYVPAPYTSKIDPLTGFVDPFVWKTIKNHESRKHFLEG





FDFLHYDVKTGDFILHFKMNRNLSFQRGLPGFMPAWDIVFEKNETQFDAK





GTPFIAGKRIVPVIENHRFTGRYRDLYPANELIALLEEKGIVFRDGSNIL





PKLLENDDSHAIDTMVALIRSVLQMRNSNAATGEDYINSPVRDLNGVCFD





SRFQNPEWPMDADANGAYHIALKGQLLLNHLKESKDLKLQNGISNQDWLA





YIQELRN






In some embodiments, the nuclease is a Cpf1 enzyme from Lachnospiraceae (species ND2006, UniProt Accession No. A0A182DWE3). An exemplary Lachnospiraceae Cpf1 sequence is provided in SEQ ID NO: 169. In some embodiments, the nuclease has at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to SEQ ID NO: 169, which is provided below:









(SEQ ID NO: 169)


AASKLEKFTNCYSISKTLRFKAIPVGKTQENIDNKRLLVEDEKRAEDYKG





VKKLLDRYYLSFINDVLHSIKLKNLNNYISLFRKKTRTEKENKELENLEI





NLRKEIAKAFKGAAGYKSLFKKDIIETILPEAADDKDEIALVNSFNGFTT





AFTGFFDNRENMFSEEAKSTSIAFRCINENLTRYISNMDIFEKVDAIFDK





HEVQEIKEKILNSDYDVEDFFEGEFFNFVLTQEGIDVYNAIIGGFVTESG





EKIKGLNEYINLYNAKTKQALPKFKPLYKQVLSDRESLSFYGEGYTSDEE





VLEVFRNTLNKNSEIFSSIKKLEKLFKNFDEYSSAGIFVKNGPAISTISK





DIFGEWNLIRDKWNAEYDDIHLKKKAVVTEKYEDDRRKSFKKIGSFSLEQ





LQEYADADLSVVEKLKEIIIQKVDEIYKVYGSSEKLFDADFVLEKSLKKN





DAVVAIMKDLLDSVKSFENYIKAFFGEGKETNRDESFYGDEVLAYDILLK





VDHIYDAIRNYVTQKPYSKDKFKLYFQNPQFMGGWDKDKETDYRATILRY





GSKYYLAIMDKKYAKCLQKIDKDDVNGNYEKINYKLLPGPNKMLPKVFFS





KKWMAYYNPSEDIQKIYKNGTFKKGDMFNLNDCHKLIDFFKDSISRYPKW





SNAYDFNFSETEKYKDIAGFYREVEEQGYKVSFESASKKEVDKLVEEGKL





YMFQIYNKDFSDKSHGTPNLHTMYFKLLFDENNHGQIRLSGGAELFMRRA





SLKKEELVVHPANSPIANKNPDNPKKTTTLSYDVYKDKRFSEDQYELHIP





IAINKCPKNIFKINTEVRVLLKHDDNPYVIGIDRGERNLLYIVVVDGKGN





IVEQYSLNEIINNFNGIRIKTDYHSLLDKKEKERFEARQNWTSIENIKEL





KAGYISQVVHKICELVEKYDAVIALEDLNSGFKNSRVKVEKQVYQKFEKM





LIDKLNYMVDKKSNPCATGGALKGYQITNKFESFKSMSTQNGFIFYIPAW





LTSKIDPSTGFVNLLKTKYTSIADSKKFISSFDRIMYVPEEDLFEFALDY





KNFSRTDADYIKKWKLYSYGNRIRIFAAAKKNNVFAWEEVCLTSAYKELF





NKYGINYQQGDIRALLCEQSDKAFYSSFMALMSLMLQMRNSITGRTDVDF





LISPVKNSDGIFYDSRNYEAQENAILPKNADANGAYNIARKVLWAIGQFK





KAEDEKLDKVKIAISNKEWLEYAQTSVK






In some embodiments, a sequence encoding the nuclease is codon optimized for expression in mammalian cells. In some embodiments, the sequence encoding the nuclease is codon optimized for expression in human cells or mouse cells.


In some embodiments, the disclosure provides a nucleic acid comprising a sequence encoding a single guide RNA (sgRNA) comprising a spacer sequence and a scaffold sequence.


A. Spacer


A spacer sequence is a short nucleic acid sequence used to target a nuclease (e.g., a Cas9 nuclease) to a specific nucleotide region of interest (e.g., a genomic DNA sequence to be cleaved).


In some embodiments, the spacer may be about 17-24 base pairs in length, such as about 20 base pairs in length. In some embodiments, the spacer may be about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, or about 30 base pairs in length. In some embodiments, the spacer may be at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, or at least 30 base pairs in length. In some embodiments, the spacer may be 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 base pairs in length. In some embodiments, the spacer sequence has between about 40% to about 80% GC content.


In some embodiments, the spacer targets a site that immediately precedes a 5′ protospacer adjacent motif (PAM). The PAM sequence may be selected based on the desired nuclease. For example, the PAM sequence may be any one of the PAM sequences shown in Table 1 below, wherein N refers to any nucleic acid, R refers to A or G, Y refers to C or T, W refers to A or T, and V refers to A or C or G.









TABLE 1







Exemplary Nucleases and PAM sequences










PAM





sequence (5′
SEQ




to 3′)
ID NO:
Nuclease
Isolated from





NGG

SpCas9

Streptococcus pyogenes



NGRRT or
128, 129
SaCas9

Staphylococcus aureus



NGRRN





NNNNGATT
130
NmeCas9

Neisseria meningitidis



NNNNRYAC
131
CjCas9

Campylobacter jejuni



NNAGAAW
132
StCas9

Streptococcus thermophilus



TTTV
133
LbCpf1

Lachnospiraceae bacterium



TTTV
134
AsCpf1

Acidaminococcus sp.










In some embodiments, a spacer may target a sequence of a mammalian gene, such as a human gene. In some embodiments, the spacer may target a mutant gene. In some embodiments, the spacer may target a coding sequence. In some embodiments, the spacer targets the dystrophin (DMD) gene. An exemplary wild-type dystrophin sequence includes the human DNA sequence (see GenBank Accession NO. NC_000023.11), located on the human X chromosome, which codes for the protein dystrophin (GenBank Accession No. AAA53189), the sequence of which is reproduced below:










(SEQ ID NO: 330)










1
MLWWEEVEDC YEREDVQKKT FTKWVNAQFS KFGKQHIENL FSDLQDGRRL LDLLEGLTGQ






61
KLPKEKGSTR VHALNNVNKA LRVLQNNNVD LVNIGSTDIV DGNHKLTLGL IWNIILHWQV





121
KNVMKNIMAG LQQTNSEKIL LSWVRQSTRN YPQVNVINFT TSWSDGLALN ALIHSHRPDL





181
FDWNSVVCQQ SATQRLEHAF NIARYQLGIE KLLDPEDVDT TYPDKKSILM YITSLFQVLP





241
QQVSIEAIQE VEMLPRPPKV TKEEHFQLHH QMHYSQQITV SLAQGYERTS SPKPRFKSYA





301
YTQAAYVTTS DPTRSPFPSQ HLEAPEDKSF GSSLMESEVN LDRYQTALEE VLSWLLSAED





361
TLQAQGEISN DVEVVKDQFH THEGYMMDLT AHQGRVGNIL QLGSKLIGTG KLSEDEETEV





421
QEQMNLLNSR WECLRVASME KQSNLHRVLM DLQNQKLKEL NDWLTKTEER TRKMEEEPLG





481
PDLEDLKRQV QQHKVLQEDL EQEQVRVNSL THMVVVVDES SGDHATAALE EQLKVLGDRW





541
ANICRWTEDR WVLLQDILLK WQRLTEEQCL FSAWLSEKED AVNKIHTTGF KDQNEMLSSL





601
QKLAVLKADL EKKKQSMGKL YSLKQDLLST LKNKSVTQKT EAWLDNFARC WDNLVQKLEK





661
STAQISQAVT TTQPSLTQTT VMETVTTVTT REQILVKHAQ EELPPPPPQK KRQITVDSEI





721
RKRLDVDITE LHSWITRSEA VLQSPEFAIF RKEGNFSDLK EKVNAIEREK AEKFRKLQDA





781
SRSAQALVEQ MVNEGVNADS IKQASEQLNS RWIEFCQLLS ERLNWLEYQN NIIAFYNQLQ





841
QLEQMTTTAE NWLKIQPTTP SEPTAIKSQL KICKDEVNRL SGLQPQIERL KIQSIALKEK





901
GQGPMFLDAD EVAFTNHFKQ VFSDVQAREK ELQTIFDTLP PMRYQETMSA IRTWVQQSET





961
KLSIPQLSVT DYEIMEQRLG ELQALQSSLQ EQQSGLYYLS TTVKEMSKKA PSEISRKYQS





1021
EFEEIEGRWK KLSSQLVEHC QKLEEQMNKL RKIQNHIQTL KKWMAEVDVF LKEEWPALGD





1081
SEILKKQLKQ CRLLVSDIQT IQPSLNSVNE GGQKIKNEAE PEFASRLETE LKELNTQWDH





1141
MCQQVYARKE ALKGGLEKTV SLQKDLSEMH EWMTQAEEEY LERDFEYKTP DELQKAVEEM





1201
KRAKEEAQQK EAKVKLLTES VNSVIAQAPP VAQEALKKEL ETLTTNYQWL CTRLNGKCKT





1261
LEEVWACWHE LLSYLEKANK WLNEVEFKLK TTENIPGGAE EISEVLDSLE NLMRHSEDNP





1321
NQIRILAQTL TDGGVMDELI NEELETFNSR WRELHEEAVR RQKLLEQSIQ SAQETEKSLH





1381
LIQESLTFID KQLAAYIADK VDAAQMPQEA QKIQSDLTSH EISLEEMKKH NQGKEAAQRV





1441
LSQIDVAQKK LQDVSMKFRL FQKPANFELR LQESKMILDE VKMHLPALET KSVEQEVVQS





1501
QLNHCVNLYK SLSEVKSEVE MVIKTGRQIV QKKQTENPKE LDERVTALKL HYNELGAKVT





1561
ERKQQLEKCL KLSRKMRKEM NVLTEWLAAT DMELTKRSAV EGMPSNLDSE VAWGKATQKE





1621
IEKQKVHLKS ITEVGEALKT VLGKKETLVE DKLSLLNSNW IAVTSRAEEW LNLLLEYQKH





1681
METFDQNVDH ITKWIIQADT LLDESEKKKP QQKEDVLKRL KAELNDIRPK VDSTRDQAAN





1741
LMANRGDHCR KLVEPQISEL NHRFAAISHR IKTGKASIPL KELEQFNSDI QKLLEPLEAE





1801
IQQGVNLKEE DFNKDMNEDN EGTVKELLQR GDNLQQRITD ERKREEIKIK QQLLQTKHNA





1861
LKDLRSQRRK KALEISHQWY QYKRQADDLL KCLDDIEKKL ASLPEPRDER KIKEIDRELQ





1921
KKKEELNAVR RQAEGLSEDG AAMAVEPTQI QLSKRWREIE SKFAQFRRLN FAQIHTVREE





1981
TMMVMTEDMP LEISYVPSTY LTEITHVSQA LLEVEQLLNA PDLCAKDFED LFKQEESLKN





2041
IKDSLQQSSG RIDIIHSKKT AALQSATPVE RVKLQEALSQ LDFQWEKVNK MYKDRQGRFD





2101
RSVEKWRRFH YDIKIFNQWL TEAEQFLRKT QIPENWEHAK YKWYLKELQD GIGQRQTVVR





2161
TLNATGEEII QQSSKTDASI LQEKLGSLNL RWQEVCKQLS DRKKRLEEQK NILSEFQRDL





2221
NEFVLWLEEA DNIASIPLEP GKEQQLKEKL EQVKLLVEEL PLRQGILKQL NETGGPVLVS





2281
APISPEEQDK LENKLKQTNL QWIKVSRALP EKQGEIEAQI KDLGQLEKKL EDLEEQLNHL





2341
LLWLSPIRNQ LEIYNQPNQE GPFDVQETEI AVQAKQPDVE EILSKGQHLY KEKPATQPVK





2401
RKLEDLSSEW KAVNRLLQEL RAKQPDLAPG LTTIGASPTQ TVTLVTQPVV TKETAISKLE





2461
MPSSLMLEVP ALADFNRAWT ELTDWLSLLD QVIKSQRVMV GDLEDINEMI IKQKATMQDL





2521
EQRRPQLEEL ITAAQNLKNK TSNQEARTII TDRIERIQNQ WDEVQEHLQN RRQQLNEMLK





2581
DSTQWLEAKE EAEQVLGQAR AKLESWKEGP YTVDAIQKKI TETKQLAKDL RQWQTNVDVA





2641
NDLALKLLRD YSADDTRKVH MITENINASW RSIHKRVSER EAALEETHRL LQQFPLDLEK





2701
FLAWLTEAET TANVLQDATR KERLLEDSKG VKELMKQWQD LQGEIEAHTD VYHNLDENSQ





2761
KILRSLEGSD DAVLLQRRLD NMNFKWSELR KKSLNIRSHL EASSDQWKRL HLSLQELLVW





2821
LQLKDDELSR QAPIGGDFPA VQKQNDVHRA FKRELKTKEP VIMSTLETVR IFLTEQPLEG





2881
LEKLYQEPRE LPPEERAQNV TRLLRKQAEE VNTEWEKLNL HSADWQRKID ETLERLQELQ





2941
EATDELDLKL RQAEVIKGSW QPVGDLLIDS LQDHLEKVKA LRGEIAPLKE NVSHVNDLAR





3001
QLTTLGIQLS PYNLSTLEDL NTRWKLLQVA VEDRVRQLHE AHRDFGPASQ HFLSTSVQGP





3061
WERAISPNKV PYYINHETQT TCWDHPKMTE LYQSLADLNN VRFSAYRTAM KLRRLQKALC





3121
LDLLSLSAAC DALDQHNLKQ NDQPMDILQI INCLTTIYDR LEQEHNNLVN VPLCVDMCLN





3181
WLLNVYDTGR TGRIRVLSFK TGIISLCKAH LEDKYRYLFK QVASSTGFCD QRRLGLLLHD





3241
SIQIPRQLGE VASFGGSNIE PSVRSCFQFA NNKPEIEAAL FLDWMRLEPQ SMVWLPVLHR





3301
VAAAETAKHQ AKCNICKECP IIGFRYRSLK HFNYDICQSC FFSGRVAKGH KMHYPMVEYC





3361
TPTTSGEDVR DFAKVLKNKF RTKRYFAKHP RMGYLPVQTV LEGDNMETPV TLINFWPVDS





3421
APASSPQLSH DDTHSRIEHY ASRLAEMENS NGSYLNDSIS PNESIDDEHL LIQHYCQSLN





3481
QDSPLSQPRS PAQILISLES EERGELERIL ADLEEENRNL QAEYDRLKQQ HEHKGLSPLP





3541
SPPEMMPTSP QSPRDAELIA EAKLLRQHKG RLEARMQILE DHNKQLESQL HRLRQLLEQP





3601
QAEAKVNGTT VSSPSTSLQR SDSSQPMLLR VVGSQTSDSM GEEDLLSPPQ DTSTGLEEVM





3661
EQLNNSFPSS RGRNTPGKPM REDTM..







In some embodiments, the spacer sequence targets a sequence of the DMD gene. In some embodiments, the spacer targets an exon of the DMD gene. In some embodiments, the spacer targets exon 43, exon 44, exon 46, exon 50 or exon 53 of the DMD gene.


In some embodiments, the spacer may have a sequence of any one of SEQ ID NOs: 135-146, 170-260, 337-339, or 617 (shown in Table 2 below). In some embodiments, a spacer may have a sequence at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to the sequence of any one of SEQ ID NOs: 135-146, 170-260, 337-339, or 617. In some embodiments, a spacer may have a sequence of any one of the spacers shown in Table 2, or a sequence that is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical thereto.









TABLE 2







Exemplary spacer sequences for targeting an exon


of the human DMD gene











sgRNA spacer
Exon


SEQ ID


name
targeted
Species
sgRNA spacer sequence
NO





hDMD-E43g1
43
Human
GTTTTAAAATTTTTATATTA
189





hDMD-E43g2
43
Human
TTTTTATATTACAGAATATAA
190





hDMD-E43g3
43
Human
ATATTACAGAATATAAAAGA
191





hDMD-E43g5
43
Human
AAATGTACAAGGACCGACAA
188





hDMD-E43g4
43
Human
TATGTGTTACCTACCCTTGT
135





hDMD-E43g6
43
Human
GTACAAGGACCGACAAGGGT
136





hDMD-E44g1
44
Human
ATCCATATGCTTTTACCTGC
137





hDMD-E44g2
44
Human
gatccatatgcttttACCTG
192





hDMD-E44g3
44
Human
CAGATCTGTCAAATCGCCTG
193





hDMD-E44g4
44
Human
TAAATACAAATGGTATCTTA
138





hDMD-E44g5
44
Human
AGATCTGTCAAATCGCCTGC
139





hDMD-E44g6
44
Human
ACAGATCTGTTGAGAAATGG
140





hDMD-E44g7
44
Human
TTAGCATGTTCCCAATTCTC
141





hDMD-E44g8
44
Human
GGGAACATGCTAAATACAAA
142





hDMD-E44g9
44
Human
TTGACAGATCTGTTGAGAAA
143





hDMD-
44
Human
AGACACAAATTCCTGAGAAT
144


E44g10









hDMD-
44
Human
GACACAAATTCCTGAGAATT
145


E44g11









hDMD-
44
Human
CGCctgcaggtaaaagcata
194


E44g12









hDMD-
44
Human
tttacctgcagGCGATTTGA
195


E44g13









hDMD-
44
Human
gGCGATTTGACAGATCTGTT
196


E44g14









hDMD-
44
Human
ATTTAATCAGTGGCTAACAG
197


E44g15









hDMD-
44
Human
AGAAACTGTTCAGCTTCTGT
198


E44g16









hDMD-
44
Human
AGTGGCTAACAGAAGCTGAA
199


E44g17









hDMD-
44
Human
AAGCTGAACAGTTTCTCAGA
200


E44g18









hDMD-
44
Human
TTTAGCATGTTCCCAATTCT
201


E44g19









hDMD-
44
Human
CTTAAGATACCATTTGTATT
202


E44g20









hDMD-
44
Human
CTAAATACAAATGGTATCTT
203


E44g21









hDMD-
44
Human
TACAAATGGTATCTTAAGgt
204


E44g22









hDMD-
44
Human
ACAAATCAAAGACTTACCTT
146


E44g23









hDMD-E45g4
45
Human
atcttacagGAACTCCAGGA
617





hDMD-E46g1
46
Human
ttattcttctttctccagGC
205





hDMD-E46g2
46
Human
AATTTTATTCTTCTTTCTCC
170





hDMD-E46g5
46
Human
ATTCTTTTGTTCTTCTAGCC
171





hDMD-E46g6
46
Human
AAATGAATTTGTTTTATGGT
172





hDMD-E46g7
46
Human
TGAATTTGTTTTATGGTTGG
173





hDMD-E46g8
46
Human
AGAAAAGCTTGAGCAAGTCA
174





hDMD-E46g9
46
Human
TTCTTCTAGCCTGGAGAAAG
206





hDMD-
46
Human
CAATTTTATTCTTCTTTCTC
207


E46g10









hDMD-
46
Human
TTGTTCTTCTAGCCTGGAGA
208


E46g11









hDMD-
46
Human
TCTTTTGTTCTTCTAGCCTG
209


E46g12









hDMD-
46
Human
TTCTTCTTTCTCCAGGCTAG
210


E46g13









hDMD-
46
Human
ACTTGCTCAAGCTTTTCTTT
211


E46g14









hDMD-
46
Human
ACTAAAAGAAAAGCTTGAGC
212


E46g15









hDMD-
46
Human
AAAATTACCTTGACTTGCTC
213


E46g16









hDMD-
46
Human
AAGAAAAGCTTGAGCAAGTC
214


E46g17









hDMD-
46
Human
TCTCCAGGCTAGAAGAACAA
337


E46g18









hDMD-
46
Human
AGAACAAAAGAATATCTTGT
338


E46g19









hDMD-
46
Human
TATCTTGTCAGAATTTCAAA
339


E46g20









hDMD-E50g1
50
Human
TGTATGCTTTTCTGTTAAAG
175





hDMD-E50g2
50
Human
ATGTGTATGCTTTTCTGTTA
215





hDMD-E50g3
50
Human
GTGTATGCTTTTCTGTTAAA
216





hDMD-E50g4
50
Human
ATGCTTTTCTGTTAAAGAGG
217





hDMD-E50g5
50
Human
TCTTCTAACTTCCTCTTTAA
218





hDMD-E50g6
50
Human
TAACTTCCTCTTTAACAGAA
219





hDMD-E50g7
50
Human
TTTTCTGTTAAAGAGGAAGT
220





hDMD-E50g8
50
Human
TCTGTTAAAGAGGAAGTTAG
221





hDMD-E50g9
50
Human
AAGAGGAAGTTAGAAGATCT
222





hDMD-
50
Human
AGTTAGAAGATCTGAGCTCT
223


E50g10









hDMD-
50
Human
TAGAAGATCTGAGCTCTGAG
224


E50g11









hDMD-
50
Human
AGATCTGAGCTCTGAGTGGA
225


E50g12









hDMD-
50
Human
ACCGCCTTCCACTCAGAGCT
226


E50g13









hDMD-
50
Human
GGTTTACCGCCTTCCACTCA
227


E50g14









hDMD-
50
Human
AAGCAGCCTGACCTAGCTCC
228


E50g15









hDMD-
50
Human
GTCAGTCCAGGAGCTAGGTC
229


E50g16









hDMD-
50
Human
GGTCAGTCCAGGAGCTAGGT
230


E50g17









hDMD-
50
Human
TAGTGGTCAGTCCAGGAGCT
231


E50g18









hDMD-
50
Human
ATAGTGGTCAGTCCAGGAGC
232


E50g19









hDMD-
50
Human
TCCAATAGTGGTCAGTCCAG
233


E50g20









hDMD-
50
Human
GCTCCAATAGTGGTCAGTCC
234


E50g21









hDMD-
50
Human
TTACAGGCTCCAATAGTGGT
235


E50g22









hDMD-
50
Human
ATACTTACAGGCTCCAATAG
236


E50g23









hDMD-
50
Human
AGTATACTTACAGGCTCCAA
237


E50g24









hDMD-
50
Human
GCTCCTGGACTGACCACTAT
238


E50g25









hDMD-
50
Human
TCCTGGACTGACCACTATTG
239


E50g26









hDMD-
50
Human
TGACCACTATTGGAGCCTGT
240


E50g27









hDMD-
50
Human
ATGGGATCCAGTATACTTAC
241


E50g28









hDMD-
50
Human
AATGGGATCCAGTATACTTA
242


E50g29









hDMD-
50
Human
ATTGGAGCCTGTAAGTATAC
243


E50g30









hDMD-E51g4
51
Human
TCATCTCGTTGATATCCTCA
244





hDMD-E51g5
51
Human
CGAGATGATCATCAAGCAGA
245





hDMD-E51g6
51
Human
GTGACCTTGAGGATATCAAC
246





hDMD-E51g7
51
Human
TCAACGAGATGATCATCAAG
247





hDMD-E51g8
51
Human
ACGAGATGATCATCAAGCAG
248





hDMD-E53g1
53
Human
ATTTATTTTTCCTTTTATTC
249





hDMD-E53g2
53
Human
TTTCCTTTTATTCTAGTTGA
250





hDMD-E53g3
53
Human
TGATTCTGAATTCTTTCAAC
251





hDMD-E53g4
53
Human
AAAGAAAATCACAGAAACCA
176





hDMD-E53g5
53
Human
AAAATCACAGAAACCAAGGT
252





hDMD-E53g6
53
Human
GGTATCTTTGATACTAACCT
177





hDMD-E53g7
53
Human
TGAAAGAATTCAGAATCAGT
178





hDMD-E53g8
53
Human
ACTGTTGCCTCCGGTTCTGA
179





hDMD-E53g9
53
Human
TACAAGAACACCTTCAGAAC
180





hDMD-
53
Human
AAGAACACCTTCAGAACCGG
181


E53g10









hDMD-
53
Human
TTTCATTCAACTGTTGCCTC
182


E53g11









hDMD-
53
Human
TGTTAAAGGATTCAACACAA
183


E53g12









hDMD-
53
Human
AAAGGATTCAACACAATGGC
184


E53g13









hDMD-
53
Human
AATTCAGAATCAGTGGGATG
185


E53g14









hDMD-
53
Human
TTGAAAGAATTCAGAATCAG
186


E53g15









hDMD-
53
Human
ACAGTTGAATGAAATGTTAA
253


E53g16









hDMD-
53
Human
ACCTTCAGAACCGGAGGCAA
254


E53g17









hDMD-
53
Human
AATTCTTTCAActagaataa
255


E53g18









hDMD-
53
Human
ttattctagTTGAAAGAATT
256


E53g19









hDMD-
53
Human
tagTTGAAAGAATTCAGAAT
257


E53g20









hDMD-
53
Human
ATGAAGTACAAGAACACCTT
258


E53g21









hDMD-
53
Human
AACTGTTGCCTCCGGTTCTG
259


E53g22









hDMD-
53
Human
CAAGAACACCTTCAGAACCG
187


E53g23









hDMD-
53
Human
AACAGTTGAATGAAATGTTA
260


E53g24









In some embodiments, the spacer may have a sequence of any one of SEQ ID NOs: 261-329 (shown in Table 3 below). In some embodiments, a spacer may have a sequence at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to the sequence of any one of SEQ ID NOs: 261-329. In some embodiments, a spacer may have a sequence of any one of the spacers shown in Table 3, or a sequence that is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical thereto.









TABLE 3







Exemplary spacer sequences for targeting an exon of


the mouse DMD gene











sgRNA spacer
Exon


SEQ ID


name
targeted
Species
sgRNA spacer sequence
NO





mDMD-E43g1
43
Mouse
ATTTGCAACAAATCTCAGGT
261





mDMD-E43g2
43
Mouse
AGAATGTACAAGGAACGACA
262





mDMD-E43g3
43
Mouse
GAATGTACAAGGAACGACAA
263





mDMD-E43g4
43
Mouse
GTACAAGGAACGACAAGGGT
264





mDMD-E44g1
44
Mouse
catccatgttttcaaattat
265





mDMD-E44g2
44
Mouse
TCGACAGATCAGTTGAAAAA
266





mDMD-E44g3
44
Mouse
AGACACAAAATCCTGAAAAC
267





mDMD-E44g4
44
Mouse
TTAGCATGTTCCCAGTTTTC
268





mDMD-E44g5
44
Mouse
GACACAAAATCCTGAAAACT
269





mDMD-E44g6
44
Mouse
GGGAACATGCTAAATACAAA
270





mDMD-E44g7
44
Mouse
TAAATACAAATGGTATCTTA
271





mDMD-E44g8
44
Mouse
tcatccatgttttcaaatta
272





mDMD-E44g9
44
Mouse
tcaaattatagGCGATTCGA
273





mDMD-
44
Mouse
AAAAACTGTTCAACTTCATT
274


E44g10









mDMD-
44
Mouse
AATGGCTGAATGAAGTTGAA
275


E44g11









mDMD-
44
Mouse
AAGTTGAACAGTTTTTCAAA
276


E44g12









mDMD-
44
Mouse
TTTAGCATGTTCCCAGTTTT
277


E44g13









mDMD-
44
Mouse
CTTAAGATACCATTTGTATT
278


E44g14









mDMD-
44
Mouse
CTAAATACAAATGGTATCTT
279


E44g15









mDMD-
44
Mouse
TACAAATGGTATCTTAAGgt
280


E44g16









mDMD-
44
Mouse
AAATCTCAAAGTCTTACCTT
281


E44g17









mDMD-
44
Mouse
ttatagGCGATTCGACAGAT
282


E44g18









mDMD-
44
Mouse
catggatgaaataaggtaag
283


E44g19









mDMD-
44
Mouse
ctgaaaaaatgaagccagca
284


E44g20









mDMD-
44
Mouse
ATTTAATCAATGGCTGAATG
285


E44g21









mDMD-E46g1
46
Mouse
aattttgttattcttaatac
286





mDMD-E46g2
46
Mouse
AAATGAATTTGTTTTGTGGC
287





mDMD-E46g3
46
Mouse
AACATTGCTATTACTCCACT
288





mDMD-E46g4
46
Mouse
AGCTGCTGCTCATCTCCAAG
289





mDMD-E46g5
46
Mouse
AGAACAACTTGAACAAGTCA
290





mDMD-E46g6
46
Mouse
gaattttgttattcttaata
291





mDMD-E46g7
46
Mouse
TTGTTCTTCAATCctgtatt
292





mDMD-E46g8
46
Mouse
ttattcttaatacagGATTG
293





mDMD-E46g9
46
Mouse
aatacagGATTGAAGAACAA
294





mDMD-
46
Mouse
TTACTCCACTTGGAGATGAG
295


E46g10









mDMD-
46
Mouse
GCTAAAAGAACAACTTGAAC
296


E46g11









mDMD-
46
Mouse
gaaattacCTTGACTTGTTC
297


E46g12









mDMD-
46
Mouse
AAGAACAACTTGAACAAGTC
298


E46g13









mDMD-
46
Mouse
ACTTGTTCAAGTTGTTCTTT
299


E46g14









mDMD-
46
Mouse
acacctctcagggatttagg
300


E46g15









mDMD-
46
Mouse
ttcccttattaaaatcctca
301


E46g16









mDMD-
46
Mouse
ctttatacaaataggccctg
302


E46g17









mDMD-E51g4
51
Mouse
TGAAATGATCATCAAACAGA
303





mDMD-E51g5
51
Mouse
TCAATGAAATGATCATCAAA
304





mDMD-E51g6
51
Mouse
ATGAAATGATCATCAAACAG
305





mDMD-E51g7
51
Mouse
TGATCATCAAACAGAAGGTA
306





mDMD-E53g1
53
Mouse
TGAAAGAATTCAGATTCAGT
307





mDMD-E53g2
53
Mouse
AATTCAGATTCAGTGGGATG
308





mDMD-E53g3
53
Mouse
TTCAAGAACAGCTGCAGAAC
309





mDMD-E53g4
53
Mouse
ACAGTTGAATGAAATGTTAA
310





mDMD-E53g5
53
Mouse
TGTTAAAGGATTCAACACAA
311





mDMD-E53g6
53
Mouse
AAAGGATTCAACACAATGGC
312





mDMD-E53g7
53
Mouse
AAAGAAGATCACAGAAACCA
313





mDMD-E53g8
53
Mouse
TTGAAAGAATTCAGATTCAG
314





mDMD-E53g9
53
Mouse
AGTGGGATGAGGTTCAAGAA
315





mDMD-
53
Mouse
AGCTGCAGAACAGGAGACAA
316


E53g10









mDMD-
53
Mouse
TGAATCTGAATTCTTTCAAC
317


E53g11









mDMD-
53
Mouse
CTTTCAACTGGAATAAAAAT
318


E53g12









mDMD-
53
Mouse
CTTATTTTTATTCCAGTTGA
319


E53g13









mDMD-
53
Mouse
TTATTCCAGTTGAAAGAATT
320


E53g14









mDMD-
53
Mouse
CAGTTGAAAGAATTCAGATT
321


E53g15









mDMD-
53
Mouse
GAATTCAGATTCAGTGGGAT
322


E53g16









mDMD-
53
Mouse
GATTCAGTGGGATGAGGTTC
323


E53g17









mDMD-
53
Mouse
ATGAGGTTCAAGAACAGCTG
324


E53g18









mDMD-
53
Mouse
GTTCAAGAACAGCTGCAGAA
325


E53g19









mDMD-
53
Mouse
AACTGTTGTCTCCTGTTCTG
326


E53g20









mDMD-
53
Mouse
CAAGAACAGCTGCAGAACAG
327


E53g21









mDMD-
53
Mouse
AAGATCACAGAAACCAAGGT
328


E53g22









mDMD-
53
Mouse
CAGAAACCAAGGTTAGTGTC
329


E53g23









B. Scaffold


The scaffold sequence is the sequence within the gRNA that is responsible for nuclease (e.g., Cas9) binding. The scaffold sequence does not include the spacer/targeting sequence.


In some embodiments, the scaffold may be about 60 to about 70, about 70 to about 80, about 80 to about 90, about 90 to about 100, about 100 to about 110, about 110 to about 120, or about 120 to about 130 nucleotides in length. In some embodiments, the scaffold may be about 60, about 61, about 62, about 63, about 64, about 65, about 66, about 67, about 68, about 69, about 70, about 71, about 72, about 73, about 74, about 75, about 76, about 77, about 78, about 79, about 80, about 81, about 82, about 83, about 84, about 85, about 86, about 87, about 88, about 89, about 90, about 91, about 92, about 93, about 94, about 95, about 96, about 97, about 98, about 99, about 100, about 101, about 102, about 103, about 104, about 105, about 106, about 107, about 108, about 109, about 110, about 111, about 112, about 113, about 114, about 115, about 116, about 117, about 118, about 119, about 120, about 121, about 122, about 123, about 124, or about 125 nucleotides in length. In some embodiments, the scaffold may be at least 60, at least 70, at least 80, at least 90, at least 100, at least 110, at least 120, or at least 125 nucleotides in length. In some embodiments, the scaffold may be 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, or 125 nucleotides in length.


In some embodiments, the scaffold may comprise a sequence of any one of SEQ ID NOs: 147-153 (shown in Table 4 below), or a sequence that is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical thereto.









TABLE 4







Exemplary scaffold sequences









SEQ ID


Sequence
NO:





GTTTAAGAGCTATGCTGGAAACAGCATAGCAAGTTTAAAT
147


AAGGCTAGTCCGTTATCAACTTGAAAAAGTGGCACCGAGT



CGGTGCTTTTTTT






GTTTTAGAGCTAGAAATAGCAGTTAAAATAAGGCTAGTCC
148


GTTATCAACTTGAAAAAGTGGCACCGAGTCGGTG






GTTGGAACCATTCAAAACAGCATAGCAAGTTAAAATAAGG
149


CTAGTCCGTTATCAACTTGAAAAAGTGGCACCGAGTCGGT



GCTTTTTT






GTTTAAGAGCTATGAAACAGCATAGCAAGTTTAAATAAGG
150


CTAGTCCGTTATCAACTTGAAAAAGTGGCACCGAGTCGGT



GCTTTTTTT






GTTTAAGAGCTATGCGAAACAGCATAGCAAGTTTAAATAA
151


GGCTAGTCCGTTATCAACTTGAAAAAGTGGCACCGAGTCG



GTGCTTTTTTT






GTTTAAGAGCTATGCTGTTTGAAACAGCATAGCAAGTTTA
152


AATAAGGCTAGTCCGTTATCAACTTGAAAAAGTGGCACCG



AGTCGGTGCTTTTTTT






GTTTAAGAGCTATGCTGTTTTGGAAACAGCATAGCAAGTT
153


TAAATAAGGCTAGTCCGTTATCAACTTGAAAAAGTGGCAC



CGAGTCGGTGCTTTTTTT









In some embodiments, a gRNA (spacer+scaffold) comprises a scaffold and a spacer as shown in Table 5 below, wherein “X” indicates that the particular combination is contemplated by the instant disclosure.









TABLE 5







Exemplary sgRNA (spacer + scaffold) sequences














Spacer
Scaffold
Scaffold
Scaffold
Scaffold
Scaffold
Scaffold
Scaffold


Sequence
sequence
sequence
sequence
sequence
sequence
sequence
sequence


(SEQ
(SEQ ID
(SEQ ID
(SEQ ID
(SEQ ID
(SEQ ID
(SEQ ID
(SEQ ID


ID NO)
NO: 147)
NO: 148)
NO: 149)
NO: 150)
NO: 151)
NO: 152)
NO: 153)





135
X
X
X
X
X
X
X


136
X
X
X
X
X
X
X


137
X
X
X
X
X
X
X


138
X
X
X
X
X
X
X


139
X
X
X
X
X
X
X


140
X
X
X
X
X
X
X


141
X
X
X
X
X
X
X


142
X
X
X
X
X
X
X


143
X
X
X
X
X
X
X


144
X
X
X
X
X
X
X


145
X
X
X
X
X
X
X


146
X
X
X
X
X
X
X


170
X
X
X
X
X
X
X


171
X
X
X
X
X
X
X


172
X
X
X
X
X
X
X


173
X
X
X
X
X
X
X


174
X
X
X
X
X
X
X


175
X
X
X
X
X
X
X


176
X
X
X
X
X
X
X


177
X
X
X
X
X
X
X


178
X
X
X
X
X
X
X


179
X
X
X
X
X
X
X


180
X
X
X
X
X
X
X


181
X
X
X
X
X
X
X


182
X
X
X
X
X
X
X


183
X
X
X
X
X
X
X


184
X
X
X
X
X
X
X


185
X
X
X
X
X
X
X


186
X
X
X
X
X
X
X


187
X
X
X
X
X
X
X


188
X
X
X
X
X
X
X


189
X
X
X
X
X
X
X


190
X
X
X
X
X
X
X


191
X
X
X
X
X
X
X


192
X
X
X
X
X
X
X


193
X
X
X
X
X
X
X


194
X
X
X
X
X
X
X


195
X
X
X
X
X
X
X


196
X
X
X
X
X
X
X


197
X
X
X
X
X
X
X


198
X
X
X
X
X
X
X


199
X
X
X
X
X
X
X


200
X
X
X
X
X
X
X


201
X
X
X
X
X
X
X


202
X
X
X
X
X
X
X


203
X
X
X
X
X
X
X


204
X
X
X
X
X
X
X


205
X
X
X
X
X
X
X


206
X
X
X
X
X
X
X


207
X
X
X
X
X
X
X


208
X
X
X
X
X
X
X


209
X
X
X
X
X
X
X


210
X
X
X
X
X
X
X


211
X
X
X
X
X
X
X


212
X
X
X
X
X
X
X


213
X
X
X
X
X
X
X


214
X
X
X
X
X
X
X


215
X
X
X
X
X
X
X


216
X
X
X
X
X
X
X


217
X
X
X
X
X
X
X


218
X
X
X
X
X
X
X


219
X
X
X
X
X
X
X


220
X
X
X
X
X
X
X


221
X
X
X
X
X
X
X


222
X
X
X
X
X
X
X


223
X
X
X
X
X
X
X


224
X
X
X
X
X
X
X


225
X
X
X
X
X
X
X


226
X
X
X
X
X
X
X


227
X
X
X
X
X
X
X


228
X
X
X
X
X
X
X


229
X
X
X
X
X
X
X


230
X
X
X
X
X
X
X


231
X
X
X
X
X
X
X


232
X
X
X
X
X
X
X


233
X
X
X
X
X
X
X


234
X
X
X
X
X
X
X


235
X
X
X
X
X
X
X


236
X
X
X
X
X
X
X


237
X
X
X
X
X
X
X


238
X
X
X
X
X
X
X


239
X
X
X
X
X
X
X


240
X
X
X
X
X
X
X


241
X
X
X
X
X
X
X


242
X
X
X
X
X
X
X


243
X
X
X
X
X
X
X


244
X
X
X
X
X
X
X


245
X
X
X
X
X
X
X


246
X
X
X
X
X
X
X


247
X
X
X
X
X
X
X


248
X
X
X
X
X
X
X


249
X
X
X
X
X
X
X


250
X
X
X
X
X
X
X


251
X
X
X
X
X
X
X


252
X
X
X
X
X
X
X


253
X
X
X
X
X
X
X


254
X
X
X
X
X
X
X


255
X
X
X
X
X
X
X


256
X
X
X
X
X
X
X


257
X
X
X
X
X
X
X


258
X
X
X
X
X
X
X


259
X
X
X
X
X
X
X


260
X
X
X
X
X
X
X









In some embodiments, the sgRNA has a sequence (spacer+scaffold) of any one of SEQ ID NO: 154 to 165 (shown in Table 6, below), or a sequence that is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical thereto.









TABLE 6







Exemplary sgRNA (spacer + scaffold) sequences









SEQ ID


sgRNA sequence (spacer + scaffold)
NO





AAAGAAAATCACAGAAACCAGTTTAAGAGCTATGCTGGAA
154


ACAGCATAGCAAGTTTAAATAAGGCTAGTCCGTTATCAAC



TTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT






AATTCAGAATCAGTGGGATGGTTTAAGAGCTATGCTGGAA
155


ACAGCATAGCAAGTTTAAATAAGGCTAGTCCGTTATCAAC



TTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT






TTGAAAGAATTCAGAATCAGGTTTAAGAGCTATGCTGGAA
156


ACAGCATAGCAAGTTTAAATAAGGCTAGTCCGTTATCAAC



TTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT






CAAGAACACCTTCAGAACCGGTTTAAGAGCTATGCTGGAA
157


ACAGCATAGCAAGTTTAAATAAGGCTAGTCCGTTATCAAC



TTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT






TATGTGTTACCTACCCTTGTGTTTAAGAGCTATGCTGGAA
158


ACAGCATAGCAAGTTTAAATAAGGCTAGTCCGTTATCAAC



TTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT






GTACAAGGACCGACAAGGGTGTTTAAGAGCTATGCTGGAA
159


ACAGCATAGCAAGTTTAAATAAGGCTAGTCCGTTATCAAC



TTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT






TAAATACAAATGGTATCTTAGTTTAAGAGCTATGCTGGAA
160


ACAGCATAGCAAGTTTAAATAAGGCTAGTCCGTTATCAAC



TTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT






GGGAACATGCTAAATACAAAGTTTAAGAGCTATGCTGGAA
161


ACAGCATAGCAAGTTTAAATAAGGCTAGTCCGTTATCAAC



TTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT






ACAAATCAAAGACTTACCTTGTTTAAGAGCTATGCTGGAA
162


ACAGCATAGCAAGTTTAAATAAGGCTAGTCCGTTATCAAC



TTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT






AATTTTATTCTTCTTTCTCCGTTTAAGAGCTATGCTGGAA
163


ACAGCATAGCAAGTTTAAATAAGGCTAGTCCGTTATCAAC



TTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT






AGAAAAGCTTGAGCAAGTCAGTTTAAGAGCTATGCTGGAA
164


ACAGCATAGCAAGTTTAAATAAGGCTAGTCCGTTATCAAC



TTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT






TGTATGCTTTTCTGTTAAAGGTTTAAGAGCTATGCTGGAA
165


ACAGCATAGCAAGTTTAAATAAGGCTAGTCCGTTATCAAC



TTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT









In some embodiments, a nucleic acid comprises one copy of the sequence encoding the sgRNA. In some embodiments, a nucleic acid comprises two, three, four, or five copies of the sequence encoding the sgRNA.


In some embodiments, a nucleic acid comprises a sequence encoding a promoter, wherein the promoter drives expression of the sgRNA. In some embodiments, the nucleic acid comprises two copies of the sequence encoding a sgRNA, wherein expression of the first copy of the sgRNA is driven by a first promoter, and expression of the second copy of the sgRNA is driven by a second promoter.


In some embodiments, the nucleic acid comprises three copies of the sequence encoding a sgRNA, wherein expression of the first copy of the sgRNA is driven by a first promoter, expression of the second copy of the sgRNA is driven by a second promoter, and expression of the third copy of the sgRNA is driven by a third promoter.


In some embodiments, the nucleic acid comprises four copies of the sequence encoding a sgRNA, wherein expression of the first copy of the sgRNA is driven by a first promoter, expression of the second copy of the sgRNA is driven by a second promoter, expression of the third copy of the sgRNA is driven by a third promoter, and expression of the fourth copy of the sgRNA is driven by a fourth promoter.


In some embodiments, the nucleic acid comprises five copies of the sequence encoding a sgRNA, wherein expression of the first copy of the sgRNA is driven by a first promoter, expression of the second copy of the sgRNA is driven by a second promoter, expression of the third copy of the sgRNA is driven by a third promoter, expression of the fourth copy of the sgRNA is driven by a fourth promoter, and expression of the fifth copy of the sgRNA is driven by a fifth promoter.


In some embodiments, a nucleic acid sequence comprising a sequence encoding a sgRNA further comprises a sequence encoding a nuclease. The nuclease may be, for example, a Type II, Type V-A, Type V-B, Type V-C, Type V-U, or Type VI-B nuclease. Exemplary nucleases include, but are not limited to a TALEN, a meganuclease, a zinc-finger nuclease, or a Cas9, Cas12a, Cas12b, Cas12c, Tnp-B like, Cas13a (C2c2), Cas13b, or Cas14 nuclease.


In some embodiments, the nuclease is a Cas9 nuclease. In some embodiments, the Cas9 nuclease is a Streptococcus pyogenes or Streptococcus aureus Cas9. In some embodiments, the nuclease is a modified Cas9 nuclease. In some embodiments, the nuclease is a modified Streptococcus pyogenes Cas9 or a modified Streptococcus aureus Cas9.


CRISPRs (clustered regularly interspaced short palindromic repeats) are DNA loci containing short repetitions of base sequences. Each repetition is followed by short segments of “spacer DNA” from previous exposures to a virus. CRISPRs are found in approximately 40% of sequenced eubacteria genomes and 90% of sequenced archaea. CRISPRs are often associated with Cas genes that code for proteins related to CRISPRs. The CRISPR/Cas system is a prokaryotic immune system that confers resistance to foreign genetic elements such as plasmids and phages and provides a form of acquired immunity. CRISPR spacers recognize and silence these exogenous genetic elements like RNAi in eukaryotic organisms.


CRISPR repeats can range in size from 24 to 48 base pairs. They usually show some dyad symmetry, implying the formation of a secondary structure such as a hairpin, but are not truly palindromic. Repeats are separated by spacers of similar length.


CRISPR-associated (cas) genes are often associated with CRISPR repeat-spacer arrays. More than forty different Cas protein families have been described. Of these protein families, Cast appears to be ubiquitous among different CRISPR/Cas systems. Particular combinations of cas genes and repeat structures have been used to define 8 CRISPR subtypes (E. coli, Ypest, Nmeni, Dvulg, Tneap, Hmari, Apern, and Mtube), some of which are associated with an additional gene module encoding repeat-associated mysterious proteins (RAMPs). More than one CRISPR subtype may occur in a single genome. The sporadic distribution of the CRISPR/Cas subtypes suggests that the system is subject to horizontal gene transfer during microbial evolution.


Exogenous DNA is processed by proteins encoded by Cas genes into small elements (˜30 base pairs in length), which are then inserted into the CRISPR locus near the leader sequence. RNAs from the CRISPR loci are constitutively expressed and are processed by Cas proteins to small RNAs composed of individual, exogenously-derived sequence elements with a flanking repeat sequence. The RNAs guide other Cas proteins to silence exogenous genetic elements at the RNA or DNA level. Evidence suggests functional diversity among CRISPR subtypes. The Cse (Cas subtype E. coli) proteins (called CasA-E in E. coli) form a functional complex, Cascade, that processes CRISPR RNA transcripts into spacer-repeat units that Cascade retains. In other prokaryotes, Cas6 processes the CRISPR transcripts. Interestingly, CRISPR-based phage inactivation in E. coli requires Cascade and Cas3, but not Cas1 and Cas2. The Cmr (Cas RAMP module) proteins found in Pyrococcus furiosus and other prokaryotes form a functional complex with small CRISPR RNAs that recognizes and cleaves complementary target RNAs. RNA-guided CRISPR enzymes are classified as type V restriction enzymes.


Cas9 is a nuclease, an enzyme specialized for cutting DNA, with two active cutting sites, one for each strand of the double helix. One or both sites may be inactivated while preserving Cas9's ability to locate its target DNA. tracrRNA (i.e., a scaffold sequence) and spacer RNA may be combined into a “single-guide RNA” molecule that, mixed with Cas9, could find and cut the correct DNA targets. Such synthetic guide RNAs can be used for gene editing.


Cas9 proteins are highly enriched in pathogenic and commensal bacteria. CRISPR/Cas-mediated gene regulation may contribute to the regulation of endogenous bacterial genes, particularly during bacterial interaction with eukaryotic hosts. For example, Cas protein Cas9 of Francisella novicida uses a unique, small, CRISPR/Cas-associated RNA (scaRNA) to repress an endogenous transcript encoding a bacterial lipoprotein that is critical for F. novicida to dampen host response and promote virulence. Delivery of Cas9 DNA sequences also is contemplated.


Clustered Regularly Interspaced Short Palindromic Repeats from Prevotella and Francisella 1 or CRISPR/Cpf1 is a DNA-editing technology analogous to the CRISPR/Cas9 system. Cpf1 is an RNA-guided endonuclease of a class II CRISPR/Cas system. This acquired immune mechanism is found in Prevotella and Francisella bacteria. It prevents genetic damage from viruses. Cpf1 genes are associated with the CRISPR locus, coding for an endonuclease that use a guide RNA to find and cleave viral DNA. Cpf1 is a smaller and simpler endonuclease than Cas9, overcoming some of the CRISPR/Cas9 system limitations. CRISPR/Cpf1 has multiple applications, including treatment of genetic illnesses and degenerative conditions.


Cpf1 appears in many bacterial species. The Two Cpf1 enzymes from Acidaminococcus and Lachnospiraceae display efficient genome-editing activity in human cells.


A smaller version of Cas9 from the bacterium Staphylococcus aureus is a potential alternative to Cpf1.


The systems CRISPR/Cas are separated into three classes. Class 1 uses several Cas proteins together with the CRISPR RNAs (crRNA) to build a functional endonuclease. Class 2 CRISPR systems use a single Cas protein with a crRNA. Cpf1 has been recently identified as a Class II, Type V CRISPR/Cas systems containing a 1,300 amino acid protein.


The Cpf1 locus contains a mixed alpha/beta domain, a RuvC-I followed by a helical region, a RuvC-II and a zinc finger-like domain. The Cpf1 protein has a RuvC-like endonuclease domain that is similar to the RuvC domain of Cas9. Furthermore, Cpf1 does not have a HNH endonuclease domain, and the N-terminal of Cpf1 does not have the alfa-helical recognition lobe of Cas9.


Cpf1 CRISPR-Cas domain architecture shows that Cpf1 is functionally unique, being classified as Class 2, type V CRISPR system. The Cpf1 loci encode Cas1, Cas2 and Cas4 proteins more similar to types I and III than from type II systems. Database searches suggest the abundance of Cpf1-family proteins in many bacterial species.


Functional Cpf1 doesn't need the tracrRNA, therefore, only crRNA is required. This benefits genome editing because Cpf1 is not only smaller than Cas9, but also it has a smaller sgRNA molecule (proximately half as many nucleotides as Cas9).


The Cpf1-crRNA complex cleaves target DNA or RNA by identification of a protospacer adjacent motif 5′-YTN-3′ (where “Y” is a pyrimidine and “N” is any nucleobase) or 5′-TTN-3′, in contrast to the G-rich PAM targeted by Cas9. After identification of PAM, Cpf1 introduces a sticky-end-like DNA double-stranded break of 4 or 5 nucleotides overhang.


The CRISPR/Cpf1 system consist of a Cpf1 enzyme and a guide RNA that finds and positions the complex at the correct spot on the double helix to cleave target DNA. CRISPR/Cpf1 systems activity has three stages:

    • Adaptation, during which Cas1 and Cas2 proteins facilitate the adaptation of small fragments of DNA into the CRISPR array;
    • Formation of crRNAs: processing of pre-cr-RNAs producing of mature crRNAs to guide the Cas protein; and
    • Interference, in which the Cpf1 is bound to a crRNA to form a binary complex to identify and cleave a target DNA sequence.


II. NUCLEIC ACID DELIVERY

As discussed above, in certain embodiments, expression cassettes are employed to express a transcription factor product, either for subsequent purification and delivery to a cell/subject, or for use directly in a genetic-based delivery approach. Expression requires that appropriate signals be provided in the vectors, and include various regulatory elements such as enhancers/promoters from both viral and mammalian sources that drive expression of the genes of interest in cells. Elements designed to optimize messenger RNA stability and translatability in host cells also are defined. The conditions for the use of a number of dominant drug selection markers for establishing permanent, stable cell clones expressing the products are also provided, as is an element that links expression of the drug selection markers to expression of the polypeptide.


A. Regulatory Elements


Throughout this application, the term “expression cassette” is meant to include any type of genetic construct containing a nucleic acid coding for a gene product in which part or all of the nucleic acid encoding sequence is capable of being transcribed and translated, i.e., is under the control of a promoter. A “promoter” refers to a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, responsible for initiating the specific transcription of a gene. The phrase “under transcriptional control” means that the promoter is in the correct location and orientation in relation to the nucleic acid to control RNA polymerase initiation and expression of the gene. An “expression vector” is meant to include expression cassettes comprised in a genetic construct that is capable of replication, and thus including one or more of origins of replication, transcription termination signals, poly-A regions, selectable markers, and multipurpose cloning sites.


The term promoter will be used here to refer to a group of transcriptional control modules that are clustered around the initiation site for RNA polymerase II. Much of the thinking about how promoters are organized derives from analyses of several viral promoters, including those for the HSV thymidine kinase (tk) and SV40 early transcription units. These studies, augmented by more recent work, have shown that promoters are composed of discrete functional modules, each consisting of approximately 7-20 bp of DNA, and containing one or more recognition sites for transcriptional activator or repressor proteins.


At least one module in each promoter functions to position the start site for RNA synthesis. The best-known example of this is the TATA box, but in some promoters lacking a TATA box, such as the promoter for the mammalian terminal deoxynucleotidyl transferase gene and the promoter for the SV40 late genes, a discrete element overlying the start site itself helps to fix the place of initiation.


In some embodiments, the sgRNA, Cas9 or Cpf1 constructs of the disclosure are expressed by a muscle-cell specific promoter. This muscle-cell specific promoter may be constitutively active or may be an inducible promoter.


Additional promoter elements regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have recently been shown to contain functional elements downstream of the start site as well. The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the tk promoter, the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline. Depending on the promoter, it appears that individual elements can function either co-operatively or independently to activate transcription.


In certain embodiments, viral promoters such as the human cytomegalovirus (CMV) immediate early gene promoter, the SV40 early promoter, the Rous sarcoma virus long terminal repeat, rat insulin promoter and glyceraldehyde-3-phosphate dehydrogenase can be used to obtain high-level expression of the coding sequence of interest. The use of other viral or mammalian cellular or bacterial phage promoters which are well-known in the art to achieve expression of a coding sequence of interest is contemplated as well, provided that the levels of expression are sufficient for a given purpose. By employing a promoter with well-known properties, the level and pattern of expression of the protein of interest following transfection or transformation can be optimized. Further, selection of a promoter that is regulated in response to specific physiologic signals can permit inducible expression of the gene product.


Enhancers are genetic elements that increase transcription from a promoter located at a distant position on the same molecule of DNA. Enhancers are organized much like promoters. That is, they are composed of many individual elements, each of which binds to one or more transcriptional proteins. The basic distinction between enhancers and promoters is operational. An enhancer region as a whole is able to stimulate transcription at a distance; this need not be true of a promoter region or its component elements. On the other hand, a promoter has one or more elements that direct initiation of RNA synthesis at a particular site and in a particular orientation, whereas enhancers lack these specificities. Promoters and enhancers are often overlapping and contiguous, often seeming to have a very similar modular organization.


Below is a list of promoters/enhancers and inducible promoters/enhancers that could be used to drive expression of a nucleic acid encoding a gene of interest in an expression construct. Additionally, any promoter/enhancer combination (as per the Eukaryotic Promoter Data Base EPDB) could also be used to drive expression of the gene. Eukaryotic cells can support cytoplasmic transcription from certain bacterial promoters if the appropriate bacterial polymerase is provided, either as part of the delivery complex or as an additional genetic expression construct.


The promoter and/or enhancer may be, for example, immunoglobulin light chain, immunoglobulin heavy chain, T-cell receptor, HLA DQ a and/or DQ β, β-interferon, interleukin-2, interleukin-2 receptor, MHC class II 5, MHC class II HLA-Dra, β-Actin, muscle creatine kinase (MCK), prealbumin (transthyretin), elastase I, metallothionein collagenase, albumin, α-fetoprotein, t-globin, β-fos, c-HA-ras, insulin, neural cell adhesion molecule (NCAM), α1-antitrypain, H2B (TH2B) histone, mouse and/or type I collagen, glucose-regulated proteins (GRP94 and GRP78), rat growth hormone, human serum amyloid A (SAA), troponin I (TN I), platelet-derived growth factor (PDGF), duchenne muscular dystrophy, SV40, polyoma, retroviruses, papilloma virus, hepatitis B virus, human immunodeficiency virus, cytomegalovirus (CMV), and gibbon ape leukemia virus.


In some embodiments, inducible elements may be used. In some embodiments, the inducible element is, for example, MTII, MMTV (mouse mammary tumor virus), β-interferon, adenovirus 5 E2, collagenase, stromelysin, SV40, murine MX gene, GRP78 gene, α-2-macroglobulin, vimentin, MHC class I gene H-2κb, HSP70, proliferin, tumor necrosis factor, and/or thyroid stimulating hormone a gene. In some embodiments, the inducer is phorbol ester (TFA), heavy metals, glucocorticoids, poly(rDx, poly(rc), ElA, phorbol ester (TPA), interferon, Newcastle Disease Virus, A23187, IL-6, serum, interferon, SV40 large T antigen, PMA, and/or thyroid hormone. Any of the inducible elements described herein may be used with any of the inducers described herein.


Of particular interest are muscle specific promoters. These include the myosin light chain-2 promoter, the α-actin promoter, the troponin 1 promoter; the Na+/Ca2+ exchanger promoter, the dystrophin promoter, the α7 integrin promoter, the brain natriuretic peptide promoter and the αB-crystallin/small heat shock protein promoter, α-myosin heavy chain promoter and the ANF promoter.


In some embodiments, the muscle specific promoter is the CK8 promoter. The CK8 promoter has the following sequence (SEQ ID NO: 331):










1
CTAGACTAGC ATGCTGCCCA TGTAAGGAGG CAAGGCCTGG



GGACACCCGA GATGCCTGGT





61
TATAATTAAC CCAGACATGT GGCTGCCCCC CCCCCCCCAA



CACCTGCTGC CTCTAAAAAT





121
AACCCTGCAT GCCATGTTCC CGGCGAAGGG CCAGCTGTCC



CCCGCCAGCT AGACTCAGCA





181
CTTAGTTTAG GAACCAGTGA GCAAGTCAGC CCTTGGGGCA



GCCCATACAA GGCCATGGGG





241
CTGGGCAAGC TGCACGCCTG GGTCCGGGGT GGGCACGGTG



CCCGGGCAAC GAGCTGAAAG





301
CTCATCTGCT CTCAGGGGCC CCTCCCTGGG GACAGCCCCT



CCTGGCTAGT CACACCCTGT





361
AGGCTCCTCT ATATAACCCA GGGGCACAGG GGCTGCCCTC



ATTCTACCAC CACCTCCACA





421
GCACAGACAG ACACTCAGGA GCCAGCCAGC.






In some embodiments, the muscle-cell cell specific promoter is a variant of the CK8 promoter, called CK8e. The CK8e promoter has the following sequence (SEQ ID NO. 332):










1
TGCCCATGTA AGGAGGCAAG GCCTGGGGAC ACCCGAGATG



CCTGGTTATA ATTAACCCAG





61
ACATGTGGCT GCCCCCCCCC CCCCAACACC TGCTGCCTCT



AAAAATAACC CTGCATGCCA





121
TGTTCCCGGC GAAGGGCCAG CTGTCCCCCG CCAGCTAGAC



TCAGCACTTA GTTTAGGAAC





181
CAGTGAGCAA GTCAGCCCTT GGGGCAGCCC ATACAAGGCC



ATGGGGCTGG GCAAGCTGCA





241
CGCCTGGGTC CGGGGTGGGC ACGGTGCCCG GGCAACGAGC



TGAAAGCTCA TCTGCTCTCA





301
GGGGCCCCTC CCTGGGGACA GCCCCTCCTG GCTAGTCACA



CCCTGTAGGC TCCTCTATAT





361
AACCCAGGGG CACAGGGGCT GCCCTCATTC TACCACCACC



TCCACAGCAC AGACAGACAC





421
TCAGGAGCCA GCCAGC.






Where a cDNA insert is employed, one will typically desire to include a polyadenylation signal to effect proper polyadenylation of the gene transcript. Any polyadenylation sequence may be employed such as human growth hormone and SV40 polyadenylation signals. Also contemplated as an element of the expression cassette is a terminator. These elements can serve to enhance message levels and to minimize read through from the cassette into other sequences.


B. Self-Cleaving Peptides


In some embodiments, the nucleic acids and/or expression constructs disclosed herein may encode a self-cleaving peptide.


In some embodiments of self-cleaving peptides of the disclosure, the self-cleaving peptide is a 2A peptide. In some embodiments, a 2A-like self-cleaving domain from the insect virus Thosea asigna (TaV 2A peptide) (SEQ ID NO: 333, EGRGSLLTCGDVEENPGP) is used. These 2A-like domains have been shown to function across eukaryotes and cause cleavage of amino acids to occur co-translationally within the 2A-like peptide domain. Therefore, inclusion of TaV 2A peptide allows the expression of multiple proteins from a single mRNA transcript. Importantly, the domain of TaV when tested in eukaryotic systems has shown greater than 99% cleavage activity. Other acceptable 2A-like peptides include, but are not limited to, equine rhinitis A virus (ERAV) 2A peptide (SEQ ID NO: 334; QCTNYALLKLAGDVESNPGP), porcine teschovirus-1 (PTV1) 2A peptide (SEQ ID NO: 335; ATNFSLLKQAGDVEENPGP) and foot and mouth disease virus (FMDV) 2A peptide (SEQ ID NO: 336; PVKQLLNFDLLKLAGDVESNPGP) or modified versions thereof.


In some embodiments, the 2A peptide is used to express a reporter and a Cas9 or a Cpf1 simultaneously. The reporter may be, for example, GFP.


Other self-cleaving peptides that may be used include, but are not limited to nuclear inclusion protein a (Nia) protease, a P1 protease, a 3C protease, an L protease, a 3C-like protease, or modified versions thereof.


C. Delivery of Expression Vectors


There are a number of other ways in which expression vectors may introduced into cells. In certain embodiments, the expression construct comprises a virus or engineered construct derived from a viral genome. The ability of certain viruses to enter cells via receptor-mediated endocytosis, to integrate into host cell genome and express viral genes stably and efficiently have made them attractive candidates for the transfer of foreign genes into mammalian cells. In some embodiments, the gene editing compositions described herein are administered to a cell or to a subject using a non-viral vector or a viral vector. In some embodiments, the gene editing compositions described herein are administered to a cell or to a subject using a recombinant vector (e.g., a recombinant viral or a recombinant non-viral vector). In some embodiments, a recombinant vector comprises a nucleic acid of the disclosure, i.e., a nucleic acid comprising a sequence encoding a single guide RNA (sgRNA) comprising a spacer sequence and a scaffold sequence, wherein the spacer sequence targets an exon sequence of the DMD gene, such as a sequence of exon 43, 44, 46, 50, or 53. In some embodiments, the recombinant vector is a plasmid. In some embodiments, the recombinant vector is an expression vector.


Exemplary non-viral vectors for use with the compositions and methods described herein comprise nanoparticles (e.g., polymeric nanoparticles), liposomes (e.g., cationic liposomes), naked DNA, cationic lipid-DNA complexes, lipid emulsions, calcium phosphate, polymer complexes, or combinations thereof.


Exemplary viral vectors for use with the compositions and methods described herein include vectors based on adeno-associated virus (AAV), adenovirus, lentivirus, retrovirus, or a hybrid virus. In some embodiments, the viral vectors of the instant disclosure are replication defective, or at least conditionally replication defective.


The AAV genome may be from any naturally derived serotype or isolate or clade of AAV. Thus, the AAV genome may be the full genome of a naturally occurring AAV virus. As is known to the skilled person, AAV viruses occurring in nature may be classified according to various biological systems.


Commonly, AAV viruses are referred to in terms of their serotype. A serotype corresponds to a variant subspecies of AAV which owing to its profile of expression of capsid surface antigens has a distinctive reactivity which can be used to distinguish it from other variant subspecies. Typically, a virus having a particular AAV serotype does not efficiently cross-react with neutralizing antibodies specific for any other AAV serotype. AAV serotypes include AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10 and AAV11, also recombinant serotypes, such as Rec2 and Rec3, recently identified from primate brain. The sequences of AAV genomes or of elements of AAV genomes including ITR sequences, rep or cap genes for use methods and compositions described herein may be derived from the following accession numbers for AAV whole genome sequences: Adeno-associated virus 1 NC_002077, AF063497; Adeno-associated virus 2 NC_001401; Adeno-associated virus 3 NC_001729; Adeno-associated virus 3B NC_001863; Adeno-associated virus 4 NC_001829; Adeno-associated virus 5 Y18065, AF085716; Adeno-associated virus 6 NC_001862; Avian AAV ATCC VR-865 AY186198, AY629583, NC_004828; Avian AAV strain DA-1 NC_006263, AY629583; Bovine AAV NC_005889, AY388617.


AAV viruses may also be referred to in terms of clades or clones. This refers to the phylogenetic relationship of naturally derived AAV viruses, and typically to a phylogenetic group of AAV viruses which can be traced back to a common ancestor, and includes all descendants thereof. Additionally, AAV viruses may be referred to in terms of a specific isolate, i.e., a genetic isolate of a specific AAV virus found in nature. The term genetic isolate describes a population of AAV viruses which has undergone limited genetic mixing with other naturally occurring AAV viruses, thereby defining a recognizably distinct population at a genetic level.


In some embodiments, the gene editing compositions of the instant disclosure are delivered to a cell or to a patient using one or more AAV vectors. An AAV vector typically comprises an AAV expression cassette encapsidated by an AAV capsid protein. The serotype of the AAV vector may be selected from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVRh74, AAV2i8, AAVRh10, AAV39, AAV43, AAVRh8, avian AAV, bovine AAV, canine AAV, equine AAV, and ovine AAV. In some embodiments, the AAV vector may be replication-defective or conditionally replication defective.


In some embodiments, the AAV vector is selected from any of the AAV vectors disclosed in Table 1 of WO 2019/028306, which is incorporated by reference herein in its entirety. In some embodiments, the AAV vector is selected from one of the serotypes listed in Table 7.









TABLE 7







AAV Serotypes











Seq



Serotype
ID No.







VOY101
1001



VOY201
2260



PHP.N/PHP.B-DGT
1002



AAVPHP.B or G2B-26
1003



AAVPHP.B
1004



AAVG2B-13
1005



AAVTH1.1-32
1006



AAVTH1.1-35
1007



PHP.S/G2Al2
1008



AAV9/hu.14 K449R
1009



AAV1
1010



AAV1
1011



AAV1
1012



AAV1.3
1013



AAV1O
1014



AAV1O
1015



AAV1O
1016



AAV11
1017



AAV12
1018



AAV2
1019



AAV2
1020



AAV2
1021



AAV2
1022



AAV2
1023



AAV2.5T
1024



AAV223.10
1025



AAV223.2
1026



AAV223.2
1027



AAV223.4
1028



AAV223.4
1029



AAV223.5
1030



AAV223.5
1031



AAV223.6
1032



AAV223.6
1033



AAV223.7
1034



AAV223.7
1035



AAV29.3
1036



AAV29.4
1037



AAV29.5
1038



AAV29.5 (AAVbb.2)
1039



AAV3
1040



AAV3
1041



AAV3
1042



AAV3.3b
1043



AAV3-3
1044



AAV3-3
1045



AAV3a
1046



AAV3a
1047



AAV3b
1048



AAV3b
1049



AAV3b
1050



AAV4
1051



AAV4
1052



AAV4
1053



AAV4
1054



AAV4
1055



AAV4
1056



AAV4
1057



AAV4
1058



AAV4
1059



AAV4
1060



AAV4
1061



AAV4
1062



AAV4
1063



AAV4
1064



AAV4
1065



AAV4
1066



AAV4
1067



AAV4
1068



AAV4
1069



AAV4
1070



AAV42.2
1071



AAV42.2
1072



AAV42.3b
1073



AAV42.3B
1074



AAV42.4
1075



AAV42.4
1076



AAV42.8
1077



AAV42.8
1078



AAV43.1
1079



AAV43.1
1080



AAV43.12
1081



AAV43.12
1082



AAV43.20
1083



AAV43.20
1084



AAV43.21
1085



AAV43.21
1086



AAV43.23
1087



AAV43.23
1088



AAV43.25
1089



AAV43.25
1090



AAV43.5
1091



AAV43.5
1092



AAV4-4
1093



AAV4-4
1094



AAV44.1
1095



AAV44.1
1096



AAV44.5
1097



AAV44.5
1098



AAV4407
1099



AAV5
1100



AAV5
1101



AAV5
1102



AAV5
1103



AAV6
1104



AAV6
1105



AAV6
1106



AAV6
1107



AAV6
1108



AAV6
1109



AAV6.1
1110



AAV6.12
1111



AAV6.2
1112



AAV7
1113



AAV7
1114



AAV7
1115



AAV7
1116



AAV7
1117



AAV7
1118



AAV7
1119



AAV8
1120



AAV8
1121



AAV8
1122



AAV8
1123



AAV8
1124



AAV8
1125



AAV-8b
1126



AAV-8b
1127



AAV-8h
1128



AAV-8h
1129



AAV9
1130



AAV9
1131



AAV9
1132



AAV9
1133



AAV9
1134



AAV9 (AAVhu.14)
1135



AAV9 (AAVhu.14)
1136



AAVA3.1
1137



AAVA3.3
1138



AAVA3.3
1139



AAVA3.4
1140



AAVA3.4
1141



AAVA3.5
1142



AAVA3.5
1143



AAVA3.7
1144



AAVA3.7
1145



AAV29.3 (AAVbb.1)
1146



AAVC2
1147



AAVCh.5
1148



AAVcy.2 (AAV13.3)
1149



AAV24.1
1150



AAVcy.3 (AAV24.1)
1151



AAV27.3
1152



AAVcy.4 (AAV27.3)
1153



AAVcy.5
1154



AAV7.2
1155



AAVcy.5 (AAV7.2)
1156



AAV16.3
1157



AAVcy.6 (AAV16.3)
1158



AAVcy.5
1159



AAVcy.5
1160



AAVCy.5R1
1161



AAVCy.5R2
1162



AAVCy.5R3
1163



AAVCy.5R4
1164



AAVDJ
1165



AAVDJ
1166



AAVDJ-8
1167



AAVDJ-8
1168



AAVF5
1169



AAVH2
1170



AAVH6
1171



AAVhE1.1
1172



AAVhEr1.14
1173



AAVhEr1.16
1174



AAVhEr1.18
1175



AAVhEr1.23 (AAVhEr2.29)
1176



AAVhEr1.35
1177



AAVhEr1.36
1178



AAVhEr1.5
1179



AAVhEr1.7
1180



AAVhEr1.8
1181



AAVhEr2.16
1182



AAVhEr2.30
1183



AAVhEr2.31
1184



AAVhEr2.36
1185



AAVhEr2.4
1186



AAVhEr3.1
1187



AAVhu.1
1188



AAVhu.1
1189



AAVhu.10 (AAV16.8)
1190



AAVhu.10 (AAV16.8)
1191



AAVhu.11 (AAV16.12)
1192



AAVhu.11 (AAV16.12)
1193



AAVhu.12
1194



AAVhu.12
1195



AAVhu.13
1196



AAVhu.13
1197



AAVhu.136.1
1198



AAVhu.140.1
1199



AAVhu.140.2
1200



AAVhu.145.6
1201



AAVhu.15
1202



AAVhu.15 (AAV33.4)
1203



AAVhu.156.1
1204



AAVhu.16
1205



AAVhu.16 (AAV33.8)
1206



AAVhu.17
1207



AAVhu.17 (AAV33.12)
1208



AAVhu.172.1
1209



AAVhu.172.2
1210



AAVhu.173.4
1211



AAVhu.173.8
1212



AAVhu.18
1213



AAVhu.18
1214



AAVhu.19
1215



AAVhu.19
1216



AAVhu.2
1217



AAVhu.2
1218



AAVhu.20
1219



AAVhu.20
1220



AAVhu.21
1221



AAVhu.21
1222



AAVhu.22
1223



AAVhu.22
1224



AAVhu.23
1225



AAVhu.23.2
1226



AAVhu.24
1227



AAVhu.24
1228



AAVhu.25
1229



AAVhu.25
1230



AAVhu.26
1231



AAVhu.26
1232



AAVhu.27
1233



AAVhu.27
1234



AAVhu.28
1235



AAVhu.28
1236



AAVhu.29
1237



AAVhu.29
1238



AAVhu.29
1239



AAVhu.29R
1240



AAVhu.3
1241



AAVhu.3
1242



AAVhu.30
1243



AAVhu.30
1244



AAVhu.31
1245



AAVhu.31
1246



AAVhu.32
1247



AAVhu.32
1248



AAVhu.33
1249



AAVhu.33
1250



AAVhu.34
1251



AAVhu.34
1252



AAVhu.35
1253



AAVhu.35
1254



AAVhu.36
1255



AAVhu.36
1256



AAVhu.37
1257



AAVhu.37 (AAV106.1)
1258



AAVhu.38
1259



AAVhu.39
1260



AAVhu.39 (AAVLG-9)
1261



AAVhu.4
1262



AAVhu.4
1263



AAVhu.40
1264



AAVhu.40 (AAV1 14.3)
1265



AAVhu.41
1266



AAVhu.41 (AAV127.2)
1267



AAVhu.42
1268



AAVhu.42 (AAV127.5)
1269



AAVhu.43
1270



AAVhu.43
1271



AAVhu.43 (AAV128.1)
1272



AAVhu.44
1273



AAVhu.44 (AAV128.3)
1274



AAVhu.44R1
1275



AAVhu.44R2
1276



AAVhu.44R3
1277



AAVhu.45
1278



AAVhu.45
1279



AAVhu.46
1280



AAVhu.46
1281



AAVhu.46
1282



AAVhu.47
1283



AAVhu.47
1284



AAVhu.48
1285



AAVhu.48
1286



AAVhu.48 (AAV130.4)
1287



AAVhu.48R1
1288



AAVhu.48R2
1289



AAVhu.48R3
1290



AAVhu.49
1291



AAVhu.49
1292



AAVhu.5
1293



AAVhu.5
1294



AAVhu.51
1295



AAVhu.51
1296



AAVhu.52
1297



AAVhu.52
1298



AAVhu.53
1299



AAVhu.53
1300



AAVhu.53 (AAV145.1)
1301



AAVhu.54
1302



AAVhu.54 (AAV145.5)
1303



AAVhu.55
1304



AAVhu.56
1305



AAVhu.56 (AAV145.6)
1306



AAVhu.56 (AAV145.6)
1307



AAVhu.57
1308



AAVhu.57
1309



AAVhu.57
1310



AAVhu.58
1311



AAVhu.58
1312



AAVhu.6 (AAV3.1)
1313



AAVhu.6 (AAV3.1)
1314



AAVhu.60
1315



AAVhu.60 (AAV161.10)
1316



AAVhu.61
1317



AAVhu.61 (AAV161.6)
1318



AAVhu.63
1319



AAVhu.63
1320



AAVhu.64
1321



AAVhu.64
1322



AAVhu.66
1323



AAVhu.67
1324



AAVhu.67
1325



AAVhu.7
1326



AAVhu.7
1327



AAVhu.7 (AAV7.3)
1328



AAVhu.71
1329



AAVhu.8
1330



AAVhu.8
1331



AAVhu.8
1332



AAVhu.9 (AAV3.1)
1333



AAVhu.9 (AAV3.1)
1334



AAV-LKO1
1335



AAV-LKO1
1336



AAV-LK02
1337



AAV-LK02
1338



AAV-LK03
1339



AAV-LK03
1340



AAV-LK04
1341



AAV-LK04
1342



AAV-LK05
1343



AAV-LK05
1344



AAV-LK06
1345



AAV-LK06
1346



AAV-LK07
1347



AAV-LK07
1348



AAV-LK08
1349



AAV-LK08
1350



AAV-LK09
1351



AAV-LK09
1352



AAV-LK1O
1353



AAV-LK1O
1354



AAV-LK11
1355



AAV-LK11
1356



AAV-LK12
1357



AAV-LK12
1358



AAV-LK13
1359



AAV-LK13
1360



AAV-LK14
1361



AAV-LK14
1362



AAV-LK15
1363



AAV-LK15
1364



AAV-LK16
1365



AAV-LK16
1366



AAV-LK17
1367



AAV-LK17
1368



AAV-LK18
1369



AAV-LK18
1370



AAV-LK19
1371



AAV-LK19
1372



AAV-PAEC
1373



AAV-PAEC
1374



AAV-PAEC11
1375



AAV-PAEC11
1376



AAV-PAEC12
1377



AAV-PAEC12
1378



AAV-PAEC13
1379



AAV-PAEC13
1380



AAV-PAEC2
1381



AAV-PAEC2
1382



AAV-PAEC4
1383



AAV-PAEC4
1384



AAV-PAEC6
1385



AAV-PAEC6
1386



AAV-PAEC7
1387



AAV-PAEC7
1388



AAV-PAEC8
1389



AAV-PAEC8
1390



AAVpi.1
1391



AAVpi.1
1392



AAVpi.2
1393



AAVpi.2
1394



AAVpi.3
1395



AAVpi.3
1396



AAVrh.10
1397



AAVrh.10
1398



AAV44.2
1399



AAVrh.10 (AAV44.2)
1400



AAV42.1B
1401



AAVrh.12 (AAV42.1b)
1402



AAVrh.13
1403



AAVrh.13
1404



AAVrh.13
1405



AAVrh.13R
1406



AAV42.3A
1407



AAVrh.14 (AAV42.3a)
1408



AAV42.5A
1409



AAVrh.17 (AAV42.5a)
1410



AAV42.5B
1411



AAVrh.18 (AAV42.5b)
1412



AAV42.6B
1413



AAVrh.19 (AAV42.6b)
1414



AAVrh.2
1415



AAVrh.2
1416



AAVrh.20
1417



AAV42.10
1418



AAVrh.21 (AAV42.10)
1419



AAV42.11
1420



AAVrh.22 (AAV42 .11)
1421



AAV42.12
1422



AAVrh.23 (AAV42.12)
1423



AAV42.13
1424



AAVrh.24 (AAV42.13)
1425



AAV42.15
1426



AAVrh.25 (AAV42.15)
1427



AAVrh.2R
1428



AAVrh.31 (AAV223.1)
1429



AAVC1
1430



AAVrh.32 (AAVC1)
1431



AAVrh.32/33
1432



AAVrh.33 (AAVC3)
1433



AAVC5
1434



AAVrh.34 (AAVC5)
1435



AAVF1
1436



AAVrh.35 (AAVF1)
1437



AAVF3
1438



AAVrh.36 (AAVF3)
1439



AAVrh.37
1440



AAVrh.37
1441



AAVrh.37
1442



AAVrh.37R2
1443



AAVrh.38 (AAVLG-4)
1444



AAVrh.38 (AAVLG-4)
1445



AAVrh.39
1446



AAVrh.39
1447



AAVrh.40
1448



AAVrh.40 (AAVLG-10)
1449



AAVrh.43 (AAVN721-8)
1450



AAVrh.43 (AAVN721-8)
1451



AAVrh.44
1452



AAVrh.44
1453



AAVrh.45
1454



AAVrh.45
1455



AAVrh.46
1456



AAVrh.46
1457



AAVrh.47
1458



AAVrh.47 (AAVbb.2)
1459



AAVrh.48
1460



AAVrh.48.1
1461



AAVrh.48.1.2
1462



AAVrh.48.2
1463



AAVrh.48 (AAV1-7)
1464



AAVrh.49 (AAV1-8)
1465



AAVrh.49 (AAV1-8)
1466



AAVrh.50 (AAV2-4)
1467



AAVrh.50 (AAV2-4)
1468



AAVrh.51 (AAV2-5)
1469



AAVrh.51 (AAV2-5)
1470



AAVrh.52 (AAV3-9)
1471



AAVrh.52 (AAV3-9)
1472



AAVrh.53
1473



AAVrh.53 (AAV3-11)
1474



AAVrh.53 (AAV3-11)
1475



AAVrh.54
1476



AAVrh.54
1477



AAVrh.55
1478



AAVrh.55 (AAV4-19)
1479



AAVrh.56
1480



AAVrh.56
1481



AAVrh.57
1482



AAVrh.57
1483



AAVrh.58
1484



AAVrh.58
1485



AAVrh.58
1486



AAVrh.59
1487



AAVrh.59
1488



AAVrh.60
1489



AAVrh.60
1490



AAVrh.61
1491



AAVrh.61 (AAV2-3)
1492



AAVrh.62 (AAV2-15)
1493



AAVrh.62 (AAV2-15)
1494



AAVrh.64
1495



AAVrh.64
1496



AAVrh.64
1497



AAVRh.64R1
1498



AAVRh.64R2
1499



AAVrh.65
1500



AAVrh.65
1501



AAVrh.67
1502



AAVrh.67
1503



AAVrh.67
1504



AAVrh.68
1505



AAVrh.68
1506



AAVrh.69
1507



AAVrh.69
1508



AAVrh.70
1509



AAVrh.70
1510



AAVrh.71
1511



AAVrh.72
1512



AAVrh.73
1513



AAVrh.74
1514



AAVrh.8
1515



AAVrh.8
1516



AAVrh.8R
1517



AAVrh.8R A586R mutant
1518



AAVrh.8R R533A mutant
1519



BAAV (bovine AAV)
1520



BAAV (bovine AAV)
1521



BAAV (bovine AAV)
1522



BAAV (bovine AAV)
1523



BAAV (bovine AAV)
1524



BAAV (bovine AAV)
1525



BAAV (bovine AAV)
1526



BAAV (bovine AAV)
1527



BAAV (bovine AAV)
1528



BAAV (bovine AAV)
1529



BAAV (bovine AAV)
1530



BAAV (bovine AAV)
1531



BAAV (bovine AAV)
1532



BNP61 AAV
1533



BNP61 AAV
1534



BNP62AAV
1535



BNP63 AAV
1536



caprine AAV
1537



caprine AAV
1538



true type AAV (ttAAV)
1539



AAAV (Avian AAV)
1540



AAAV (Avian AAV)
1541



AAAV (Avian AAV)
1542



AAAV (Avian AAV)
1543



AAAV (Avian AAV)
1544



AAAV (Avian AAV)
1545



AAAV (Avian AAV)
1546



AAAV (Avian AAV)
1547



AAAV (Avian AAV)
1548



AAAV (Avian AAV)
1549



AAAV (Avian AAV)
1550



AAAV (Avian AAV)
1551



AAAV (Avian AAV)
1552



AAAV (Avian AAV)
1553



AAAV (Avian AAV)
1554



AAV Shuffle 100-1
1555



AAV Shuffle 100-1
1556



AAV Shuffle 100-2
1557



AAV Shuffle 100-2
1558



AAV Shuffle 100-3
1559



AAV Shuffle 100-3
1560



AAV Shuffle 100-7
1561



AAV Shuffle 100-7
1562



AAV Shuffle 10-2
1563



AAV Shuffle 10-2
1564



AAV Shuffle 10-6
1565



AAV Shuffle 10-6
1566



AAV Shuffle 10-8
1567



AAV Shuffle 10-8
1568



AAVSM 100-10
1569



AAVSM 100-10
1570



AAVSM 100-3
1571



AAVSM 100-3
1572



AAVSM 10-1
1573



AAVSM 10-1
1574



AAVSM 10-2
1575



AAVSM 10-2
1576



AAVSM 10-8
1577



AAVSM 10-8
1578



AAVF1/HSC1
1579



AAVF2/HSC2
1580



AAVF3/HSC3
1581



AAVF4/HSC4
1582



AAVF5/HSC5
1583



AAVF6/HSC6
1584



AAVF7/HSC7
1585



AAVF8/HSC8
1586



AAVF9/HSC9
1587



AAVF1 1/HSC11
1588



AAVF12/HSC12
1589



AAVF13/HSC13
1590



AAVF14/HSC14
1591



AAVF15/HSC15
1592



AAVF16/HSC16
1593



AAVF17/HSC17
1594



AAVF1/HSC1
1595



AAVF2/HSC2
1596



AAVF3/HSC3
1597



AAVF4/HSC4
1598



AAVF5/HSC5
1599



AAVF6/HSC6
1600



AAVF7/HSC7
1601



AAVF8/HSC8
1602



AAVF9/HSC9
1603



AAVF1 1/HSC1 1
1604



AAVF12/HSC12
1605



AAVF13/HSC13
1606



AAVF14/HSC14
1607



AAVF15/HSC15
1608



AAVF16/HSC16
1609



AAVF17/HSC17
1610



AAVCBr-E1
1611



AAVCBr-E2
1612



AAVCBr-E3
1613



AAVCBr-E4
1614



AAVCBr-E5
1615



AAVCBr-e5
1616



AAVCBr-E6
1617



AAVCBr-E7
1618



AAVCBr-E8
1619



AAVCLv-D1
1620



AAVCLv-D2
1621



AAVCLv-D3
1622



AAVCLv-D4
1623



AAVCLv-D5
1624



AAVCLv-D6
1625



AAVCLv-D7
1626



AAVCLv-D8
1627



AAVCLv-E1
1628



AAVCLv-R1
1629



AAVCLv-R2
1630



AAVCLv-R3
1631



AAVCLv-R4
1632



AAVCLv-R5
1633



AAVCLv-R6
1634



AAVCLv-R7
1635



AAVCLv-R8
1636



AAVCLv-R9
1637



AAVCLg-F1
1638



AAVCLg-F2
1639



AAVCLg-F3
1640



AAVCLg-F4
1641



AAVCLg-F5
1642



AAVCLg-F6
1643



AAVCLg-F7
1644



AAVCLg-F8
1645



AAVCSp-1
1646



AAVCSp-10
1647



AAVCSp-11
1648



AAVCSp-2
1649



AAVCSp-3
1650



AAVCSp-4
1651



AAVCSp-6
1652



AAVCSp-7
1653



AAVCSp-8
1654



AAVCSp-9
1655



AAVCHt-2
1656



AAVCHt-3
1657



AAVCKd-1
1658



AAVCKd-10
1659



AAVCKd-2
1660



AAVCKd-3
1661



AAVCKd-4
1662



AAVCKd-6
1663



AAVCKd-7
1664



AAVCKd-8
1665



AAVCLv-1
1666



AAVCLv-12
1667



AAVCLv-13
1668



AAVCLv-2
1669



AAVCLv-3
1670



AAVCLv-4
1671



AAVCLv-6
1672



AAVCLv-8
1673



AAVCKd-B1
1674



AAVCKd-B2
1675



AAVCKd-B3
1676



AAVCKd-B4
1677



AAVCKd-B5
1678



AAVCKd-B6
1679



AAVCKd-B7
1680



AAVCKd-B8
1681



AAVCKd-H1
1682



AAVCKd-H2
1683



AAVCKd-H3
1684



AAVCKd-H4
1685



AAVCKd-H5
1686



AAVCKd-H6
1687



AAV CHt-1
1688



AAVCLv1-1
1689



AAVCLv1-2
1690



AAVCLv1-3
1691



AAVCLv1-4
1692



AAVC1v1-7
1693



AAVC1v1-8
1694



AAVC1v1-9
1695



AAVC1v1-10
1696



AAV.VR-355
1697



AAV.hu.48R3
1698



AAVCBr-E1
1699



AAVCBr-E2
1700



AAVCBr-E3
1701



AAVCBr-E4
1702



AAVCBr-E5
1703



AAVCBr-e5
1704



AAVCBr-E6
1705



AAVCBr-E7
1706



AAVCBr-E8
1707



AAVCLv-D1
1708



AAVCLv-D2
1709



AAVCLv-D3
1710



AAVCLv-D4
1711



AAVCLv-D5
1712



AAVCLv-D6
1713



AAVCLv-D7
1714



AAVCLv-D8
1715



AAVCLv-E1
1716



AAVCLv-R1
1717



AAVCLv-R2
1718



AAVCLv-R3
1719



AAVCLv-R4
1720



AAVCLv-R5
1721



AAVCLv-R6
1722



AAVCLv-R7
1723



AAVCLv-R8
1724



AAVCLv-R9
1725



AAVCLg-F1
1726



AAVCLg-F2
1727



AAVCLg-F3
1728



AAVCLg-F4
1729



AAVCLg-F5
1730



AAVCLg-F6
1731



AAVCLg-F7
1732



AAVCLg-F8
1733



AAVCSp-1
1734



AAVCSp-10
1735



AAVCSp-11
1736



AAVCSp-2
1737



AAVCSp-3
1738



AAVCSp-4
1739



AAVCSp-6
1740



AAVCSp-7
1741



AAVCSp-8
1742



AAVCSp-9
1743



AAVCHt-2
1744



AAVCHt-3
1745



AAVCKd-1
1746



AAVCKd-10
1747



AAVCKd-2
1748



AAVCKd-3
1749



AAVCKd-4
1750



AAVCKd-6
1751



AAVCKd-7
1752



AAVCKd-8
1753



AAVCLv-1
1754



AAVCLv-12
1755



AAVCLv-13
1756



AAVCLv-2
1757



AAVCLv-3
1758



AAVCLv-4
1759



AAVCLv-6
1760



AAVCLv-8
1761



AAVCKd-B1
1762



AAVCKd-B2
1763



AAVCKd-B3
1764



AAVCKd-B4
1765



AAVCKd-B5
1766



AAVCKd-B6
1767



AAVCKd-B7
1768



AAVCKd-B8
1769



AAVCKd-H1
1770



AAVCKd-H2
1771



AAVCKd-H3
1772



AAVCKd-H4
1773



AAVCKd-H5
1774



AAVCKd-H6
1775



AAVCHt-1
1776



AAVCHt-P2
1777



AAVCHt-P5
1778



AAVCHt-P9
1779



AAVCBr-7.1
1780



AAVCBr-7.2
1781



AAVCBr-7.3
1782



AAVCBr-7.4
1783



AAVCBr-7.5
1784



AAVCBr-7.7
1785



AAVCBr-7.8
1786



AAV CBr-7.10
1787



AAVCKd-N3
1788



AAVCKd-N4
1789



AAVCKd-N9
1790



AAVCLv-L4
1791



AAVCLv-L5
1792



AAVCLv-L6
1793



AAVCLv-K1
1794



AAVCLv-K3
1795



AAVCLv-K6
1796



AAVCLv-M1
1797



AAVCLv-M11
1798



AAVCLv-M2
1799



AAVCLv-M5
1800



AAVCLv-M6
1801



AAVCLv-M7
1802



AAVCLv-M8
1803



AAVCLv-M9
1804



AAVCHt-P1
1805



AAVCHt-P6
1806



AAVCHt-P8
1807



AAVCHt-6.1
1808



AAV CHt-6.10
1809



AAVCHt-6.5
1810



AAVCHt-6.6
1811



AAVCHt-6.7
1812



AAVCHt-6.8
1813



AAVCSp-8.10
1814



AAVCSp-8.2
1815



AAVCSp-8.4
1816



AAVCSp-8.5
1817



AAVCSp-8.6
1818



AAVCSp-8.7
1819



AAVCSp-8.8
1820



AAVCSp-8.9
1821



AAVCBr-B7.3
1822



AAVCBr-B7.4
1823



AAV3B
1824



AAV4
1825



AAV5
1826



AAVCHt-P2
1827



AAVCHt-P5
1828



AAVCHt-P9
1829



AAVCBr-7.1
1830



AAVCBr-7.2
1831



AAVCBr-7.3
1832



AAVCBr-7.4
1833



AAVCBr-7.5
1834



AAVCBr-7.7
1835



AAVCBr-7.8
1836



AAV CBr-7.10
1837



AAVCKd-N3
1838



AAVCKd-N4
1839



AAVCKd-N9
1840



AAV CLv-L4
1841



AAVCLv-L5
1842



AAVCLv-L6
1843



AAVCLv-K1
1844



AAVCLv-K3
1845



AAV CLv-K6
1846



AAVCLv-M1
1847



AAVCLv-M11
1848



AAVCLv-M2
1849



AAVCLv-M5
1850



AAVCLv-M6
1851



AAVCLv-M7
1852



AAVCLv-M8
1853



AAVCLv-M9
1854



AAVCHt-P1
1855



AAVCHt-P6
1856



AAVCHt-P8
1857



AAVCHt-6.1
1858



AAV CHt-6.10
1859



AAVCHt-6.5
1860



AAVCHt-6.6
1861



AAVCHt-6.7
1862



AAVCHt-6.8
1863



AAVCSp-8.10
1864



AAVCSp-8.2
1865



AAVCSp-8.4
1866



AAVCSp-8.5
1867



AAVCSp-8.6
1868



AAV CSp-8.7
1869



AAVCSp-8.8
1870



AAVCSp-8.9
1871



AAV CBr-B7.3
1872



AAV CBr-B7.4
1873



AAV3B
1874



AAV4
1875



AAV5
1876



GPV
1877



B19
1878



MVM
1879



FPV
1880



CPV
1881



AAV6
1882



AAV6
1883



AAV2
1884



ShH1O
1885



ShH13
1886



ShH1O
1887



ShH1O
1888



ShH1O
1889



ShH1O
1890



ShH1O
1891



rh74
1892



rh74
1893



AAV8
1894



rh74
1895



rh74 (RHM4-1)
1896



rh74 (RHM15-1)
1897



rh74 (RHM15-2)
1898



rh74 (RHM15-3/RHM15-5)
1899



rh74 (RHM15-4)
1900



rh74 (RHM15-6)
1901



rh74 (RHM4-1)
1902



rh74 (RHM15-1)
1903



rh74 (RHM15-2)
1904



rh74 (RHM15-3/RHM15-5)
1905



rh74 (RHM15-4)
1906



rh74 (RHM15-6)
1907



AAV2 (comprising lung
1908



specific polypeptide)




AAV2 (comprising lung
1909



specific polypeptide)




Anc80
1910



Anc80
1911



Anc81
1912



Anc80
1913



Anc82
1914



Anc82
1915



Anc83
1916



Anc83
1917



Anc84
1918



Anc84
1919



Anc94
1920



Anc94
1921



Anc113
1922



Anc113
1923



Anc126
1924



Anc126
1925



Anc127
1926



Anc127
1927



Anc80L27
1928



Anc80L59
1929



Anc80L60
1930



Anc80L62
1931



Anc80L65
1932



Anc80L33
1933



Anc80L36
1934



Anc80L44
1935



Anc80L1
1936



Anc80L1
1937



AAV-X1
1938



AAV-X1b
1939



AAV-X5
1940



AAV-X19
1941



AAV-X21
1942



AAV-X22
1943



AAV-X23
1944



AAV-X24
1945



AAV-X25
1946



AAV-X26
1947



AAV-X1
1948



AAV-X1b
1949



AAV-X5
1950



AAV-X19
1951



AAV-X21
1952



AAV-X22
1953



AAV-X23
1954



AAV-X24
1955



AAV-X25
1956



AAV-X26
1957



AAVrh8
1958



AAVrh8VP2FC5
1959



AAVrh8VP2FC44
1960



AAVrh8VP2ApoB100
1961



AAVrh8VP2RVG
1962



AAVrh8VP2Angiopep-2 VP2
1963



AAV9.47VP1.3
1964



AAV9.47VP2ICAMg3
1965



AAV9.47VP2RVG
1966



AAV9.47VP2Angiopep-2
1967



AAV9.47VP2A-string
1968



AAVrh8VP2FC5 VP2
1969



AAVrh8VP2FC44 VP2
1970



AAVrh8VP2ApoB100 VP2
1971



AAVrh8VP2RVG VP2
1972



AAVrh8VP2Angiopep-2 VP2
1973



AAV9.47VP2ICAMg3 VP2
1974



AAV9.47VP2RVG VP2
1975



AAV9.47VP2Angiopep-2 VP2
1976



AAV9.47VP2A-string VP2
1977



rAAV-B1
1978



rAAV-B2
1979



rAAV-B3
1980



rAAV-B4
1981



rAAV-B1
1982



rAAV-B2
1983



rAAV-B3
1984



rAAV-B4
1985



rAAV-L1
1986



rAAV-L2
1987



rAAV-L3
1988



rAAV-L4
1989



rAAV-L1
1990



rAAV-L2
1991



rAAV-L3
1992



rAAV-L4
1993



AAV9
1994



rAAV
1995



rAAV
1996



rAAV
1997



rAAV
1998



rAAV
1999



rAAV
2000



rAAV
2001



rAAV
2002



rAAV
2003



rAAV
2004



rAAV
2005



rAAV
2006



rAAV
2007



rAAV
2008



rAAV
2009



rAAV
2010



rAAV
2011



rAAV
2012



rAAV
2013



rAAV
2014



rAAV
2015



rAAV
2016



rAAV
2017



rAAV
2018



rAAV
2019



rAAV
2020



rAAV
2021



rAAV
2022



rAAV
2023



rAAV
2024



rAAV
2025



rAAV
2026



rAAV
2027



rAAV
2028



rAAV
2029



rAAV
2030



rAAV
2031



rAAV
2032



rAAV
2033



rAAV
2034



rAAV
2035



rAAV
2036



rAAV
2037



rAAV
2038



rAAV
2039



rAAV
2040



rAAV
2041



rAAV
2042



rAAV
2043



rAAV
2044



rAAV
2045



rAAV
2046



rAAV
2047



rAAV
2048



rAAV
2049



rAAV
2050



rAAV
2051



rAAV
2052



rAAV
2053



rAAV
2054



rAAV
2055



rAAV
2056



rAAV
2057



rAAV
2058



rAAV
2059



rAAV
2060



rAAV
2061



rAAV
2062



rAAV
2063



rAAV
2064



rAAV
2065



rAAV
2066



rAAV
2067



rAAV
2068



rAAV
2069



rAAV
2070



rAAV
2071



rAAV
2072



rAAV
2073



rAAV
2074



rAAV
2075



rAAV
2076



rAAV
2077



rAAV
2078



rAAV
2079



rAAV
2080



rAAV
2081



rAAV
2082



rAAV
2083



rAAV
2084



rAAV
2085



rAAV
2086



rAAV
2087



rAAV
2088



rAAV
2089



rAAV
2090



rAAV
2091



rAAV
2092



rAAV
2093



rAAV
2094



rAAV
2095



rAAV
2096



rAAV
2097



rAAV
2098



rAAV
2099



rAAV
2100



rAAV
2101



rAAV
2102



rAAV
2103



rAAV
2104



rAAV
2105



rAAV
2106



rAAV
2107



rAAV
2108



rAAV
2109



rAAV
2110



rAAV
2111



rAAV
2112



rAAV
2113



rAAV
2114



rAAV
2115



rAAV
2116



rAAV
2117



rAAV
2118



rAAV
2119



rAAV
2120



rAAV
2121



rAAV
2122



AAV8E532K
2123



AAV8E532K
2124



rAAV4
2125



rAAV4
2126



rAAV4
2127



rAAV4
2128



rAAV4
2129



rAAV4
2130



rAAV4
2131



rAAV4
2132



rAAV4
2133



rAAV4
2134



rAAV4
2135



rAAV4
2136



rAAV4
2137



rAAV4
2138



rAAV4
2139



rAAV4
2140



rAAV4
2141



rAAV4
2142



rAAV4
2143



rAAV4
2144



AAV11
2145



AAV12
2146



rh32
2147



Th33
2148



Th34
2149



rAAV4
2150



rAAV4
2151



rAAV4
2152



rAAV4
2153



rAAV4
2154



rAAV4
2155



AAV2/8
2156



AAV2/8
2157



ancestral AAV
2158



ancestral AAV variant C4
2159



ancestral AAV variant C7
2160



ancestral AAV variant G4
2161



consensus amino acid
2162



sequence of ancestral AAV




variants, C4, C7 and G4




consensus amino acid
2163



sequence of ancestral AAV




variants, C4 and C7




AAVS (with an AAV2
2164



phospholipase domain)




AAVVR-942n
2165



AAVS-A (M569V)
2166



AAVS-A (M569V)
2167



AAVS-A (Y585V)
2168



AAVS-A (Y585V)
2169



AAVS-A (L587T)
2170



AAVS-A (L587T)
2171



AAVS-A (Y585V/L587T)
2172



AAVS-A (Y585V/L587T)
2173



AAV5-B (D652A)
2174



AAV5-B (D652A)
2175



AAV5-B (T362M)
2176



AAV5-B (T362M)
2177



AAV5-B (Q359D)
2178



AAV5-B (Q359D)
2179



AAV5-B (E350Q)
2180



AAV5-B (E350Q)
2181



AAV5-B (P533S)
2182



AAV5-B (P533S)
2183



AAV5-B (P533G)
2184



AAV5-B (P533G)
2185



AAVS-mutation in loop V11
2186



AAVS-mutation in loop V11
2187



AAVS
2188



Mut A (LK03/AAVS)
2189



Mut B (LK03/AAVS)
2190



Mut C (AAV8/AAV3B)
2191



MutD (AAV5/AAV3B)
2192



Mut E (AAV8/AAV3B)
2193



Mut F (AAV3B/AAV8)
2194



AAV44.9
2195



AAV44.9
2196



AAVrh8
2197



AAV44.9 (S470N)
2198



Th74 VP1
2199



AAV-LK03 (L125I)
2200



AAV3B (S663V + T492V)
2201



Anc80
2202



Anc80
2203



Anc81
2204



Anc81
2205



Anc82
2206



Anc82
2207



Anc83
2208



Anc83
2209



Anc84
2210



Anc84
2211



Anc94
2212



Anc94
2213



Anc113
2214



Anc113
2215



Anc126
2216



Anc126
2217



Anc127
2218



Anc127
2219



Anc80L27
2220



Anc80L59
2221



Anc80L60
2222



Anc80L62
2223



Anc80L65
2224



Anc80L33
2225



Anc80L36
2226



Anc80L44
2227



Anc80L1
2228



Anc80L1
2229



AAVrh1O
2230



Anc11O
2231



Anc11O
2232



AAVrh32.33
2233



AAVrh74
2234



AAV2
2235



AAV2
2236



AAV2
2237



Pallo-like vims
2238



Pallo-like vims
2239



Pallo-like vims
2240



Pallo-like vims
2241



Pallo-like vims
2242



Pallo-like vims
2243



AAVrh.10
2244



AAVrh.10
2245



AAV2tYF
2246



AAV-SPK
2247



AAV2.5
2248



AAV1.1
2249



AAV6.1
2250



AAV6.3.1
2251



AAV2i8
2252



AAV2i8
2253



ttAAV
2254



ttAAV-S312N
2255



ttAAV-S312N
2256



AAV6 (Y705, Y731, and T492)
2257



AAV2
2258



AAV2
2259










The single-stranded DNA genome of wild-type AAV is about 4.7 kilobases (kb). In practice, AAV genomes of up to about 5.0 kb appear to be completely packaged, i.e., be full-length, into AAV virus particles. With two AAV inverted terminal repeats (ITRs) of about 145 bases, the DNA packaging capacity of an AAV vector is such that a maximum of about 4.4 kb of protein.


The wild-type AAV genome comprises two open reading frames, Rep and Cap, flanked by two inverted terminal repeats (ITRs). Typically, when producing a recombinant AAV, the sequence between the two ITRs is replaced with one or more sequence of interest (e.g., a transgene), and the Rep and Cap sequences are provided in trans. The recombinant AAV genome construct, comprising two ITRs flanking a sequence of interest (such as a transgene), is referred to herein as an AAV expression cassette. The disclosure provides AAV expression cassettes for production of AAV viral vectors.


In some embodiments, an AAV expression cassette comprises a nucleic acid of the disclosure, i.e., a nucleic acid comprising a sequence encoding a single guide RNA (sgRNA) comprising a spacer sequence and a scaffold sequence wherein the spacer sequence targets an exon sequence of the DMD gene, such as a sequence of exon 43, 44, 45, 50, or 53 of the DMD gene.


In some embodiments, an AAV expression cassette comprises a first ITR, a transgene sequence, and a second ITR. In some embodiments, an AAV expression cassette comprises a first ITR, an expression control sequence (such as a promoter or enhancer), a transgene sequence, and a second ITR. In some embodiments, an AAV expression cassette comprises a first ITR, an expression control sequence (such as a promoter or enhancer), a transgene sequence, a stuffer sequence, and a second ITR. The transgene may comprise all or part of a nucleic acid of the disclosure. For example, the transgene may comprise a gRNA sequence (i.e., spacer+scaffold sequences), wherein the gRNA targets an exon sequence of the DMD gene, such as a sequence of exon 43, 44, 45, 50, or 53 of the DMD gene.


In some embodiments, an AAV expression cassette comprises a first ITR, a gRNA sequence, and a second ITR. In some embodiments, an AAV expression cassette comprises a first ITR, an expression control sequence (such as a promoter or enhancer), a gRNA sequence, and a second ITR. In some embodiments, an AAV expression cassette comprises a first ITR, an expression control sequence (such as a promoter or enhancer), a gRNA sequence, a stuffer sequence, and a second ITR.


In some embodiments, the transgene comprises more than one guide RNA sequence, such as two, three, four, five, six, seven, or eight gRNA sequences. In some embodiments, the transgene comprises three, four or five gRNA sequences. In some embodiments, each gRNA sequence is operably linked to an expression control sequence (such as a promoter or enhancer).


In some embodiments, an AAV expression cassette comprises a first ITR, a first expression control sequence (such as a promoter or enhancer), a first gRNA sequence, a second expression control sequence (such as a promoter or enhancer), a second gRNA sequence, and a second ITR.


In some embodiments, an AAV expression cassette comprises a first ITR, a first expression control sequence (such as a promoter or enhancer), a first gRNA sequence, a second expression control sequence (such as a promoter or enhancer), a second gRNA sequence, a third expression control sequence (such as a promoter or enhancer), a third gRNA sequence, and a second ITR.


In some embodiments, an AAV expression cassette comprises a first ITR, a first expression control sequence (such as a promoter or enhancer), a first gRNA sequence, a second expression control sequence (such as a promoter or enhancer), a second gRNA sequence, a third expression control sequence (such as a promoter or enhancer), a third gRNA sequence, a fourth expression control sequence (such as a promoter or enhancer), a fourth gRNA sequence, and a second ITR.


In some embodiments, an AAV expression cassette comprises a first ITR, a first expression control sequence (such as a promoter or enhancer), a first gRNA sequence, a second expression control sequence (such as a promoter or enhancer), a second gRNA sequence, a third expression control sequence (such as a promoter or enhancer), a third gRNA sequence, a fourth expression control sequence (such as a promoter or enhancer), a fourth gRNA sequence, a fifth expression control sequence (such as a promoter or enhancer), a fifth gRNA sequence, and a second ITR.


In some embodiments, all of the gRNA sequences are the same. In some embodiments, two or more of the gRNA sequences are different. In some embodiments, all of the gRNA sequences are different. In some embodiments, the AAV expression cassette further comprises a stuffer sequence. In some embodiments, the AAV expression cassette further comprises a polyadenosine (polyA) sequence.


In some embodiments, an AAV expression cassette comprises sequences encoding a first ITR, a first promoter, a first gRNA comprising a first spacer sequence, a second promoter, a second gRNA comprising a second spacer sequence, a third promoter, a third gRNA comprising a third spacer sequence; and a second ITR. At least one of the first, second, and third spacer sequences may target a sequence of the DMD gene (e.g., exon 43, exon 44, exon 46, exon 50 or exon 53 of the DMD gene). In some embodiments, the first, second, and third spacer sequences are each individually selected from any one of the gRNA spacer sequences in Table 2, or a sequence at least 95% identical thereto. In some embodiments, at least two of the first, second, and third spacer sequences are different. In some embodiments, the first, second, and third spacer sequences are the same. In some embodiments, the first, second, and/or third spacer sequences have a sequence that is at least 95% identical or 100% identical to the sequence of any one of SEQ ID NOs: 135-146, 170-260, 337-339, or 617. In some embodiments, the first, second, and/or third spacer sequences have a sequence that is at least 95% identical or 100% identical to the sequence of any one of SEQ ID NOs: 135-146, 170-260, 337-339, or 617.


In some embodiments, an AAV expression cassette comprises a first gRNA comprising a first spacer sequence, a second gRNA comprising a second spacer sequence, a third gRNA comprising a third spacer sequence, and a fourth gRNA comprising a fourth spacer sequence. In some embodiments, two, three, or four of the gRNAs are the same. In some embodiments, two, three, or four of the gRNAs are different. In some embodiments, an AAV expression cassette comprises a first promoter, a first gRNA comprising a first spacer sequence, a second promoter, a second gRNA comprising a second spacer sequence, a third promoter, a third gRNA comprising a third spacer sequence, a fourth promoter, and a fourth gRNA comprising a fourth spacer sequence. In some embodiments, an AAV expression cassette comprises a first ITR, a first promoter, a first gRNA comprising a first spacer sequence, a second promoter, a second gRNA comprising a second spacer sequence, a third promoter, a third gRNA comprising a third spacer sequence, a fourth promoter, a fourth gRNA comprising a fourth spacer sequence, and a second ITR. In some embodiments, the expression cassette further comprises a stuffer sequence.


In some embodiments, an AAV expression cassette comprises a first gRNA comprising a first spacer sequence, a second gRNA comprising a second spacer sequence, a third gRNA comprising a third spacer sequence, a fourth gRNA comprising a fourth spacer sequence, and a fifth gRNA comprising a fifth spacer sequence. In some embodiments, two, three, four, or five of the gRNAs are the same. In some embodiments, two, three, four or five of the gRNAs are different. In some embodiments, an AAV expression cassette comprises a first promoter, a first gRNA comprising a first spacer sequence, a second promoter, a second gRNA comprising a second spacer sequence, a third promoter, a third gRNA comprising a third spacer sequence, a fourth promoter, a fourth gRNA comprising a fourth spacer sequence, a fifth promoter, and a fifth gRNA comprising a fifth spacer sequence. In some embodiments, an AAV expression cassette comprises a first ITR, a first promoter, a first gRNA comprising a first spacer sequence, a second promoter, a second gRNA comprising a second spacer sequence, a third promoter, a third gRNA comprising a third spacer sequence, a fourth promoter, a fourth gRNA comprising a fourth spacer sequence, a fifth promoter, a fifth gRNA comprising a fifth spacer sequence, and a second ITR. In some embodiments, the expression cassette further comprises a stuffer sequence.


In some embodiments, an AAV expression cassette comprises sequences encoding a first inverted terminal repeat (ITR), a first promoter, a first gRNA comprising a first spacer sequence (e.g., a sequence at least 95% or 100% identical to any one of SEQ ID NOs: 135-146, 170-260, 337-339, or 617) and a scaffold sequence (e.g., a scaffold sequence at least 95% or 100% identical to any of SEQ ID NO: 147 to 153); and a second ITR.


In some embodiments, an AAV expression cassette comprises sequences encoding a first inverted terminal repeat (ITR), a first promoter, a first gRNA comprising a first spacer sequence (e.g., a sequence at least 95% or 100% identical to any one of SEQ ID NOs: 135-146, 170-260, 337-339, or 617) and a scaffold sequence (e.g., a scaffold sequence at least 95% or 100% identical to any of SEQ ID NO: 147 to 153), a second promoter, a second gRNA comprising a second spacer sequence (e.g., a sequence at least 95% or 100% identical to any one of SEQ ID NOs: 135-146, 170-260, 337-339, or 617) and a scaffold sequence (e.g., a scaffold sequence at least 95% or 100% identical to any of SEQ ID NO: 147 to 153); and a second ITR.


In some embodiments, an AAV expression cassette comprises sequences encoding a first inverted terminal repeat (ITR), a first promoter, a first gRNA comprising a first spacer sequence (e.g., a sequence at least 95% or 100% identical to any one of SEQ ID NOs: 135-146, 170-260, 337-339, or 617) and a scaffold sequence (e.g., a scaffold sequence at least 95% or 100% identical to any of SEQ ID NO: 147 to 153), a second promoter, a second gRNA comprising a second spacer sequence (e.g., a sequence at least 95% or 100% identical to any one of SEQ ID NOs: 135-146, 170-260, 337-339, or 617) and a scaffold sequence (e.g., a scaffold sequence at least 95% or 100% identical to any of SEQ ID NO: 147 to 153), a third promoter, a third gRNA comprising a third spacer sequence (e.g., a sequence at least 95% or 100% identical to any one of SEQ ID NOs: 135-146, 170-260, 337-339, or 617) and a scaffold sequence (e.g., a scaffold sequence at least 95% or 100% identical to any of SEQ ID NO: 147 to 153); and a second ITR.


In some embodiments, an AAV expression cassette comprises a first inverted terminal repeat (ITR), a first promoter, a nucleic acid comprising a gRNA targeting a sequence of the DMD gene, such as a sequence of Exon 43, 44, 45, 50, or 53 of the DMD gene, and a second ITR. In some embodiments, the AAV expression cassette further comprises a polyadenosine (polyA) sequence.


In some embodiments, one or both of the first ITR and the second ITR are isolated or derived from any one of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVRh74, AAV2i8, AAVRh10, AAV39, AAV43, AAVRh8, avian AAV, bovine AAV, canine AAV, equine AAV, and ovine AAV. In some embodiments, the expression cassette comprises multiple copies of the gRNA, such as 2, 3, 4, or 5 copies of the gRNA.


In some embodiments, an AAV expression cassette comprises a sequence to make the AAV vector less immunogenic (e.g., a “cloaking” sequence). In some embodiments, the sequence is isolated or derived from a telomere sequence. In some embodiments, the nucleotide sequence binds to a toll-like receptor, such as TLR9.


In some embodiments, an AAV expression cassette comprises sequences encoding a first ITR, a first promoter, a first gRNA comprising a first spacer sequence, a second promoter, a second gRNA comprising the first spacer sequence, a third promoter, a third gRNA comprising the first spacer sequence, and a second ITR.


In some embodiments, an AAV expression cassette comprises sequences encoding a first ITR, a first promoter, a first gRNA comprising a first spacer sequence, a second promoter, a second gRNA comprising the first spacer sequence, a third promoter, a third gRNA comprising the first spacer sequence, (optionally) a first stuffer sequence, and a second ITR. The first spacer sequence may target the DMD gene, for example it may target exon 43, exon 44, exon 46, exon 50 or exon 53 of the DMD gene. In some embodiments, the first spacer sequence is selected from any one of the gRNA sequences in Table 2, or a sequence at least 95% identical thereto.


The AAV expression cassettes described herein may be incorporated into an AAV vector. In some embodiments, an AAV vector comprises an AAV expression cassette encapsidated by an AAV capsid protein.


In some embodiments, the AAV vector is based on one or more of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVRh74, AAV2i8, AAVRh10, AAV39, AAV43, AAVRh8, avian AAV, bovine AAV, canine AAV, equine AAV, or ovine AAV. In some embodiments, the AAV vector is based on a modified AAV, comprising one or more non-naturally occurring sequences. In some embodiments, the AAV vector is based on a chimeric AAV. The AAV vector may be replication-defective or conditionally replication defective.


Adenoviruses. “Adenovirus expression vector” is meant to include those constructs containing adenovirus sequences sufficient to (a) support packaging of the construct and (b) to express an antisense polynucleotide that has been cloned therein. In this context, expression does not require that the gene product be synthesized.


The expression vector comprises a genetically engineered form of adenovirus. Knowledge of the genetic organization of adenovirus, a 36 kB, linear, double-stranded DNA virus, allows substitution of large pieces of adenoviral DNA with foreign sequences up to 7 kB. In contrast to retrovirus, the adenoviral infection of host cells does not result in chromosomal integration because adenoviral DNA can replicate in an episomal manner without potential genotoxicity. Also, adenoviruses are structurally stable, and no genome rearrangement has been detected after extensive amplification. Adenovirus can infect virtually all epithelial cells regardless of their cell cycle stage. So far, adenoviral infection appears to be linked only to mild disease such as acute respiratory disease in humans.


Adenovirus is particularly suitable for use as a gene transfer vector because of its mid-sized genome, ease of manipulation, high titer, wide target cell range and high infectivity. Both ends of the viral genome contain 100-200 base pair inverted repeats (ITRs), which are cis elements necessary for viral DNA replication and packaging. The early (E) and late (L) regions of the genome contain different transcription units that are divided by the onset of viral DNA replication. The E1 region (E1A and E1B) encodes proteins responsible for the regulation of transcription of the viral genome and a few cellular genes. The expression of the E2 region (E2A and E2B) results in the synthesis of the proteins for viral DNA replication. These proteins are involved in DNA replication, late gene expression and host cell shut-off. The products of the late genes, including the majority of the viral capsid proteins, are expressed only after significant processing of a single primary transcript issued by the major late promoter (MLP). The MLP, (located at 16.8 m.u.) is particularly efficient during the late phase of infection, and all the mRNAs issued from this promoter possess a 5′-tripartite leader (TPL) sequence which makes them preferred mRNA's for translation.


In one system, recombinant adenovirus is generated from homologous recombination between shuttle vector and provirus vector. Due to the possible recombination between two proviral vectors, wild-type adenovirus may be generated from this process. Therefore, it is critical to isolate a single clone of virus from an individual plaque and examine its genomic structure.


Generation and propagation of the current adenovirus vectors, which are replication deficient, depend on a unique helper cell line, designated 293, which was transformed from human embryonic kidney cells by Ad5 DNA fragments and constitutively expresses E1 proteins. Since the E3 region is dispensable from the adenovirus genome, the current adenovirus vectors, with the help of 293 cells, carry foreign DNA in either the E1, the D3 or both regions. In nature, adenovirus can package approximately 105% of the wild-type genome, providing capacity for about 2 extra kb of DNA. Combined with the approximately 5.5 kb of DNA that is replaceable in the E1 and E3 regions, the maximum capacity of the current adenovirus vector is under 7.5 kb, or about 15% of the total length of the vector. More than 80% of the adenovirus viral genome remains in the vector backbone and is the source of vector-borne cytotoxicity. Also, the replication deficiency of the E1-deleted virus is incomplete.


Helper cell lines may be derived from human cells such as human embryonic kidney cells, muscle cells, hematopoietic cells or other human embryonic mesenchymal or epithelial cells. Alternatively, the helper cells may be derived from the cells of other mammalian species that are permissive for human adenovirus. Such cells include, e.g., Vero cells or other monkey embryonic mesenchymal or epithelial cells. As stated above, the preferred helper cell line is 293.


The adenovirus may be of any of the 42 different known serotypes or subgroups A-F. Adenovirus type 5 of subgroup C is the preferred starting material in order to obtain the conditional replication-defective adenovirus vector for use as described herein. This is because Adenovirus type 5 is a human adenovirus about which a great deal of biochemical and genetic information is known, and it has historically been used for most constructions employing adenovirus as a vector.


As stated above, the typical vector according to the present disclosure is replication defective and will not have an adenovirus E1 region. Thus, it will be most convenient to introduce the polynucleotide encoding the gene of interest at the position from which the E1-coding sequences have been removed. However, the position of insertion of the construct within the adenovirus sequences is not critical. The polynucleotide encoding the gene of interest may also be inserted in lieu of the deleted E3 region in E3 replacement vectors, or in the E4 region where a helper cell line or helper virus complements the E4 defect.


Adenovirus is easy to grow and manipulate and exhibits broad host range in vitro and in vivo. This group of viruses can be obtained in high titers, e.g., 109-1012 plaque-forming units per ml, and they are highly infective. The life cycle of adenovirus does not require integration into the host cell genome. The foreign genes delivered by adenovirus vectors are episomal and, therefore, have low genotoxicity to host cells. No side effects have been reported in studies of vaccination with wild-type adenovirus, demonstrating their safety and therapeutic potential as in vivo gene transfer vectors.


Retroviruses. The retroviruses are a group of single-stranded RNA viruses characterized by an ability to convert their RNA to double-stranded DNA in infected cells by a process of reverse-transcription. The resulting DNA then stably integrates into cellular chromosomes as a provirus and directs synthesis of viral proteins. The integration results in the retention of the viral gene sequences in the recipient cell and its descendants. The retroviral genome contains three genes, gag, pol, and env that code for capsid proteins, polymerase enzyme, and envelope components, respectively. A sequence found upstream from the gag gene contains a signal for packaging of the genome into virions. Two long terminal repeat (LTR) sequences are present at the 5′ and 3′ ends of the viral genome.


In order to construct a retroviral vector, a nucleic acid encoding a gene of interest is inserted into the viral genome in the place of certain viral sequences to produce a virus that is replication-defective. In order to produce virions, a packaging cell line containing the gag, pol, and env genes but without the LTR and packaging components is constructed. When a recombinant plasmid containing a cDNA, together with the retroviral LTR and packaging sequences is introduced into this cell line (by calcium phosphate precipitation for example), the packaging sequence allows the RNA transcript of the recombinant plasmid to be packaged into viral particles, which are then secreted into the culture media. The media containing the recombinant retroviruses is then collected, optionally concentrated, and used for gene transfer. Retroviral vectors are able to infect a broad variety of cell types. However, integration and stable expression require the division of host cells.


A novel approach designed to allow specific targeting of retrovirus vectors was recently developed based on the chemical modification of a retrovirus by the chemical addition of lactose residues to the viral envelope. This modification could permit the specific infection of hepatocytes via sialoglycoprotein receptors.


A different approach to targeting of recombinant retroviruses was designed in which biotinylated antibodies against a retroviral envelope protein and against a specific cell receptor were used. The antibodies are coupled via the biotin components by using streptavidin. Using antibodies against major histocompatibility complex class I and class II antigens, a variety of human cells that bear those surface antigens may be infected with an ecotropic virus in vitro.


Other viral vectors. Other viral vectors may be employed as expression constructs. For example, vectors derived from viruses such as vaccinia virus and herpesviruses may be employed. They offer several attractive features for various mammalian cells.


Non-viral methods. Several non-viral methods for the transfer of expression constructs into cultured mammalian cells also are contemplated. These include calcium phosphate precipitation, DEAE-dextran, electroporation, direct microinjection, DNA-loaded liposomes and lipofectamine-DNA complexes, cell sonication, gene bombardment using high velocity microprojectiles, and receptor-mediated transfection. Some of these techniques may be successfully adapted for in vivo or ex vivo use.


Once the expression construct has been delivered into the cell the nucleic acid encoding the gene of interest may be positioned and expressed at different sites. In certain embodiments, the nucleic acid encoding the gene may be stably integrated into the genome of the cell. This integration may be in the cognate location and orientation via homologous recombination (gene replacement) or it may be integrated in a random, non-specific location (gene augmentation). In yet further embodiments, the nucleic acid may be stably maintained in the cell as a separate, episomal segment of DNA. Such nucleic acid segments or “episomes” encode sequences sufficient to permit maintenance and replication independent of or in synchronization with the host cell cycle. How the expression construct is delivered to a cell and where in the cell the nucleic acid remains is dependent on the type of expression construct employed.


In yet another embodiment, the expression construct may simply consist of naked recombinant DNA or plasmids. Transfer of the construct may be performed by any of the methods mentioned above which physically or chemically permeabilize the cell membrane. This is particularly applicable for transfer in vitro but it may be applied to in vivo use as well. Polyomavirus DNA has been successfully injected in the form of calcium phosphate precipitates into liver and spleen of adult and newborn mice demonstrating active viral replication and acute infection. Direct intraperitoneal injection of calcium phosphate-precipitated plasmids, resulting in expression of the transfected genes, may also be used. It is envisioned that DNA encoding a gene of interest may also be transferred in a similar manner in vivo and express the gene product.


In still another embodiment for transferring a naked DNA expression construct into cells may involve particle bombardment. This method depends on the ability to accelerate DNA-coated microprojectiles to a high velocity allowing them to pierce cell membranes and enter cells without killing them. Several devices for accelerating small particles have been developed. One such device relies on a high voltage discharge to generate an electrical current, which in turn provides the motive force. The microprojectiles used have consisted of biologically inert substances such as tungsten or gold beads.


Selected organs including the liver, skin, and muscle tissue of rats and mice have been bombarded in vivo. This may require surgical exposure of the tissue or cells, to eliminate any intervening tissue between the gun and the target organ, i.e., ex vivo treatment. Again, DNA encoding a particular gene may be delivered via this method and still be incorporated by the instant disclosure.


In a further embodiment, the expression construct may be entrapped in a liposome. Liposomes are vesicular structures characterized by a phospholipid bilayer membrane and an inner aqueous medium. Multilamellar liposomes have multiple lipid layers separated by aqueous medium. They form spontaneously when phospholipids are suspended in an excess of aqueous solution. The lipid components undergo self-rearrangement before the formation of closed structures and entrap water and dissolved solutes between the lipid bilayers. Also contemplated are lipofectamine-DNA complexes.


Liposome-mediated nucleic acid delivery and expression of foreign DNA in vitro has been very successful. A reagent known as Lipofectamine 2000™ is widely used and commercially available.


In certain embodiments, the liposome may be complexed with a hemagglutinating virus (HVJ). This has been shown to facilitate fusion with the cell membrane and promote cell entry of liposome-encapsulated DNA. In other embodiments, the liposome may be complexed or employed in conjunction with nuclear non-histone chromosomal proteins (HMG-1). In yet further embodiments, the liposome may be complexed or employed in conjunction with both HVJ and HMG-1. In that such expression constructs have been successfully employed in transfer and expression of nucleic acid in vitro and in vivo, then they are applicable. Where a bacterial promoter is employed in the DNA construct, it also will be desirable to include within the liposome an appropriate bacterial polymerase.


Other expression constructs which can be employed to deliver a nucleic acid encoding a particular gene into cells are receptor-mediated delivery vehicles. These take advantage of the selective uptake of macromolecules by receptor-mediated endocytosis in almost all eukaryotic cells. Because of the cell type-specific distribution of various receptors, the delivery can be highly specific.


Receptor-mediated gene targeting vehicles generally consist of two components: a cell receptor-specific ligand and a DNA-binding agent. Several ligands have been used for receptor-mediated gene transfer. The most extensively characterized ligands are asialoorosomucoid (ASOR) and transferrin. A synthetic neoglycoprotein, which recognizes the same receptor as ASOR, may be used as a gene delivery vehicle. In some embodiments, epidermal growth factor (EGF) may be used to deliver genes.


III. METHODS OF MAKING TRANSGENIC MICE

A particular embodiment provides transgenic animals that contain mutations in the dystrophin gene. Also, transgenic animals may express a marker that reflects the production of mutant or normal dystrophin gene product.


In a general aspect, a transgenic animal is produced by the integration of a given construct into the genome in a manner that permits the expression of the transgene using methods discussed above. Methods for producing transgenic animals are generally described by Wagner and Hoppe (U.S. Pat. No. 4,873,191; incorporated herein by reference), and Brinster et al. (1985; incorporated herein by reference).


Typically, the construct is transferred by microinjection into a fertilized egg. The microinjected eggs are implanted into a host female, and the progeny are screened for the expression of the transgene. Transgenic animals may be produced from the fertilized eggs from a number of animals including, but not limited to reptiles, amphibians, birds, mammals, and fish.


RNA for microinjection can be prepared by any means known in the art. For example, RNA for microinjection can be cleaved with enzymes appropriate for removing the bacterial plasmid sequences, and the RNA fragments electrophoresed on 1% agarose gels in TBE buffer, using standard techniques. The RNA bands are visualized by staining with ethidium bromide, and the band containing the expression sequences is excised. The excised band is then placed in dialysis bags containing 0.3 M sodium acetate, pH 7.0. RNA is electroeluted into the dialysis bags, extracted with a 1:1 phenol:chloroform solution and precipitated by two volumes of ethanol. The RNA is redissolved in 1 ml of low salt buffer (0.2 M NaCl, 20 mM Tris, pH 7.4, and 1 mM EDTA) and purified on an Elutip-D® column. The column is first primed with 3 ml of high salt buffer (1 M NaCl, 20 mM Tris, pH 7.4, and 1 mM EDTA) followed by washing with 5 ml of low salt buffer. The DNA solutions are passed through the column three times to bind RNA to the column matrix. After one wash with 3 ml of low salt buffer, the RNA is eluted with 0.4 ml high salt buffer and precipitated by two volumes of ethanol. RNA concentrations are measured by absorption at 260 nm in a UV spectrophotometer. For microinjection, DNA concentrations are adjusted to 3 μg/ml in 5 mM Tris, pH 7.4 and 0.1 mM EDTA.


In an exemplary microinjection procedure, female mice six weeks of age are induced to superovulate with a 5 IU injection (0.1 cc, ip) of pregnant mare serum gonadotropin (PMSG; Sigma) followed 48 hours later by a 5 IU injection (0.1 cc, ip) of human chorionic gonadotropin (hCG; Sigma). Females are placed with males immediately after hCG injection. Twenty-one hours after hCG injection, the mated females are sacrificed by CO2 asphyxiation or cervical dislocation and embryos are recovered from excised oviducts and placed in Dulbecco's phosphate buffered saline with 0.5% bovine serum albumin (BSA; Sigma). Surrounding cumulus cells are removed with hyaluronidase (1 mg/ml). Pronuclear embryos are then washed and placed in Earle's balanced salt solution containing 0.5% BSA (EBSS) in a 37.5° C. incubator with a humidified atmosphere at 5% CO2, 95% air until the time of injection. Embryos can be implanted at the two-cell stage.


Randomly cycling adult female mice are paired with vasectomized males. C57BL/6 or Swiss mice or other comparable strains can be used for this purpose. Recipient females are mated at the same time as donor females. At the time of embryo transfer, the recipient females are anesthetized with an intraperitoneal injection of 0.015 ml of 2.5% avertin per gram of body weight. The oviducts are exposed by a single midline dorsal incision. An incision is then made through the body wall directly over the oviduct. The ovarian bursa is then torn with watchmakers forceps. Embryos to be transferred are placed in DPBS (Dulbecco's phosphate buffered saline) and in the tip of a transfer pipet (about 10 to 12 embryos). The pipet tip is inserted into the infundibulum and the embryos transferred. After the transfer, the incision is closed by two sutures.


IV. PHARMACEUTICAL COMPOSITIONS AND DELIVERY METHODS

Where clinical applications are contemplated, pharmaceutical compositions will be prepared in a form appropriate for the intended application. Generally, this will entail preparing compositions that are essentially free of pyrogens, as well as other impurities that could be harmful to humans or animals.


One will generally desire to employ appropriate salts and buffers to render drugs, proteins or delivery vectors stable and allow for uptake by target cells. Aqueous compositions of the disclosure may comprise an effective amount of the drug, vector or proteins, dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium. The phrase “pharmaceutically or pharmacologically acceptable” refer to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or a human. As used herein, “pharmaceutically acceptable carrier” includes solvents, buffers, solutions, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like acceptable for use in formulating pharmaceuticals, such as pharmaceuticals suitable for administration to humans. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredients of the present disclosure, its use in therapeutic compositions is contemplated. Supplementary active ingredients also can be incorporated into the compositions, provided they do not inactivate the vectors or cells of the compositions.


The active compositions of the present disclosure may include classic pharmaceutical preparations. Administration of these compositions according to the present disclosure may be via any common route so long as the target tissue is available via that route, but generally including systemic administration. This includes oral, nasal, or buccal. Alternatively, administration may be by intradermal, subcutaneous, intramuscular, intraperitoneal or intravenous injection, or by direct injection into muscle tissue. Such compositions would normally be administered as pharmaceutically acceptable compositions, as described supra.


The active compounds may also be administered parenterally or intraperitoneally. By way of illustration, solutions of the active compounds as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations generally contain a preservative to prevent the growth of microorganisms.


The pharmaceutical forms suitable for injectable use include, for example, sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. Generally, these preparations are sterile and fluid to the extent that easy injectability exists. Preparations should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms, such as bacteria and fungi. Appropriate solvents or dispersion media may contain, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.


Sterile injectable solutions may be prepared by incorporating the active compounds in an appropriate amount into a solvent along with any other ingredients (for example as enumerated above) as desired, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the desired other ingredients, e.g., as enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation include vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient(s) plus any additional desired ingredient from a previously sterile-filtered solution thereof.


The compositions generally may be formulated in a neutral or salt form. Pharmaceutically-acceptable salts include, for example, acid addition salts (formed with the free amino groups of the protein) derived from inorganic acids (e.g., hydrochloric or phosphoric acids, or from organic acids (e.g., acetic, oxalic, tartaric, mandelic, and the like). Salts formed with the free carboxyl groups of the protein can also be derived from inorganic bases (e.g., sodium, potassium, ammonium, calcium, or ferric hydroxides) or from organic bases (e.g., isopropylamine, trimethylamine, histidine, procaine and the like.


Upon formulation, solutions are preferably administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations may easily be administered in a variety of dosage forms such as injectable solutions, drug release capsules and the like. For parenteral administration in an aqueous solution, for example, the solution generally is suitably buffered, and the liquid diluent first rendered isotonic for example with sufficient saline or glucose. Such aqueous solutions may be used, for example, for intravenous, intramuscular, subcutaneous and intraperitoneal administration. Preferably, sterile aqueous media are employed as is known to those of skill in the art, particularly in light of the present disclosure. By way of illustration, a single dose may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, “Remington's Pharmaceutical Sciences” 15th Edition, pages 1035-1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologics standards.


V. DMD SUBJECT CHARACTERISTICS AND CLINICAL PRESENTATION

Duchenne muscular dystrophy (DMD) is a recessive X-linked form of muscular dystrophy, affecting around 1 in 5000 boys, which results in muscle degeneration and premature death. The disorder is caused by a mutation in the gene dystrophin, located on the human X chromosome, which codes for the protein dystrophin. Dystrophin is an important component within muscle tissue that provides structural stability to the dystroglycan complex (DGC) of the cell membrane. While both sexes can carry the mutation, females are rarely affected with the skeletal muscle form of the disease.


Mutations vary in nature and frequency. Large genetic deletions are found in about 60-70% of cases, large duplications are found in about 10% of cases, and point mutants or other small changes account for about 15-30% of cases. An examination of some 7000 mutations catalogued a total of 5,682 large mutations (80% of total mutations), of which 4,894 (86%) were deletions (1 exon or larger) and 784 (14%) were duplications (1 exon or larger). There were 1,445 small mutations (smaller than 1 exon, 20% of all mutations), of which 358 (25%) were small deletions and 132 (9%) small insertions, while 199 (14%) affected the splice sites. Point mutations totaled 756 (52% of small mutations) with 726 (50%) nonsense mutations and 30 (2%) missense mutations. Finally, 22 (0.3%) mid-intronic mutations were observed. In addition, mutations were identified within the database that would potentially benefit from novel genetic therapies for DMD including stop codon read-through therapies (10% of total mutations) and exon skipping therapy (80% of deletions and 55% of total mutations).


A. Symptoms


Symptoms usually appear in boys between the ages of 2 and 3 and may be visible in early infancy. Even though symptoms do not appear until early infancy, laboratory testing can identify children who carry the active mutation at birth. Progressive proximal muscle weakness of the legs and pelvis associated with loss of muscle mass is observed first. Eventually this weakness spreads to the arms, neck, and other areas. Early signs may include pseudohypertrophy (enlargement of calf and deltoid muscles), low endurance, and difficulties in standing unaided or inability to ascend staircases. As the condition progresses, muscle tissue experiences wasting and is eventually replaced by fat and fibrotic tissue (fibrosis). By age 10, braces may be required to aid in walking but most patients are wheelchair dependent by age 12. Later symptoms may include abnormal bone development that lead to skeletal deformities, including curvature of the spine. Due to progressive deterioration of muscle, loss of movement occurs, eventually leading to paralysis. Intellectual impairment may or may not be present but if present, does not progressively worsen as the child ages. The average life expectancy for males afflicted with DMD is around 25.


The main symptom of Duchenne muscular dystrophy, a progressive neuromuscular disorder, is muscle weakness associated with muscle wasting with the voluntary muscles being first affected, especially those of the hips, pelvic area, thighs, shoulders, and calves. Muscle weakness also occurs later, in the arms, neck, and other areas. Calves are often enlarged. Symptoms usually appear before age 6 and may appear in early infancy. Other physical symptoms are:

    • Awkward manner of walking, stepping, or running—(patients tend to walk on their forefeet, because of an increased calf muscle tone. Also, toe walking is a compensatory adaptation to knee extensor weakness.)
    • Frequent falls
    • Fatigue
    • Difficulty with motor skills (running, hopping, jumping)
    • Lumbar hyperlordosis, possibly leading to shortening of the hip-flexor muscles. This has an effect on overall posture and a manner of walking, stepping, or running.
    • Muscle contractures of Achilles tendon and hamstrings impair functionality because the muscle fibers shorten and fibrose in connective tissue
    • Progressive difficulty walking
    • Muscle fiber deformities
    • Pseudohypertrophy (enlarging) of tongue and calf muscles. The muscle tissue is eventually replaced by fat and connective tissue, hence the term pseudohypertrophy.
    • Higher risk of neurobehavioral disorders (e.g., ADHD), learning disorders (dyslexia), and non-progressive weaknesses in specific cognitive skills (in particular short-term verbal memory), which are believed to be the result of absent or dysfunctional dystrophin in the brain.
    • Eventual loss of ability to walk (usually by the age of 12)
    • Skeletal deformities (including scoliosis in some cases)
    • Trouble getting up from lying or sitting position


      The condition can often be observed clinically from the moment the patient takes his first steps, and the ability to walk usually completely disintegrates between the time the boy is 9 to 12 years of age. Most men affected with DMD become essentially “paralyzed from the neck down” by the age of 21. Muscle wasting begins in the legs and pelvis, then progresses to the muscles of the shoulders and neck, followed by loss of arm muscles and respiratory muscles. Calf muscle enlargement (pseudohypertrophy) is quite obvious. Cardiomyopathy particularly (dilated cardiomyopathy) is common, but the development of congestive heart failure or arrhythmia (irregular heartbeat) is only occasional.


A positive Gowers' sign reflects the more severe impairment of the lower extremities muscles. The child helps himself to get up with upper extremities: first by rising to stand on his arms and knees, and then “walking” his hands up his legs to stand upright. Affected children usually tire more easily and have less overall strength than their peers. Creatine kinase (CPK-MM) levels in the bloodstream are extremely high. An electromyography (EMG) shows that weakness is caused by destruction of muscle tissue rather than by damage to nerves. Genetic testing can reveal genetic errors in the Xp21 gene. A muscle biopsy (immunohistochemistry or immunoblotting) or genetic test (blood test) confirms the absence of dystrophin, although improvements in genetic testing often make this unnecessary.


Additional symptoms may include:

    • Abnormal heart muscle (cardiomyopathy)
    • Congestive heart failure or irregular heart rhythm (arrhythmia)
    • Deformities of the chest and back (scoliosis)
    • Enlarged muscles of the calves, buttocks, and shoulders (around age 4 or 5). These muscles are eventually replaced by fat and connective tissue (pseudohypertrophy).
    • Loss of muscle mass (atrophy)
    • Muscle contractures in the heels, legs
    • Muscle deformities
    • Respiratory disorders, including pneumonia and swallowing with food or fluid passing into the lungs (in late stages of the disease)


B. Causes


Duchenne muscular dystrophy (DMD) is caused by a mutation of the dystrophin gene at locus Xp21, located on the short arm of the X chromosome. Dystrophin is responsible for connecting the cytoskeleton of each muscle fiber to the underlying basal lamina (extracellular matrix), through a protein complex containing many subunits. The absence of dystrophin permits excess calcium to penetrate the sarcolemma (the cell membrane). Alterations in calcium and signaling pathways cause water to enter into the mitochondria, which then burst.


In skeletal muscle dystrophy, mitochondrial dysfunction gives rise to an amplification of stress-induced cytosolic calcium signals and an amplification of stress-induced reactive-oxygen species (ROS) production. In a complex cascading process that involves several pathways and is not clearly understood, increased oxidative stress within the cell damages the sarcolemma and eventually results in the death of the cell. Muscle fibers undergo necrosis and are ultimately replaced with adipose and connective tissue.


DMD is inherited in an X-linked recessive pattern. Females will typically be carriers for the disease while males will be affected. Typically, a female carrier will be unaware they carry a mutation until they have an affected son. The son of a carrier mother has a 50% chance of inheriting the defective gene from his mother. The daughter of a carrier mother has a 50% chance of being a carrier and a 50% chance of having two normal copies of the gene. In all cases, an unaffected father will either pass a normal Y to his son or a normal X to his daughter. Female carriers of an X-linked recessive condition, such as DMD, can show symptoms depending on their pattern of X-inactivation.


Duchenne muscular dystrophy has an incidence of 1 in 5,000 male infants. Mutations within the dystrophin gene can either be inherited or occur spontaneously during germline transmission.


C. Diagnosis


Genetic counseling is advised for people with a family history of the disorder. Duchenne muscular dystrophy can be detected with about 95% accuracy by genetic studies.


DNA test. The muscle-specific isoform of the dystrophin gene is composed of 79 exons, and DNA testing and analysis can usually identify the specific type of mutation of the exon or exons that are affected. DNA testing confirms the diagnosis in most cases.


Muscle biopsy. If DNA testing fails to find the mutation, a muscle biopsy test may be performed. A small sample of muscle tissue is extracted (usually with a scalpel instead of a needle) and a dye is applied that reveals the presence of dystrophin. Complete absence of the protein indicates the condition.


Over the past several years DNA tests have been developed that detect more of the many mutations that cause the condition, and muscle biopsy is not required as often to confirm the presence of Duchenne's.


Prenatal tests. DMD is carried by an X-linked recessive gene. Males have only one X chromosome, so one copy of the mutated gene will cause DMD. Fathers cannot pass X-linked traits on to their sons, so the mutation is transmitted by the mother.


If the mother is a carrier, and therefore one of her two X chromosomes has a DMD mutation, there is a 50% chance that a female child will inherit that mutation as one of her two X chromosomes, and be a carrier. There is a 50% chance that a male child will inherit that mutation as his one X chromosome, and therefore have DMD.


Prenatal tests can tell whether their unborn child has the most common mutations. There are many mutations responsible for DMD, and some have not been identified, so genetic testing only works when family members with DMD have a mutation that has been identified.


Prior to invasive testing, determination of the fetal sex is important; while males are sometimes affected by this X-linked disease, female DMD is extremely rare. This can be achieved by ultrasound scan at 16 weeks or more recently by free fetal DNA testing. Chorion villus sampling (CVS) can be done at 11-14 weeks, and has a 1% risk of miscarriage. Amniocentesis can be done after 15 weeks, and has a 0.5% risk of miscarriage. Fetal blood sampling can be done at about 18 weeks. Another option in the case of unclear genetic test results is fetal muscle biopsy.


D. Treatment


There is no current cure for DMD, and an ongoing medical need has been recognized by regulatory authorities. Phase 1-2a trials with exon skipping treatment for certain mutations have halted decline and produced clinical improvements in walking. Sarepta's drug Exondys 51 (eteplirsen) has recently received FDA approval. However, treatment is generally aimed at controlling the onset of symptoms to maximize the quality of life, and include the following:

    • Corticosteroids such as prednisolone and deflazacort increase energy and strength and defer severity of some symptoms.
    • Randomized control trials have shown that beta-2-agonists increase muscle strength but do not modify disease progression. Follow-up time for most RCTs on beta2-agonists is only around 12 months and hence results cannot be extrapolated beyond that time frame.
    • Mild, non jarring physical activity such as swimming is encouraged. Inactivity (such as bed rest) can worsen the muscle disease.
    • Physical therapy is helpful to maintain muscle strength, flexibility, and function.
    • Orthopedic appliances (such as braces and wheelchairs) may improve mobility and the ability for self-care. Form-fitting removable leg braces that hold the ankle in place during sleep can defer the onset of contractures.
    • Appropriate respiratory support as the disease progresses is important.


      Comprehensive multi-disciplinary care standards/guidelines for DMD have been developed by the Centers for Disease Control and Prevention (CDC), and and are available at treat-nmd.eu/dmd/care/diagnosis-management-DMD.


1. Physical Therapy


Physical therapists are concerned with enabling patients to reach their maximum physical potential. Their aim is to:

    • minimize the development of contractures and deformity by developing a programme of stretches and exercises where appropriate
    • anticipate and minimize other secondary complications of a physical nature by recommending bracing and durable medical equipment
    • monitor respiratory function and advise on techniques to assist with breathing exercises and methods of clearing secretions


2. Respiration Assistance


Modern “volume ventilators/respirators,” which deliver an adjustable volume (amount) of air to the person with each breath, are valuable in the treatment of people with muscular dystrophy related respiratory problems. The ventilator may require an invasive endotracheal or tracheotomy tube through which air is directly delivered, but, for some people non-invasive delivery through a face mask or mouthpiece is sufficient. Positive airway pressure machines, particularly bi-level ones, are sometimes used in this latter way. The respiratory equipment may easily fit on a ventilator tray on the bottom or back of a power wheelchair with an external battery for portability.


Ventilator treatment may start in the mid to late teens when the respiratory muscles can begin to collapse. If the vital capacity has dropped below 40% of normal, a volume ventilator/respirator may be used during sleeping hours, a time when the person is most likely to be under ventilating (“hypoventilating”). Hypoventilation during sleep is determined by a thorough history of sleep disorder with an oximetry study and a capillary blood gas (See Pulmonary Function Testing). A cough assist device can help with excess mucus in lungs by hyperinflation of the lungs with positive air pressure, then negative pressure to get the mucus up. If the vital capacity continues to decline to less than 30 percent of normal, a volume ventilator/respirator may also be needed during the day for more assistance. The person gradually will increase the amount of time using the ventilator/respirator during the day as needed.


E. Prognosis


Duchenne muscular dystrophy is a progressive disease which eventually affects all voluntary muscles and involves the heart and breathing muscles in later stages. The life expectancy is currently estimated to be around 25, but this varies from patient to patient. Recent advancements in medicine are extending the lives of those afflicted. The Muscular Dystrophy Campaign, which is a leading UK charity focusing on all muscle disease, states that “with high standards of medical care young men with Duchenne muscular dystrophy are often living well into their 30s.”


In rare cases, persons with DMD have been seen to survive into the forties or early fifties, with the use of proper positioning in wheelchairs and beds, ventilator support (via tracheostomy or mouthpiece), airway clearance, and heart medications, if required. Early planning of the required supports for later-life care has shown greater longevity in people living with DMD.


Curiously, in the mdx mouse model of Duchenne muscular dystrophy, the lack of dystrophin is associated with increased calcium levels and skeletal muscle myonecrosis. The intrinsic laryngeal muscles (ILM) are protected and do not undergo myonecrosis. ILM have a calcium regulation system profile suggestive of a better ability to handle calcium changes in comparison to other muscles, and this may provide a mechanistic insight for their unique pathophysiological properties. The ILM may facilitate the development of novel strategies for the prevention and treatment of muscle wasting in a variety of clinical scenarios.


VI. SEQUENCE TABLES









TABLE 8







Sequence of primers for generating sgRNA targeting


human Dmd exon 44, exon 46, and exon 53









ID
Sequence (5′-3′)
SEQ ID NO.












hDMD-E43g1-top
CACCGTTTTAAAATTTTTATATTA
340





hDMD-E43g1-bottom
aaacTAATATAAAAATTTTAAAAC
341





hDMD-E43g2-top
CACCGTTTTTATATTACAGAATATAA
342





hDMD-E43g2-bottom
aaacTTATATTCTGTAATATAAAAA
343





hDMD-E43g3-top
CACCGATATTACAGAATATAAAAGA
344





hDMD-E43g3-bottom
aaacTCTTTTATATTCTGTAATAT
345





hDMD-E43g5-top
CACCGAAATGTACAAGGACCGACAA
346





hDMD-E43g5-bottom
aaacTTGTCGGTCCTTGTACATTTC
347





hDMD-E43g4-top
CACCGTATGTGTTACCTACCCTTGT
348





hDMD-E43g4-bottom
aaacACAAGGGTAGGTAACACATA
349





hDMD-E43g6-top
CACCGTACAAGGACCGACAAGGGT
350





hDMD-E43g6-bottom
aaacACCCTTGTCGGTCCTTGTAC
351





hDMD-E44g1-top
CACCGATCCATATGCTTTTACCTGC
352





hDMD-E44g1-bottom
aaacGCAGGTAAAAGCATATGGAT
353





hDMD-E44g2-top
CACCgatccatatgcttttACCTG
354





hDMD-E44g2-bottom
aaacCAGGTAAAAGCATATGGATC
355





hDMD-E44g3-top
CACCGCAGATCTGTCAAATCGCCTG
356





hDMD-E44g3-bottom
aaacCAGGCGATTTGACAGATCTG
357





hDMD-E44g4-top
CACCGTAAATACAAATGGTATCTTA
358





hDMD-E44g4-bottom
aaacTAAGATACCATTTGTATTTA
359





hDMD-E44g5-bottom
aaacGCAGGCGATTTGACAGATCTC
1





hDMD-E44g5-top
CACCGAGATCTGTCAAATCGCCTGC
2





hDMD-E44g6-top
CACCGACAGATCTGTTGAGAAATGG
3





hDMD-E44g6-bottom
aaacCCATTTCTCAACAGATCTGTC
4





hDMD-E44g7-bottom
aaacGAGAATTGGGAACATGCTAAC
5





hDMD-E44g7-top
CACCGTTAGCATGTTCCCAATTCTC
6





hDMD-E44g8-bottom
aaacTTTGTATTTAGCATGTTCCC
7





hDMD-E44g8-top
CACCGGGAACATGCTAAATACAAA
8





hDMD-E44g9-top
CACCGTTGACAGATCTGTTGAGAAA
9





hDMD-E44g9-bottom
aaacTTTCTCAACAGATCTGTCAAC
10





hDMD-E44g10-bottom
aaacATTCTCAGGAATTTGTGTCTC
11





hDMD-E44g10-top
CACCGAGACACAAATTCCTGAGAAT
12





hDMD-E44g11-bottom
aaacAATTCTCAGGAATTTGTGTC
13





hDMD-E44g11-top
CACCGACACAAATTCCTGAGAATT
14





hDMD-E44g12-bottom
aaactatgcttttACCTGCAGGCGC
360





hDMD-E44g12-top
CACCGCGCCTGCAGGTaaaagcata
361





hDMD-E44g13-top
CACCGtttACCTGCAGGCGATTTGA
362





hDMD-E44g13-bottom
aaacTCAAATCGCCTGCAGGTaaaC
363





hDMD-E44g14-bottom
aaacAACAGATCTGTCAAATCGCC
364





hDMD-E44g14-top
CACCGGCGATTTGACAGATCTGTT
365





hDMD-E44g15-bottom
aaacCTGTTAGCCACTGATTAAATC
366





hDMD-E44g15-top
CACCGATTTAATCAGTGGCTAACAG
367





hDMD-E44g16-bottom
aaacACAGAAGCTGAACAGTTTCTC
368





hDMD-E44g16-top
CACCGAGAAACTGTTCAGCTTCTGT
369





hDMD-E44g17-bottom
aaacTTCAGCTTCTGTTAGCCACTC
370





hDMD-E44g17-top
CACCGAGTGGCTAACAGAAGCTGAA
371





hDMD-E44g18-bottom
aaacTCTGAGAAACTGTTCAGCTTC
372





hDMD-E44g18-top
CACCGAAGCTGAACAGTTTCTCAGA
373





hDMD-E44g19-bottom
aaacAGAATTGGGAACATGCTAAAC
374





hDMD-E44g19-top
CACCGTTTAGCATGTTCCCAATTCT
375





hDMD-E44g20-bottom
aaacAATACAAATGGTATCTTAAGC
376





hDMD-E44g20-top
CACCGCTTAAGATACCATTTGTATT
377





hDMD-E44g21-bottom
aaacAAGATACCATTTGTATTTAGC
378





hDMD-E44g21-top
CACCGCTAAATACAAATGGTATCTT
379





hDMD-E44g22-bottom
aaacACCTTAAGATACCATTTGTAC
380





hDMD-E44g22-top
CACCGTACAAATGGTATCTTAAGGT
381





hDMD-E44g23-bottom
aaacAAGGTAAGTCTTTGATTTGTC
382





hDMD-E44g23-top
CACCGACAAATCAAAGACTTACCTT
383





hDMD-E46g1-top
CACCGttattcttctttctccagGC
384





hDMD-E46g1-bottom
aaacGCCTGGAGAAAGAAGAATAA
385





hDMD-E46g2-top
CACCGAATTTTATTCTTCTTTCTCC
386





hDMD-E46g2-bottom
aaacGGAGAAAGAAGAATAAAATT
387





hDMD-E46g5-bottom
aaacGGCTAGAAGAACAAAAGAATC
29





hDMD-E46g5-top
CACCGATTCTTTTGTTCTTCTAGCC
30





hDMD-E46g6-bottom
aaacACCATAAAACAAATTCATTTC
31





hDMD-E46g6-top
CACCGAAATGAATTTGTTTTATGGT
32





hDMD-E46g7-top
CACCGTGAATTTGTTTTATGGTTGG
33





hDMD-E46g7-bottom
aaacCCAACCATAAAACAAATTCAC
34





hDMD-E46g8-bottom
aaacTGACTTGCTCAAGCTTTTCTC
35





hDMD-E46g8-top
CACCGAGAAAAGCTTGAGCAAGTCA
36





hDMD-E46g9-top
CACCGTTCTTCTAGCCTGGAGAAAG
388





hDMD-E46g9-bottom
aaacCTTTCTCCAGGCTAGAAGAAC
389





hDMD-E46g10-bottom
aaacGAGAAAGAAGAATAAAATTGC
390





hDMD-E46g10-top
CACCGCAATTTTATTCTTCTTTCTC
391





hDMD-E46g11-bottom
aaacTCTCCAGGCTAGAAGAACAAC
392





hDMD-E46g11-top
CACCGTTGTTCTTCTAGCCTGGAGA
393





hDMD-E46g12-bottom
aaacCAGGCTAGAAGAACAAAAGAC
394





hDMD-E46g12-top
CACCGTCTTTTGTTCTTCTAGCCTG
395





hDMD-E46g13-bottom
aaacCTAGCCTGGAGAAAGAAGAAC
396





hDMD-E46g13-top
CACCGTTCTTCTTTCTCCAGGCTAG
397





hDMD-E46g14-bottom
aaacAAAGAAAAGCTTGAGCAAGTC
398





hDMD-E46g14-top
CACCGACTTGCTCAAGCTTTTCTTT
399





hDMD-E46g15-bottom
aaacGCTCAAGCTTTTCTTTTAGTC
400





hDMD-E46g15-top
CACCGACTAAAAGAAAAGCTTGAGC
401





hDMD-E46g16-bottom
aaacGAGCAAGTCAAGGTAATTTTC
402





hDMD-E46g16-top
CACCGAAAATTACCTTGACTTGCTC
403





hDMD-E46g17-bottom
aaacGACTTGCTCAAGCTTTTCTTC
404





hDMD-E46g17-top
CACCGAAGAAAAGCTTGAGCAAGTC
405





hDMD-E46g18-top
CACCGTCTCCAGGCTAGAAGAACAA
406





hDMD-E46g18-bottom
aaacTTGTTCTTCTAGCCTGGAGA
407





hDMD-E46g19-top
CACCGAGAACAAAAGAATATCTTGT
408





hDMD-E46g19-bottom
aaacACAAGATATTCTTTTGTTCT
409





hDMD-E46g20-top
CACCGTATCTTGTCAGAATTTCAAA
410





hDMD-E46g20-bottom
aaacTTTGAAATTCTGACAAGATA
411





hDMD-E50g1-top
CACCGTGTATGCTTTTCTGTTAAAG
412





hDMD-E50g1-bottom
aaacCTTTAACAGAAAAGCATACA
413





hDMD-E50g2-top
CACCGATGTGTAT GCTTTT CT GTTA
414





hDMD-E50g2-bottom
aaacTAACAGAAAAGCATACACAT
415





hDMD-E50g3-top
CACCGTGTATGCTTTTCTGTTAAA
416





hDMD-E50g3-bottom
aaacTTTAACAGAAAAGCATACAC
417





hDMD-E50g4-top
CACCGATGCTTTTCTGTTAAAGAGG
418





hDMD-E50g4-bottom
aaacCCTCTTTAACAGAAAAGCAT
419





hDMD-E50g5-top
CACCGTCTTCTAACTTCCTCTTTAA
420





hDMD-E50g5-bottom
aaacTTAAAGAGGAAGTTAGAAGA
421





hDMD-E50g6-top
CACCGTAACTTCCTCTTTAACAGAA
422





hDMD-E50g6-bottom
aaacTTCTGTTAAAGAGGAAGTTA
423





hDMD-E50g7-top
CACCGTTTTCTGTTAAAGAGGAAGT
424





hDMD-E50g7-bottom
aaacACTTCCTCTTTAACAGAAAA
425





hDMD-E50g8-top
CACCGTCTGTTAAAGAGGAAGTTAG
426





hDMD-E50g8-bottom
aaacCTAACTTCCTCTTTAACAGA
427





hDMD-E50g9-top
CACCGAAGAGGAAGTTAGAAGATCT
428





hDMD-E50g9-bottom
aaacAGATCTTCTAACTTCCTCTT
429





hDMD-E50g10-top
CACCGAGTTAGAAGATCTGAGCTCT
430





hDMD-E50g10-bottom
aaacAGAGCTCAGATCTTCTAACT
431





hDMD-E50g11-top
CACCGTAGAAGATCTGAGCTCTGAG
432





hDMD-E50g11-bottom
aaacCTCAGAGCTCAGATCTTCTA
433





hDMD-E50g12-top
CACCGAGATCTGAGCTCTGAGTGGA
434





hDMD-E50g12-bottom
aaacTCCACTCAGAGCTCAGATCT
435





hDMD-E50g13-top
CACCGACCGCCTTCCACTCAGAGCT
436





hDMD-E50g13-bottom
aaacAGCTCTGAGTGGAAGGCGGT
437





hDMD-E50g14-top
CACCGGTTTACCGCCTTCCACTCA
438





hDMD-E50g14-bottom
aaacTGAGTGGAAGGCGGTAAACC
439





hDMD-E50g15-top
CACCGAAGCAGCCTGACCTAGCTCC
440





hDMD-E50g15-bottom
aaacGGAGCTAGGTCAGGCTGCTT
441





hDMD-E50g16-top
CACCGTCAGTCCAGGAGCTAGGTC
442





hDMD-E50g16-bottom
aaacGACCTAGCTCCTGGACTGA
443





hDMD-E50g17-top
CACCGGTCAGTCCAGGAGCTAGGT
444





hDMD-E50g17-bottom
aaacACCTAGCTCCTGGACTGACC
445





hDMD-E50g18-top
CACCGTAGTGGTCAGTCCAGGAGCT
446





hDMD-E50g18-bottom
aaacAGCTCCTGGACTGACCACTA
447





hDMD-E50g19-top
CACCGATAGTGGTCAGTCCAGGAGC
448





hDMD-E50g19-bottom
aaacGCTCCTGGACTGACCACTAT
449





hDMD-E50g20-top
CACCGTCCAATAGTGGTCAGTCCAG
450





hDMD-E50g20-bottom
aaacCTGGACTGACCACTATTGGA
451





hDMD-E50g21-top
CACCGCTCCAATAGTGGTCAGTCC
452





hDMD-E50g21-bottom
aaacGGACTGACCACTATTGGAGC
453





hDMD-E50g22-top
CACCGTTACAGGCTCCAATAGTGGT
454





hDMD-E50g22-bottom
aaacACCACTATTGGAGCCTGTAA
455





hDMD-E50g23-top
CACCGATACTTACAGGCTCCAATAG
456





hDMD-E50g23-bottom
aaacCTATTGGAGCCTGTAAGTAT
457





hDMD-E50g24-top
CACCGAGTATACTTACAGGCTCCAA
458





hDMD-E50g24-bottom
aaacTTGGAGCCTGTAAGTATACT
459





hDMD-E50g25-top
CACCGCTCCTGGACTGACCACTAT
460





hDMD-E50g25-bottom
aaacATAGTGGTCAGTCCAGGAGC
461





hDMD-E50g26-top
CACCGTCCTGGACTGACCACTATTG
462





hDMD-E50g26-bottom
aaacCAATAGTGGTCAGTCCAGGA
463





hDMD-E50g27-top
CACCGTGACCACTATTGGAGCCTGT
464





hDMD-E50g27-bottom
aaacACAGGCTCCAATAGTGGTCA
465





hDMD-E50g28-top
CACCGATGGGATCCAGTATACTTAC
466





hDMD-E50g28-bottom
aaacGTAAGTATACTGGATCCCAT
467





hDMD-E50g29-top
CACCGAATGGGATCCAGTATACTTA
468





hDMD-E50g29-bottom
aaacTAAGTATACTGGATCCCATT
469





hDMD-E50g30-top
CACCGATTGGAGCCTGTAAGTATAC
470





hDMD-E50g30-bottom
aaacATTGGAGCCTGTAAGTATAC
471





hDMD-E51g4-top
CACCGTCATCTCGTTGATATCCTCA
472





hDMD-E51g4-bottom
aaacTGAGGATATCAACGAGATGA
473





hDMD-E51g5-top
CACCGCGAGATGATCATCAAGCAGA
474





hDMD-E51g5-bottom
aaacTCTGCTTGATGATCATCTCG
475





hDMD-E51g6-top
CACCGTGACCTTGAGGATATCAAC
476





hDMD-E51g6-bottom
aaacGTTGATATCCTCAAGGTCAC
477





hDMD-E51g7-top
CACCGTCAACGAGATGATCATCAAG
478





hDMD-E51g7-bottom
aaacCTTGATGATCATCTCGTTGA
479





hDMD-E51g8-top
CACCGACGAGATGATCATCAAGCAG
480





hDMD-E51g8-bottom
aaacCTGCTTGATGATCATCTCGT
481





hDMD-E53g1-top
CACCGATTTATTTTTCCTTTTATTC
482





hDMD-E53g1-bottom
aaacGAATAAAAGGAAAAATAAAT
483





hDMD-E53g2-top
CACCGTTTCCTTTTATTCTAGTTGA
484





hDMD-E53g2-bottom
aaacTCAACTAGAATAAAAGGAAA
485





hDMD-E53g3-top
CACCGTGATTCTGAATTCTTTCAAC
486





hDMD-E53g3-bottom
aaacGTTGAAAGAATTCAGAATCA
487





hDMD-E53g4-top
CACCGAAAGAAAATCACAGAAACCA
488





hDMD-E53g4-bottom
aaacTGGTTTCTGTGATTTTCTTT
489





hDMD-E53g5-top
CACCGAAAATCACAGAAACCAAGGT
490





hDMD-E53g5-bottom
aaacACCTTGGTTTCTGTGATTTT
491





hDMD-E53g6-top
CACCGGTATCTTTGATACTAACCT
492





hDMD-E53g6-bottom
aaacAGGTTAGTATCAAAGATACC
493





hDMD-E53g7-bottom
aaacACTGATTCTGAATTCTTTCAC
47





hDMD-E53g7-top
CACCGTGAAAGAATTCAGAATCAGT
48





hDMD-E53g8-bottom
aaacTCAGAACCGGAGGCAACAGTC
49





hDMD-E53g8-top
CACCGACTGTTGCCTCCGGTTCTGA
50





hDMD-E53g9-top
CACCGTACAAGAACACCTTCAGAAC
51





hDMD-E53g9-bottom
aaacGTTCTGAAGGTGTTCTTGTAC
52





hDMD-E53g10-bottom
aaacCCGGTTCTGAAGGTGTTCTTC
53





hDMD-E53g10-top
CACCGAAGAACACCTTCAGAACCGG
54





hDMD-E53g11-bottom
aaacGAGGCAACAGTTGAATGAAAC
55





hDMD-E53g11-top
CACCGTTTCATTCAACTGTTGCCTC
56





hDMD-E53g12-top
CACCGTGTTAAAGGATTCAACACAA
57





hDMD-E53g12-bottom
aaacTTGTGTTGAATCCTTTAACAC
58





hDMD-E53g13-bottom
aaacGCCATTGTGTTGAATCCTTTC
59





hDMD-E53g13-top
CACCGAAAGGATTCAACACAATGGC
60





hDMD-E53g14-top
CACCGAATTCAGAATCAGTGGGATG
494





hDMD-E53g14-bottom
aaacCATCCCACTGATTCTGAATT
495





hDMD-E53g15-bottom
aaacCTGATTCTGAATTCTTTCAAC
496





hDMD-E53g15-top
CACCGTTGAAAGAATTCAGAATCAG
497





hDMD-E53g16-top
CACCGACAGTTGAATGAAATGTTAA
498





hDMD-E53g16-bottom
aaacTTAACATTTCATTCAACTGTC
499





hDMD-E53g17-top
CACCGACCTTCAGAACCGGAGGCAA
500





hDMD-E53g17-bottom
aaacTTGCCTCCGGTTCTGAAGGTC
501





hDMD-E53g18-top
CACCGAATTCTTTCAACTAGAATAA
502





hDMD-E53g18-bottom
aaacTTATTCTAGTTGAAAGAATTC
503





hDMD-E53g19-top
CACCGTTATTCTAGTTGAAAGAATT
504





hDMD-E53g19-bottom
aaacAATTCTTTCAACTAGAATAAC
505





hDMD-E53g20-top
CACCGTAGTTGAAAGAATTCAGAAT
506





hDMD-E53g20-bottom
aaacATTCTGAATTCTTTCAACTAC
507





hDMD-E53g21-top
CACCGATGAAGTACAAGAACACCTT
508





hDMD-E53g21-bottom
aaacAAGGTGTTCTTGTACTTCATC
509





hDMD-E53g22-top
CACCGAACTGTTGCCTCCGGTTCTG
510





hDMD-E53g22-bottom
aaacCAGAACCGGAGGCAACAGTTC
511





hDMD-E53g23-top
CACCGCAAGAACACCTTCAGAACCG
512





hDMD-E53g23-bottom
aaacCGGTTCTGAAGGTGTTCTTGC
513





hDMD-E53g24-top
CACCGCAAGAACACCTTCAGAACCG
514





hDMD-E53g24-bottom
aaacCGGTTCTGAAGGTGTTCTTGC
515
















TABLE 9







Sequence of primers for generating sgRNA targeting mouse


Dmd exon 44, exon 46, and exon 53









ID
Sequence (5′-3′)
SEQ ID NO.












mDmd-E43g1-top
CACCGATTTGCAACAAATCTCAGGT
516





mDmd-E43g1-bottom
aaacACCTGAGATTTGTTGCAAAT
517





mDmd-E43g2-top
CACCGAGAATGTACAAGGAACGACA
518





mDmd-E43g2-bottom
aaacTGTCGTTCCTTGTACATTCT
519





mDmd-E43g3-top
CACCGAATGTACAAGGAACGACAA
520





mDmd-E43g3-bottom
aaacTTGTCGTTCCTTGTACATTC
521





mDmd-E43g4-bottom
aaacACCCTTGTCGTTCCTTGTAC
522





mDmd-E43g4-top
CACCGTACAAGGAACGACAAGGGT
523





mDmd-E44g1-bottom
aaacATAATTTGAAAACATGGATGC
15





mDmd-E44g1-top
CACCGCATCCATGTTTTCAAATTAT
16





mDmd-E44g2-bottom
aaacTTTTTCAACTGATCTGTCGAC
17





mDmd-E44g2-top
CACCGTCGACAGATCAGTTGAAAAA
18





mDmd-E44g3-bottom
aaacGTTTTCAGGATTTTGTGTCTC
19





mDmd-E44g3-top
CACCGAGACACAAAATCCTGAAAAC
20





mDmd-E44g4-bottom
aaacGAAAACTGGGAACATGCTAAC
21





mDmd-E44g4-top
CACCGTTAGCATGTTCCCAGTTTTC
22





mDmd-E44g5-bottom
aaacAGTTTTCAGGATTTTGTGTC
23





mDmd-E44g5-top
CACCGACACAAAATCCTGAAAACT
24





mDmd-E44g6-bottom
aaacTTTGTATTTAGCATGTTCCC
25





mDmd-E44g6-top
CACCGGGAACATGCTAAATACAAA
26





mDmd-E44g7-bottom
aaacTAAGATACCATTTGTATTTAC
27





mDmd-E44g7-top
CACCGTAAATACAAATGGTATCTTA
28





mDmd-E44g8-bottom
aaacTAATTTGAAAACATGGATGAC
524





mDmd-E44g8-top
CACCGTCATCCATGTTTTCAAATTA
525





mDmd-E44g9-bottom
aaacTCGAATCGCCTATAATTTGAC
526





mDmd-E44g9-top
CACCGTCAAATTATAGGCGATTCGA
527





mDmd-E44g10-bottom
aaacAATGAAGTTGAACAGTTTTTC
528





mDmd-E44g10-top
CACCGAAAAACTGTTCAACTTCATT
529





mDmd-E44g11-bottom
aaacTTCAACTTCATTCAGCCATTC
530





mDmd-E44g11-top
CACCGAATGGCTGAATGAAGTTGAA
531





mDmd-E44g12-top
CACCGAAGTTGAACAGTTTTTCAAA
532





mDmd-E44g12-bottom
aaacTTTGAAAAACTGTTCAACTTC
533





mDmd-E44g13-bottom
aaacAAAACTGGGAACATGCTAAAC
534





mDmd-E44g13-top
CACCGTTTAGCATGTTCCCAGTTTT
535





mDmd-E44g14-bottom
aaacAATACAAATGGTATCTTAAGC
536





mDmd-E44g14-top
CACCGCTTAAGATACCATTTGTATT
537





mDmd-E44g15-bottom
aaacAAGATACCATTTGTATTTAGC
538





mDmd-E44g15-top
CACCGCTAAATACAAATGGTATCTT
539





mDmd-E44g16-bottom
aaacACCTTAAGATACCATTTGTAC
540





mDmd-E44g16-top
CACCGTACAAATGGTATCTTAAGGT
541





mDmd-E44g17-bottom
aaacAAGGTAAGACTTTGAGATTTC
542





mDmd-E44g17-top
CACCGAAATCTCAAAGTCTTACCTT
543





mDmd-E44g18-top
CACCGTTATAGGCGATTCGACAGAT
544





mDmd-E44g18-bottom
aaacATCTGTCGAATCGCCTATAA
545





mDmd-E44g19-top
CACCGcatggatgaaataaggtaag
546





mDmd-E44g19-bottom
aaacCTTACCTTATTTCATCCATG
547





mDmd-E44g20-top
CACCGctgaaaaaatgaagccagca
548





mDmd-E44g20-bottom
aaacTGCTGGCTTCATTTTTTCAG
549





mDmd-E44g21-top
CACCGATTTAATCAATGGCTGAATG
550





mDmd-E44g21-bottom
aaacCATTCAGCCATTGATTAAAT
551





mDmd-E46g1-top
CACCGAATTTTGTTATTCTTAATAC
37





mDmd-E46g1-bottom
aaacGTATTAAGAATAACAAAATTC
38





mDmd-E46g2-bottom
aaacGCCACAAAACAAATTCATTTC
39





mDmd-E46g2-top
CACCGAAATGAATTTGTTTTGTGGC
40





mDmd-E46g3-bottom
aaacAGTGGAGTAATAGCAATGTTC
41





mDmd-E46g3-top
CACCGAACATTGCTATTACTCCACT
42





mDmd-E46g4-top
CACCGAGCTGCTGCTCATCTCCAAG
43





mDmd-E46g4-bottom
aaacCTTGGAGATGAGCAGCAGCTC
44





mDmd-E46g5-top
CACCGAGAACAACTTGAACAAGTCA
45





mDmd-E46g5-bottom
aaacTGACTTGTTCAAGTTGTTCTC
46





mDmd-E46g6-bottom
aaacTATTAAGAATAACAAAATTC
552





mDmd-E46g6-top
CACCGAATTTTGTTATTCTTAATA
553





mDmd-E46g7-top
CACCGTTGTTCTTCAATCCTGTATT
554





mDmd-E46g7-bottom
aaacAATACAGGATTGAAGAACAAC
555





mDmd-E46g8-bottom
aaacCAATCCTGTATTAAGAATAAC
556





mDmd-E46g8-top
CACCGTTATTCTTAATACAGGATTG
557





mDmd-E46g9-bottom
aaacTTGTTCTTCAATCCTGTATTC
558





mDmd-E46g9-top
CACCGAATACAGGATTGAAGAACAA
559





mDmd-E46g10-bottom
aaacCTCATCTCCAAGTGGAGTAAC
560





mDmd-E46g10-top
CACCGTTACTCCACTTGGAGATGAG
561





mDmd-E46g11-bottom
aaacGTTCAAGTTGTTCTTTTAGC
562





mDmd-E46g11-top
CACCGCTAAAAGAACAACTTGAAC
563





mDmd-E46g12-top
CACCGAAATTACCTTGACTTGTTC
564





mDmd-E46g12-bottom
aaacGAACAAGTCAAGGTAATTTC
565





mDmd-E46g13-top
CACCGAAGAACAACTTGAACAAGTC
566





mDmd-E46g13-bottom
aaacGACTTGTTCAAGTTGTTCTTC
567





mDmd-E46g14-top
CACCGACTTGTTCAAGTTGTTCTTT
568





mDmd-E46g14-bottom
aaacAAAGAACAACTTGAACAAGT
569





mDmd-E46g15-top
CACCGacacctctcagggatttagg
570





mDmd-E46g15-bottom
aaacCCTAAATCCCTGAGAGGTGT
571





mDmd-E46g16-top
CACCGttcccttattaaaatcctca
572





mDmd-E46g16-bottom
aaacTGAGGATTTTAATAAGGGAA
573





mDmd-E46g17-top
CACCGctttatacaaataggccctg
574





mDmd-E46g17-bottom
aaacCAGGGCCTATTTGTATAAAG
575





mDmd-E51g4-top
CACCGTGAAATGATCATCAAACAGA
576





mDmd-E51g4-bottom
aaacTCTGTTTGATGATCATTTCA
577





mDmd-E51g5-top
CACCGTCAATGAAATGATCATCAAA
578





mDmd-E51g5-bottom
aaacTTTGATGATCATTTCATTGA
579





mDmd-E51g6-top
CACCGATGAAATGATCATCAAACAG
580





mDmd-E51g6-bottom
aaacCTGTTTGATGATCATTTCAT
581





mDmd-E51g7-top
CACCGTGATCATCAAACAGAAGGTA
582





mDmd-E51g7-bottom
aaacTACCTTCTGTTTGATGATCA
583





mDmd-E53g1-top
CACCGTGAAAGAATTCAGATTCAGT
61





mDmd-E53g1-bottom
aaacACTGAATCTGAATTCTTTCAC
62





mDmd-E53g2-bottom
aaacCATCCCACTGAATCTGAATTC
63





mDmd-E53g2-top
CACCGAATTCAGATTCAGTGGGATG
64





mDmd-E53g3-bottom
aaacGTTCTGCAGCTGTTCTTGAAC
65





mDmd-E53g3-top
CACCGTTCAAGAACAGCTGCAGAAC
66





mDmd-E53g4-bottom
aaacTTAACATTTCATTCAACTGTC
67





mDmd-E53g4-top
CACCGACAGTTGAATGAAATGTTAA
68





mDmd-E53g5-bottom
aaacTTGTGTTGAATCCTTTAACAC
69





mDmd-E53g5-top
CACCGTGTTAAAGGATTCAACACAA
70





mDmd-E53g6-bottom
aaacGCCATTGTGTTGAATCCTTTC
71





mDmd-E53g6-top
CACCGAAAGGATTCAACACAATGGC
72





mDmd-E53g7-top
CACCGAAAGAAGATCACAGAAACCA
584





mDmd-E53g7-bottom
aaacTGGTTTCTGTGATCTTCTTT
585





mDmd-E53g8-top
CACCGTTGAAAGAATTCAGATTCAG
586





mDmd-E53g8-bottom
aaacCTGAATCTGAATTCTTTCAA
587





mDmd-E53g9-top
CACCGAGTGGGATGAGGTTCAAGAA
588





mDmd-E53g9-bottom
aaacTTCTTGAACCTCATCCCACTC
589





mDmd-E53g10-top
CACCGAGCTGCAGAACAGGAGACAA
590





mDmd-E53g10-bottom
aaacTTGTCTCCTGTTCTGCAGCTC
591





mDmd-E53g11-top
CACCGTGAATCTGAATTCTTTCAAC
592





mDmd-E53g11-bottom
aaacGTTGAAAGAATTCAGATTCAC
593





mDmd-E53g12-top
CACCGCTTTCAACTGGAATAAAAAT
594





mDmd-E53g12-bottom
aaacATTTTTATTCCAGTTGAAAGC
595





mDmd-E53g13-top
CACCGCTTATTTTTATTCCAGTTGA
596





mDmd-E53g13-bottom
aaacTCAACTGGAATAAAAATAAGC
597





mDmd-E53g14-top
CACCGTTATTCCAGTTGAAAGAATT
598





mDmd-E53g14-bottom
aaacAATTCTTTCAACTGGAATAAC
599





mDmd-E53g15-top
CACCGCAGTTGAAAGAATTCAGATT
600





mDmd-E53g15-bottom
aaacAATCTGAATTCTTTCAACTGC
601





mDmd-E53g16-top
CACCGAATTCAGATTCAGTGGGAT
602





mDmd-E53g16-bottom
aaacATCCCACTGAATCTGAATTC
603





mDmd-E53g17-top
CACCGATTCAGTGGGATGAGGTTC
604





mDmd-E53g17-bottom
aaacGAACCTCATCCCACTGAATC
605





mDmd-E53g18-top
CACCGATGAGGTTCAAGAACAGCTG
606





mDmd-E53g18-bottom
aaacCAGCTGTTCTTGAACCTCATC
607





mDmd-E53g19-top
CACCGTTCAAGAACAGCTGCAGAA
608





mDmd-E53g19-bottom
aaacTTCTGCAGCTGTTCTTGAAC
609





mDmd-E53g20-top
CACCGAACTGTTGTCTCCTGTTCTG
610





mDmd-E53g20-bottom
aaacCAGAACAGGAGACAACAGTTC
611





mDmd-E53g21-top
CACCGCAAGAACAGCTGCAGAACAG
612





mDmd-E53g21-bottom
aaacCTGTTCTGCAGCTGTTCTTGC
613





mDmd-E53g22-top
CACCGAAGATCACAGAAACCAAGGT
614





mDmd-E53g22-bottom
aaacACCTTGGTTTCTGTGATCTTC
615





mDmd-E53g23-top
CACCGCAGAAACCAAGGTTAGTGTC
616





mDmd-E53g23-bottom
aaacGACACTAACCTTGGTTTCTGC
2261
















TABLE 10







Sequence of primers for generating sgRNA targeting Dmd exon 43,


exon 45, and exon 52 to generate the mouse models











Mouse

SEQ


ID
Model
Sequence (5′-3′)
ID NO.













mDmd-Ex43-N1-Top
Δ43 DMD
caccgaagtgagttccaggacagcc
73





mDmd-Ex43-N1-Bottom
Δ43 DMD
aaacGGCTGTCCTGGAACTCACTTc
74





mDmd-Ex43-N2-Top
Δ43 DMD
caccgTTATTAGTACTAACTCAGAA
75





mDmd-Ex43-N2-Bottom
Δ43 DMD
aaacTTCTGAGTTAGTACTAATAAc
76





mDmd-Ex43-N3-Top
Δ43 DMD
caccgtagtactaataaaagtagtc
77





mDmd-Ex43-N3-Bottom
Δ43 DMD
aaacGACTACTTTTATTAGTACTAc
78





mDmd-Ex43-C1-Top
Δ43 DMD
caccgATGTTGAAATACACTGCTCT
79





mDmd-Ex43-C1-Bottom
Δ43 DMD
aaacAGAGCAGTGTATTTCAACATc
80





mDmd-Ex43-C2-Top
Δ43 DMD
caccgGTAAATATCAACTTCTAAAT
81





mDmd-Ex43-C2-Bottom
Δ43 DMD
aaacATTTAGAAGTTGATATTTACc
82





mDmd-Ex43-C3-Top
Δ43 DMD
caccggtatttccctatttttaatg
83





mDmd-Ex43-C3-Bottom
Δ43 DMD
aaacCATTAAAAATAGGGAAATACc
84





mDmd-Exon45-5-G1-top
Δ45 DMD
CACCGaactaatatatccaaatact
85





mDmd-Exon45-5-G1-bot
Δ45 DMD
AAACAGTATTTGGATATATTAGTT C
86





mDmd-Exon45-5-G2-top
Δ45 DMD
CACCGttaaacatagaacatccttg
87





mDmd-Exon45-5-G2-bot
Δ45 DMD
AAACCAAGGATGTTCTATGTTTAA C
88





mDmd-Exon45-5-G3-top
Δ45 DMD
CACCGaatcgaatttgctcttgaga
89





mDmd-Exon45-5-G3-bot
Δ45 DMD
AAACTCTCAAGAGCAAATTCGATT C
90





mDmd-Exon45-3-G4-top
Δ45 DMD
CACCGagtttgtgctaaaaatcatg
91





mDmd-Exon45-3-G4-bot
Δ45 DMD
AAACCATGATTTTTAGCACAAACT C
92





mDmd-Exon45-3-G5-top
Δ45 DMD
CACCGgctttacccagctgaatcac
93





mDmd-Exon45-3-G5-bot
Δ45 DMD
AAACGTGATTCAGCTGGGTAAAGC C
94





mDmd-Exon45-3-G6-top
Δ45 DMD
CACCGaattttcacagcattgctta
95





mDmd-Exon45-3-G6-bot
Δ45 DMD
AAACTAAGCAATGCTGTGAAAATT C
96





mDmd-Ex52-N1-Top
Δ52 DMD
CACCGATATATCTTAAATGATGTAT
97





mDmd-Ex52-N1-bottom
Δ52 DMD
AAACATACATCATTTAAGATATATC
98





mDmd-Ex52-N2-Top
Δ52 DMD
CACCGAATAATAATGCTGTTTGATG
99





mDmd-Ex52-N2-bottom
Δ52 DMD
aaacCATCAAACAGCATTATTATTc
100





mDmd-Ex52-N3-Top
Δ52 DMD
CACCGGATAGTTAGAAATGACTCCA
101





mDmd-Ex52-N3-bottom
Δ52 DMD
AAACTGGAGTCATTTCTAACTATCC
102





mDmd-Ex52-C1-Top
Δ52 DMD
CACCGtctttaatgtctgtctacta
103





mDmd-Ex52-C1-Bottom
Δ52 DMD
aaacTAGTAGACAGACATTAAAGAc
104





mDmd-Ex52-C2-Top
Δ52 DMD
caccgtgcaatttcatagtatattt
105





mDmd-Ex52-C2-Bottom
Δ52 DMD
aaacAAATATACTATGAAATTGCAc
106





mDmd-Ex52-C3-Top
Δ52 DMD
caccgccaagttaatcaaattgttc
107





mDmd-Ex52-C3-Bottom
Δ52 DMD
aaacGAACAATTTGATTAACTTGGc
108
















TABLE 11







Sequence of primers


for in vitro transcription of sgRNA











Mouse

SEQ ID


ID
Model
Sequence (5′-3′)
 NO.





IVTT7-mDmd-
Δ43
gaattgTAATACGACTCACTATA
109


E43-N1-F
DMD
GGGaagtgagttccaggacagcc






IVTT7-mDmd-
Δ43
gaattgTAATACGACTCACTATA
110


E43-N2-F
DMD
GGGTTATTAGTACTAACTCAGAA






IVTT7-mDmd-
Δ43
gaattgTAATACGACTCACTATA
111


E43-N3-F
DMD
GGGtagtactaataaaagtagtc






IVTT7-mDmd-
Δ43
gaattgTAATACGACTCACTATA
112


E43-C1-F
DMD
GGGATGTTGAAATACACTGCTCT






IVTT7-mDmd-
Δ43
gaattgTAATACGACTCACTATA
113


E43-C2-F
DMD
GGGGTAAATATCAACTTCTAAAT






IVTT7-mDmd-
Δ43
gaattgTAATACGACTCACTATA
114


E43-C3-F
DMD
GGGgtatttccctatttttaatg






IVTT7-mDmd-
Δ52
gaattgTAATACGACTCACTATA
115


E52-N1-F
DMD
GGGatatatcttaaatgatgtat






IVTT7-mDmd-
Δ52
gaattgTAATACGACTCACTATA
116


E52-N2-F
DMD
GGGaataataatgctgtttgatg






IVTT7-mDmd-
Δ52
gaattgTAATACGACTCACTATA
117


E52-N3-F
DMD
GGGGATAGTTAGAAATGACTCCA






IVTT7-mDmd-
Δ52
gaattgTAATACGACTCACTATA
118


E52-C1-F
DMD
GGGtctttaatgtctgtctacta






IVTT7-mDmd-
Δ52
gaattgTAATACGACTCACTATA
119


E52-C2-F
DMD
GGGtgcaatttcatagtatattt






IVTT7-mDmd-
Δ52
gaattgTAATACGACTCACTATA
120


E52-C3-F
DMD
GGGccaagttaatcaaattgttc






T7-Rv
All
AAAAGCACCGACTCGGTGCCAC
121
















TABLE 12







Sequence of primers for genotyping











Mouse

SEQ ID


ID
Model
Sequence (5′-3′)
NO.





mDmd-Ex43-
Δ43 DMD
gcacacctttaatcccagca
122


geno-F1








mDmd-Ex43-
Δ43 DMD
tctgcagccgaataccttca
123


geno-R1








mDmd-Ex45-
Δ45 DMD
tctcattctgtgcattcttggt
124


geno-F1








mDmd-Ex45-
Δ45 DMD
gctttccaattaccatagcatgc
125


geno-R2








mDmd-Ex52-
Δ52 DMD
agggaatctgctgtccttga
126


geno-F1








mDmd-Ex52-
Δ52 DMD
tggaggttagatttcacaactgt
127


geno-R1









VII. EXAMPLES

The following examples are included to demonstrate preferred embodiments of the disclosure. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the disclosure, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the disclosure.


Example 1
Materials and Methods

Study Approval. All experimental procedures involving animals in this study were reviewed and approved by the University of Texas Southwestern Medical Center's Institutional Animal Care and Use Committee.


Plasmids. The pSpCas9(BB)-2A-GFP (PX458) plasmid containing the human codon optimized SpCas9 gene with 2A-EGFP and the backbone of sgRNA was purchased from Addgene (Plasmid #48138). Cloning of sgRNA was done using Bbs I sites. The AAV TRISPR-CK8-GFP plasmid containing three sgRNAs driven by U6, H1 or 7SK promoter and GFP driven by CK8 promoter.


Human iPSCs maintenance and nucleofection. Human iPSCs were cultured in mTeSR™ 1 media (STEMCELL Technologies) and passaged approximately every 4 days (1:14 split ratio). One hour before nucleofection, iPSCs were treated with 10 μM ROCK inhibitor (Y-27632) and dissociated into single cells using Accutase (Innovative Cell Technologies, Inc.). 1×106 iPSCs were mixed with 5 μg of pX458-gRNA-2A-GFP plasmid and nucleofected using the P3 Primary Cell 4D-Nucleofector X kit (Lonza) according to manufacturer's protocol. After nucleofection, iPSCs were cultured in mTeSR™ 1 media supplemented with 10 μM ROCK inhibitor, penicillin-streptomycin (1:100) (ThermoFisher Scientific) and 100 μg/ml Primosin (InvivoGen). Three days post-nucleofection, GFP(+) and (−) cells were sorted by FACS and subjected to T7E1 assay. Single clones derived from GFP(+) iPSCs were picked and sequenced.


Genomic DNA isolation. Genomic DNA of mouse 10T1/2 fibroblasts, mouse N2a cells, human 293 and human iPSCs was isolated using DirectPCR (cell) lysis reagent (VIAGEN) according to manufacturer's protocol.


PCR amplification of genomic DNA. Genomic DNA was PCR-amplified using GoTaq DNA polymerase (Promega) with primers. PCR products were gel purified and subcloned into pCRII-TOPO vector (Invitrogen) according to the manufacturer's protocol. Individual clones were picked and the DNA was sequenced.


T7E1 analysis of PCR products. Mismatched duplex DNA was obtained by denaturing/renaturing of 25 μl of the genomic PCR product using the following conditions: 95° C. for 5 mins, 95° C. to 85° C. (−2.0° C./seconds), 85° C. to 25° C. (−0.1° C./seconds), hold at 4° C. Then 25 μl of the mismatched duplex DNA was incubated with 2.7 μl of 10× NEB buffer 2 and 0.3 μl of T7E1 (New England BioLabs) at 37° C. for 90 minutes. The T7E1 digested PCR product was analyzed by 2% agarose gel electrophoresis.


Human cardiomyocyte differentiation. Human iPSCs were cultured in mTeSR™ 1 media for 3 days until they reached 90% confluence. To differentiate the iPSCs to cardiomyocytes, the iPSCs were cultured in CDM3-C media for 2 days, followed by CDM3-59 media for 2 days, followed by CDM3 media for 6 days, followed by selective media for 10 days and lastly by basal media for 2 days. Then, the cardiomyocytes were dissociated using TrypLE media and re-plated at 2×106 per well in a 6-well dish.


Dystrophin Western blot analysis. After 30 days post-differentiation, 2×106 cardiomyocytes were harvested and lysed with lysis buffer (10% SDS, 62.5 mM Tris pH=6.8, 1 mM EDTA, and protease inhibitor). Cell lysates were passed through a 22 G syringe and then a 27 G syringe, 10 times each. Protein concentration was determined by BCA assay and 50 ug of total protein was loaded onto an acrylamide gel. After running at 100V (20 mA) for 5 hours and followed by 1 hour 20 min transfer to PVDF membrane at 35V (200 mA) at 4° C. The blot was incubated with mouse anti-dystrophin antibody (MANDYS8, Sigma-Aldrich, D8168) at 4° C. overnight and with goat anti-mouse HRP antibody (Bio-Rad Laboratories) at RT for 1 hour. The blot was developed using Western Blotting Luminol Reagent (Santa Cruz, sc-2048). The loading control was determined by blotting with mouse anti-vinculin antibody (Sigma-Aldrich, V9131).


CRISPR/Cas9-mediated exon deletion in mice. Two single-guide RNA (sgRNA) specific intronic regions surrounding exon 43, exon 45, or exon 52 sequence of the mouse Dmd locus were cloned into vector pX458 using the primers from Table 10. For the in vitro transcription of sgRNA, T7 promoter sequence was added to the sgRNA template by PCR using the primers from Table 11. The gel purified PCR products were used as template for in vitro transcription using the MEGAshortscript T7 Kit (Life Technologies). sgRNA were purified by MEGAclear kit (Life Technologies) and eluted with nuclease-free water (Ambion). The concentration of guide RNA was measured by a NanoDrop instrument (Thermo Scientific).


Genotyping of 443, 445, and 452 DMD Mice. 443, 445, and 452 DMD mice were genotyped using primers encompassing the targeted region from Table 12. Tail biopsies were digested in 100 μL of 25 mM NaOH, 0.2 mM EDTA (pH 12) for 20 min at 95° C. Tails were briefly centrifuged followed by addition of 100 μL of 40 mM Tris.HCl (pH 5) and mixed to homogenize. Two microliters of this reaction was used for subsequent PCR reactions with the primers below, followed by gel electrophoresis.


Histological analysis of muscles. Skeletal muscles from WT, Δ43 DMD, Δ45 DMD, and Δ52 DMD mice were individually dissected and cryoembedded in a 1:2 volume mixture of Gum Tragacanth powder (Sigma-Aldrich) to Tissue Freezing Medium (TFM) (Triangle Bioscience). All embeds were snap frozen in isopentane heat extractant supercooled to −155° C. Resulting blocks were stored overnight at −80° C. prior to sectioning. Eight-micron transverse sections of skeletal muscle, and frontal sections of heart were prepared on a Leica CM3050 cryostat and air-dried prior to same day staining. H&E staining was performed according to established staining protocols and dystrophin immunohistochemistry was performed using MANDYS8 monoclonal antibody (Sigma-Aldrich) with modifications to manufacturer's instructions. In brief, cryostat sections were thawed and rehydrated/delipidated in 1% triton/phosphate-buffered-saline, pH 7.4 (PBS). Following delipidation, sections were washed free of Triton, incubated with mouse IgG blocking reagent (M.O.M. Kit, Vector Laboratories), washed, and sequentially equilibrated with MOM protein concentrate/PBS, and MANDYS8 diluted 1:1800 in MOM protein concentrate/PBS. Following overnight primary antibody incubation at 4° C., sections were washed, incubated with MOM biotinylated anti-mouse IgG, washed, and detection completed with incubation of Vector fluorescein-avidin DCS. Nuclei were counterstained with propidium iodide (Molecular Probes) prior to cover slipping with Vectashield.


Example 2
Results

Δ43, Δ45, and Δ52 DMD mouse models recapitulate muscle dystrophy phenotype. To investigate CRISPR/Cas9-mediated exon skipping and reframing in vivo, three mimics of the human “hot spot” regions were generated in three mouse models by deleting the exon 43, exon 45, and exon 52, respectively, using CRISPR/Cas9 system directed by 2 single guide RNAs (sgRNA) (FIG. 1A and Table 9). The inventors designed and validated sgRNAs targeting 5′ end and 3′ end of Dmd exon 43, exon 45, and exon 52. C57BL/6 zygotes were co-injected with in vitro transcribed Cas9 mRNA and in vitro transcribed sgRNAs, and then re-implanted into pseudo-pregnant females.


The deletion of Dmd exon 43, exon 45, and exon 52 was confirmed by DNA genotyping. 1-month old mice lacking exon 43, exon 45, or exon 52 showed pronounced dystrophic muscle changes. (FIG. 1B). The deletion of these exons placed the dystrophin gene out of frame leading to the absence of dystrophin protein in skeletal muscle and heart (FIG. 1C). Serum analysis of the Δ43, Δ45, and Δ52 DMD mice shows a significant increase of creatine kinase (CK) level, which is a sign of muscle damage (FIG. 1D). Taken together, dystrophin protein expression, muscle histology, and serum creatine kinase level validated dystrophic phenotype of the Δ43, Δ45, and Δ52 DMD mouse models.


Identification of optimal sgRNAs for CRISPR/Cas9 correction of DMD exon 43, exon 45, and exon 52 deletions. Skipping or reframing of exon 44, exon 46, and exon 53 would apply to about 18% of DMD patients. To restore the ORF of the DMD patient with exon 43, exon 45, or exon 53 deletion, the inventors applied single guide RNA to disrupt the splicing junction of exon 44, exon 46, and exon 53 respectively, which results in reframing of the exon downstream of the deleted exon and restoration of the protein reading frame (FIGS. 2A, 4A, 5A, 6A, 7A, 8A). To test sgRNA efficiency within these regions, the inventors designed sgRNAs to target the region flanking splicing junctions of exon 44, exon 46 or exon 53 to reframe or skip the targeting exon (Table 2, Table 3, Table 8, Table 9, FIGS. 4B, 5B, 6B, 7B). The inventors validated the cleavage efficiency of these gRNAs in both mouse 10T1/2 or mouse N2a cells or human 293 cells. By T7E1 assay, the inventors demonstrated that exon 44 sgRNAs, exon 46 sgRNAs, and exon 53 sgRNAs efficiently cause DNA cleavage at Dmd exon target locus in mouse cells (FIG. 2B). By T7E1 assay, the inventors also demonstrated that exon 44 sgRNAs, exon 46 sgRNAs, and exon 53 sgRNAs efficiently cause DNA cleavage at DMD exon target locus in human cells (FIG. 2C). Additionally, by TIDE assay, the inventors demonstrated that 3 exon 44 sgRNAs (hDMD-E44g4, hDMD-E44g8, hDMD-E44g11), 2 exon 46 sgRNAs (hDMD-E46g2, hDMD-E46g8), and 3 exon 53 sgRNAs (hDMD-E53g14, hDMD-e53g15, hDMD-E53g23) efficiently cause DNA cleavage at a DMD exon target locus in human cells (FIG. 8B). hDMD-E45g4 was included as a positive control.


DMD iPSC-derived cardiomyocytes express dystrophin after CRISPR/Cas9 mediated genome editing by exon skipping and exon reframing. The inventors then generated iPSCs from DMD patients (TX16) that have deletion of exon 52 and an isogenic iPSC line with deletion of exon 43 (Δ43 DMD) and a deletion of exon 45 (Δ45 DMD). The inventors then tested exon 44 editing sgRNAs on Δ43 and Δ45 DMD iPSCs and showed restoration of dystrophin protein expression by Western blot analysis and immunostaining of iPSC-derived cardiomyocytes (FIGS. 3A, 3C, 6C, 6D). The inventors also tested exon 53 editing sgRNAs on TX16 patient derived iPSCs. The restoration of dystrophin in these TX16 DMD patient iPSCs was confirmed by Western blot analysis and immunostaining of iPSC-derived cardiomyocytes (FIG. 3B and FIG. 3C).


All of the compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this disclosure have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the disclosure. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the disclosure as defined by the appended claims.


VIII. REFERENCES

The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference.

  • Angel et al., Mol. Cell. Biol., 7:2256, 1987a.
  • Angel et al., Cell, 49:729, 1987b.
  • Baichwal and Sugden, In: Gene Transfer, Kucherlapati (Ed), NY, Plenum Press, 117-148, 1986.
  • Banerji et al., Cell, 27(2 Pt 1):299-308, 1981.
  • Banerji et al., Cell, 33(3):729-740, 1983.
  • Barnes et al., J. Biol. Chem., 272(17):11510-7, 1997.
  • Benvenisty and Neshif, Proc. Natl. Acad. Sci. USA, 83:9551-9555, 1986.
  • Berkhout et al., Cell, 59:273-282, 1989.
  • Bhaysar et al., Genomics, 35(1):11-23, 1996.
  • Bikard et al., Nucleic Acids Res. 41(15): 7429-7437, 2013.
  • Blanar et al., EMBO J., 8:1139, 1989.
  • Bodine and Ley, EMBO J., 6:2997, 1987.
  • Boshart et al., Cell, 41:521, 1985.
  • Bosze et al., EMBO J., 5(7):1615-1623, 1986.
  • Braddock et al., Cell, 58:269, 1989.
  • Brinster et al., Proc. Natl. Acad. Sci. USA, 82(13):4438-4442, 1985.
  • Bulla and Siddiqui, J. Virol., 62:1437, 1986.
  • Calcedo et al., J Infect Dis, 199(3): p. 381-90, 2009.
  • Campbell and Villarreal, Mol. Cell. Biol., 8:1993, 1988.
  • Campere and Tilghman, Genes and Dev., 3:537, 1989.
  • Campo et al., Nature, 303:77, 1983.
  • Celander and Haseltine, J. Virology, 61:269, 1987.
  • Celander et al., J. Virology, 62:1314, 1988.
  • Chandler et al., Cell, 33:489, 1983.
  • Chang et al., Mol. Cell. Biol., 9:2153, 1989.
  • Chang et al., Stem Cells., 27:1042-1049, 2009.
  • Chatterjee et al., Proc. Natl. Acad. Sci. USA, 86:9114, 1989.
  • Chen and Okayama, Mol. Cell Biol., 7:2745-2752, 1987.
  • Cho et al., Nat. Biotechnol. 31(3): 230-232, 2013.
  • Choi et al., Cell, 53:519, 1988.
  • Choi et al., Curr Gene Ther. 5(3); 299-310, 2005.
  • Coffin, In: Virology, Fields et al. (Eds.), Raven Press, NY, 1437-1500, 1990.
  • Cohen et al., J. Cell. Physiol., 5:75, 1987.
  • Costa et al., Mol. Cell. Biol., 8:81, 1988.
  • Couch et al., Am. Rev. Resp. Dis., 88:394-403, 1963.
  • Coupar et al., Gene, 68:1-10, 1988.
  • Cripe et al., EMBO J., 6:3745, 1987.
  • Culotta and Hamer, Mol. Cell. Biol., 9:1376, 1989.
  • Dandolo et al., J. Virology, 47:55-64, 1983.
  • De Villiers et al., Nature, 312(5991):242-246, 1984.
  • Deschamps et al., Science, 230:1174-1177, 1985.
  • Donnelly et al., J. Gen. Virol. 82, 1027-1041, 2001.
  • Dubensky et al., Proc. Natl. Acad. Sci. USA, 81:7529-7533, 1984.
  • Edbrooke et al., Mol. Cell. Biol., 9:1908, 1989.
  • Edlund et al., Science, 230:912-916, 1985.
  • EP 0273085
  • Fechheimer et al., Proc Natl. Acad. Sci. USA, 84:8463-8467, 1987.
  • Feng and Holland, Nature, 334:6178, 1988.
  • Ferkol et al., FASEB J., 7:1081-1091, 1993.
  • Firak et al., Mol. Cell. Biol., 6:3667, 1986.
  • Foecking et al., Gene, 45(1):101-105, 1986.
  • Fraley et al., Proc Natl. Acad. Sci. USA, 76:3348-3352, 1979
  • Franz et al., Cardoscience, 5(4):235-43, 1994.
  • Friedmann, Science, 244:1275-1281, 1989.
  • Fujita et al., Cell, 49:357, 1987.
  • Gao et al., J Virol, 78(12): p. 6381-8, 2004.
  • Ghosh and Bachhawat, In: Liver Diseases, Targeted Diagnosis and Therapy Using Specific Receptors and Ligands, Wu et al. (Eds.), Marcel Dekker, NY, 87-104, 1991.
  • Ghosh-Choudhury et al., EMBO J., 6:1733-1739, 1987.
  • Gilles et al., Cell, 33:717, 1983.
  • Gloss et al., EMBO J., 6:3735, 1987.
  • Godbout et al., Mol. Cell. Biol., 8:1169, 1988.
  • Gomez-Foix et al., J. Biol. Chem., 267:25129-25134, 1992.
  • Goodbourn and Maniatis, Proc. Natl. Acad. Sci. USA, 85:1447, 1988.
  • Goodbourn et al., Cell, 45:601, 1986.
  • Gopal, Mol. Cell Biol., 5:1188-1190, 1985.
  • Gopal-Srivastava et al., J. Mol. Cell. Biol., 15(12):7081-90, 1995.
  • Graham and Prevec, In: Methods in Molecular Biology: Gene Transfer and Expression Protocol, Murray (Ed.), Humana Press, Clifton, N.J., 7:109-128, 1991.
  • Graham and van der Eb, Virology, 52:456-467, 1973.
  • Graham et al., J. Gen. Virol., 36:59-72, 1977.
  • Greene et al., Immunology Today, 10:272, 1989
  • Grosschedl and Baltimore, Cell, 41:885, 1985.
  • Grunhaus and Horwitz, Seminar in Virology, 3:237-252, 1992.
  • Harland and Weintraub, J. Cell Biol., 101:1094-1099, 1985.
  • Haslinger and Karin, Proc. Natl. Acad. Sci. USA, 82:8572, 1985.
  • Hauber and Cullen, J. Virology, 62:673, 1988.
  • Hen et al., Nature, 321:249, 1986.
  • Hensel et al., Lymphokine Res., 8:347, 1989.
  • Hermonat and Muzycska, Proc. Nat'l Acad. Sci. USA, 81:6466-6470, 1984.
  • Herr and Clarke, Cell, 45:461, 1986.
  • Hersdorffer et al., DNA Cell Biol., 9:713-723, 1990.
  • Herz and Gerard, Proc. Nat'l. Acad. Sci. USA 90:2812-2816, 1993.
  • Hirochika et al., J. Virol., 61:2599, 1987.
  • Holbrook et al., Virology, 157:211, 1987.
  • Horlick and Benfield, Mol. Cell. Biol., 9:2396, 1989.
  • Horwich et al., J. Virol., 64:642-650, 1990.
  • Hsu et al., Natl Biotechnol. 31:827-832, 2013
  • Huang et al., Cell, 27:245, 1981.
  • Hug et al., Mol. Cell. Biol., 8:3065, 1988.
  • Hwang et al., Mol. Cell. Biol., 10:585, 1990.
  • Imagawa et al., Cell, 51:251, 1987.
  • Imbra and Karin, Nature, 323:555, 1986.
  • Imler et al., Mol. Cell. Biol., 7:2558, 1987.
  • Imperiale and Nevins, Mol. Cell. Biol., 4:875, 1984.
  • Jakobovits et al., Mol. Cell. Biol., 8:2555, 1988.
  • Jameel and Siddiqui, Mol. Cell. Biol., 6:710, 1986.
  • Jaynes et al., Mol. Cell. Biol., 8:62, 1988.
  • Jinek et al., Science 337, 816-821, 2012.
  • Johnson et al., Mol. Cell. Biol., 9:3393, 1989.
  • Jones and Shenk, Cell, 13:181-188, 1978.
  • Kadesch and Berg, Mol. Cell. Biol., 6:2593, 1986.
  • Kaneda et al., Science, 243:375-378, 1989.
  • Karin et al., Mol. Cell. Biol., 7:606, 1987.
  • Karlsson et al., EMBO J., 5:2377-2385, 1986.
  • Katinka et al., Cell, 20:393, 1980.
  • Kato et al., J Biol Chem., 266(6):3361-3364, 1991.
  • Kawamoto et al., Mol. Cell. Biol., 8:267, 1988.
  • Kelly et al., J. Cell Biol., 129(2):383-96, 1995.
  • Kiledjian et al., Mol. Cell. Biol., 8:145, 1988.
  • Kimura et al., Dev. Growth Differ., 39(3):257-65, 1997.
  • Klamut et al., Mol. Cell. Biol., 10:193, 1990.
  • Klein et al., Nature, 327:70-73, 1987.
  • Koch et al., Mol. Cell. Biol., 9:303, 1989.
  • Kriegler and Botchan, In: Eukaryotic Viral Vectors, Gluzman (Ed.), Cold Spring Harbor: Cold Spring Harbor Laboratory, NY, 1982.
  • Kriegler and Botchan, Mol. Cell. Biol., 3:325, 1983.
  • Kriegler et al., Cell, 38:483, 1984.
  • Kriegler et al., Cell, 53:45, 1988.
  • Kuhl et al., Cell, 50:1057, 1987.
  • Kunz et al., Nucl. Acids Res., 17:1121, 1989.
  • LaPointe et al., Hypertension, 27(3):715-22, 1996
  • LaPointe et al., J. Biol. Chem., 263(19):9075-8, 1988.
  • Larsen et al., Proc. Natl. Acad. Sci. USA., 83:8283, 1986.
  • Laspia et al., Cell, 59:283, 1989.
  • Latimer et al., Mol. Cell. Biol., 10:760, 1990.
  • Le Gal La Salle et al., Science, 259:988-990, 1993.
  • Lee et al., Nature, 294:228, 1981.
  • Levinson et al., Nature, 295:79, 1982.
  • Levrero et al., Gene, 101:195-202, 1991.
  • Lin et al., Mol. Cell. Biol., 10:850, 1990.
  • Luria et al., EMBO J., 6:3307, 1987.
  • Lusky and Botchan, Proc. Natl. Acad. Sci. USA, 83:3609, 1986.
  • Lusky et al., Mol. Cell. Biol., 3:1108, 1983.
  • Majors and Varmus, Proc. Natl. Acad. Sci. USA, 80:5866, 1983.
  • Mali et al., Science 339, 823-826, 2013a.
  • Mali et al., Nat Methods 10, 957-963, 2013b.
  • Mali et al., Nat. Biotechnol. 31:833-838, 2013c.
  • Mann et al., Cell, 33:153-159, 1983.
  • Markowitz et al., J. Virol., 62:1120-1124, 1988.
  • McNeall et al., Gene, 76:81, 1989.
  • Melnick et al., J Bacteriol, 90(1): p. 271-4, 1965.
  • Miksicek et al., Cell, 46:203, 1986.
  • Mordacq and Linzer, Genes and Dev., 3:760, 1989.
  • Moreau et al., Nucl. Acids Res., 9:6047, 1981.
  • Moss et al., J. Gen. Physiol., 108(6):473-84, 1996.
  • Muesing et al., Cell, 48:691, 1987.
  • Nathwani et al., New Engl. J. Med. 365:2357-2365, 2011.
  • Nicolas and Rubinstein, In: Vectors: A survey of molecular cloning vectors and their uses, Rodriguez and Denhardt (Eds.), Stoneham: Butterworth, 494-513, 1988.
  • Nicolau and Sene, Biochim. Biophys. Acta, 721:185-190, 1982.
  • Nicolau et al., Methods Enzymol., 149:157-176, 1987.
  • Ondek et al., EMBO J., 6:1017, 1987.
  • Ornitz et al., Mol. Cell. Biol., 7:3466, 1987.
  • Palmiter et al., Cell, 29:701, 1982a.
  • Palmiter et al., Nature, 300:611, 1982b.
  • Paskind et al., Virology, 67:242-248, 1975.
  • Pech et al., Mol. Cell. Biol., 9:396, 1989.
  • Perales et al., Proc. Natl. Acad. Sci. USA, 91(9):4086-4090, 1994.
  • Perez-Stable and Constantin, Mol. Cell. Biol., 10:1116, 1990.
  • Picard et al., Nature, 307:83, 1984.
  • Pinkert et al., Genes and Dev., 1:268, 1987.
  • Ponta et al., Proc. Natl. Acad. Sci. USA, 82:1020, 1985.
  • Porton et al., Mol. Cell. Biol., 10:1076, 1990.
  • Potter et al., Proc. Natl. Acad. Sci. USA, 81:7161-7165, 1984.
  • Queen and Baltimore, Cell, 35:741, 1983.
  • Quinn et al., Mol. Cell. Biol., 9:4713, 1989.
  • Racher et al., Biotech. Techniques, 9:169-174, 1995.
  • Ragot et al., Nature, 361:647-650, 1993.
  • Redondo et al., Science, 247:1225, 1990.
  • Reisman and Rotter, Mol. Cell. Biol., 9:3571, 1989.
  • Renan, Radiother. Oncol., 19:197-218, 1990.
  • Resendez Jr. et al., Mol. Cell. Biol., 8:4579, 1988.
  • Rich et al., Hum. Gene Ther., 4:461-476, 1993.
  • Ridgeway, In: Vectors: A survey of molecular cloning vectors and their uses, Stoneham: Butterworth, 467-492, 1988.
  • Ripe et al., Mol. Cell. Biol., 9:2224, 1989.
  • Rippe et al., Mol. Cell Biol., 10:689-695, 1990.
  • Riffling et al., Nuc. Acids Res., 17:1619, 1989.
  • Rosen et al., Cell, 41:813, 1988.
  • Rosenfeld et al., Cell, 68:143-155, 1992.
  • Rosenfeld et al., Science, 252:431-434, 1991.
  • Roux et al., Proc. Natl. Acad. Sci. USA, 86:9079-9083, 1989.
  • Sambrook and Russell, Molecular Cloning: A Laboratory Manual 3rd Ed., Cold Spring Harbor Laboratory Press, 2001.
  • Sakai et al., Genes and Dev., 2:1144, 1988.
  • Satake et al., J. Virology, 62:970, 1988.
  • Schaffner et al., J. Mol. Biol., 201:81, 1988.
  • Searle et al., Mol. Cell. Biol., 5:1480, 1985.
  • Sharp et al., Cell, 59:229, 1989.
  • Shaul and Ben-Levy, EMBO J., 6:1913, 1987.
  • Sherman et al., Mol. Cell. Biol., 9:50, 1989.
  • Sleigh et al., J. EMBO, 4:3831, 1985.
  • Spalholz et al., Cell, 42:183, 1985.
  • Spandau and Lee, J. Virology, 62:427, 1988.
  • Spandidos and Wilkie, EMBO J., 2:1193, 1983.
  • Stephens and Hentschel, Biochem. 1, 248:1, 1987.
  • Stratford-Perricaudet and Perricaudet, In: Human Gene Transfer, Cohen-Haguenauer and Boiron (Eds.), John Libbey Eurotext, France, 51-61, 1991.
  • Stratford-Perricaudet et al., Hum. Gene. Ther., 1:241-256, 1990.
  • Stuart et al., Nature, 317:828, 1985.
  • Sullivan and Peterlin, Mol. Cell. Biol., 7:3315, 1987.
  • Swartzendruber and Lehman, J. Cell. Physiology, 85:179, 1975.
  • Takebe et al., Mol. Cell. Biol., 8:466, 1988.
  • Tavernier et al., Nature, 301:634, 1983.
  • Taylor and Kingston, Mol. Cell. Biol., 10:165, 1990a.
  • Taylor and Kingston, Mol. Cell. Biol., 10:176, 1990b.
  • Taylor et al., J. Biol. Chem., 264:15160, 1989.
  • Temin, In: Gene Transfer, Kucherlapati (Ed.), NY, Plenum Press, 149-188, 1986.
  • Thiesen et al., J. Virology, 62:614, 1988.
  • Top et al., J. Infect. Dis., 124:155-160, 1971.
  • Tronche et al., Mol. Biol. Med., 7:173, 1990.
  • Trudel and Constantini, Genes and Dev. 6:954, 1987.
  • Tur-Kaspa et al., Mol. Cell Biol., 6:716-718, 1986.
  • Tyndell et al., Nuc. Acids. Res., 9:6231, 1981.
  • Vannice and Levinson, J. Virology, 62:1305, 1988.
  • Varmus et al., Cell, 25:23-36, 1981.
  • Vasseur et al., Proc Natl. Acad. Sci. USA., 77:1068, 1980.
  • Wagner et al., Proc. Natl. Acad. Sci. USA, 87(9):3410-3414, 1990.
  • Wang and Calame, Cell, 47:241, 1986.
  • Wang et al., Cell, 153:910-910, 2013.
  • Weber et al., Cell, 36:983, 1984.
  • Weinberger et al. Mol. Cell. Biol., 8:988, 1984.
  • Winoto et al., Cell, 59:649, 1989.
  • Wong et al., Gene, 10:87-94, 1980.
  • Wu and Wu, Adv. Drug Delivery Rev., 12:159-167, 1993.
  • Wu and Wu, Biochemistry, 27:887-892, 1988.
  • Wu and Wu, J. Biol. Chem., 262:4429-4432, 1987.
  • Wu et al., Molecular Therapy 14(3), 316-327, 2006.
  • Wu et al., Cell Stem Cell 13, 659-662, 2013.
  • Wu et al., Nat Biotechnol 32, 670-676, 2014.
  • Yamauchi-Takihara et al., Proc. Natl. Acad. Sci. USA, 86(10):3504-3508, 1989.
  • Yang et al., Proc. Natl. Acad. Sci. USA, 87:9568-9572, 1990.
  • Yutzey et al. Mol. Cell. Biol., 9:1397, 1989.
  • Zelenin et al., FEBS Lett., 280:94-96, 1991.
  • Ziober and Kramer, J. Bio. Chem., 271(37):22915-22922, 1996.

Claims
  • 1. A nucleic acid comprising: a sequence encoding a single guide RNA (sgRNA) comprising a spacer sequence and a scaffold sequence;wherein the spacer sequence comprises the sequence of any one of SEQ ID NOs: 135-146, 170-260, 337-339, or 617.
  • 2. The nucleic acid of claim 1, wherein the scaffold sequence comprises the sequence of any one of SEQ ID NO: 147-153.
  • 3. The nucleic acid of claim 1, wherein the nucleic acid comprises one copy of the sequence encoding the sgRNA.
  • 4. The nucleic acid of claim 1, wherein the nucleic acid comprises two, three, four, or five copies of the sequence encoding the sgRNA.
  • 5. The nucleic acid of claim 1, wherein the nucleic acid comprises a sequence encoding a promoter, wherein the promoter drives expression of the sgRNA.
  • 6. The nucleic acid of claim 5, wherein the nucleic acid comprises three copies of the sequence encoding the sgRNA, wherein the nucleic acid comprises a sequence encoding a first promoter and expression of the first copy of the sgRNA is driven by the first promoter, wherein the nucleic acid comprises a sequence encoding a second promoter and expression of the second copy of the sgRNA is driven by the second promoter, and wherein the nucleic acid comprises a sequence encoding a third promoter and expression of the third copy of the sgRNA is driven by the third promoter.
  • 7. The nucleic acid of claim 1, wherein the nucleic acid further comprises a sequence encoding a nuclease.
  • 8. The nucleic acid of claim 7, wherein the nuclease is a Type II, Type V-A, Type V-B, Type V-C, Type V-U, or Type VI-B nuclease.
  • 9. The nucleic acid of claim 7, wherein the nuclease is a TAL nuclease, a meganuclease, or a zinc-finger nuclease.
  • 10. The nucleic acid of claim 7, wherein the nuclease is a Cas9, Cas12a, Cas12b, Cas12c, Tnp-B like, Cas13a (C2c2), Cas13b, or Cas14 nuclease.
  • 11. The nucleic acid of claim 10, wherein the nuclease is a Cas9 nuclease.
  • 12. The nucleic acid of claim 11, wherein the Cas9 is a Streptococcus pyogenes or Streptococcus aureus Cas9.
  • 13. The nucleic acid of claim 11, wherein the nuclease is a modified Cas9 nuclease.
  • 14. The nucleic acid of claim 12, wherein the nuclease is a modified Streptococcus pyogenes Cas9 or a modified Streptococcus aureus Cas9.
  • 15. A recombinant vector comprising the nucleic acid of claim 1.
  • 16. The recombinant vector of claim 15, wherein the recombinant vector is a plasmid.
  • 17. The recombinant vector of claim 15, wherein the recombinant vector is an expression vector.
  • 18. The recombinant vector of claim 15, wherein the recombinant vector is a viral vector.
  • 19. The recombinant vector of claim 18, wherein the viral vector is a lentiviral vector, a retroviral vector, an adenoviral vector, or an adeno-associated virus (AAV) vector.
  • 20. The recombinant vector of claim 19, wherein the viral vector is an adeno-associated virus (AAV) vector.
  • 21. The recombinant vector of claim 20, wherein the serotype of the AAV vector is selected from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVRh74, AAV2i8, AAVRh10, AAV39, AAV43, AAVRh8, avian AAV, bovine AAV, canine AAV, equine AAV, and ovine AAV.
  • 22. The recombinant vector of claim 20, wherein the AAV vector is replication-defective or conditionally replication defective.
  • 23. The recombinant vector of claim 21, wherein the serotype of the AAV vector is AAV9.
  • 24. A non-viral vector comprising the nucleic acid of claim 1, wherein the non-viral vector comprises calcium phosphate, liposomes, nanoparticles, and/or lipid emulsions.
  • 25. An AAV expression cassette comprising: a first inverted terminal repeat (ITR);a first promoter;the nucleic acid of claim 1; anda second ITR.
  • 26. The AAV expression cassette of claim 25, wherein the AAV expression cassette further comprises a polyadenosine (polyA) sequence.
  • 27. The AAV expression cassette of claim 25, wherein one or both of the first ITR and the second ITR are isolated or derived from any one of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVRh74, AAV2i8, AAVRh10, AAV39, AAV43, AAVRh8, avian AAV, bovine AAV, canine AAV, equine AAV, or ovine AAV.
  • 28. The AAV expression cassette of claim 25.
  • 29. An AAV vector comprising the nucleic acid of claim 1.
  • 30. The AAV vector of claim 28, wherein the AAV vector has the serotype of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVRh74, AAV2i8, AAVRh10, AAV39, AAV43, AAVRh8, avian AAV, bovine AAV, canine AAV, equine AAV, or ovine AAV.
  • 31. The AAV vector of claim 29, wherein the AAV vector is replication-defective or conditionally replication defective.
  • 32. The AAV vector of claim 30, wherein the serotype of the AAV vector is AAV9.
  • 33. A composition comprising the nucleic acid of claim 1.
  • 34. The composition of claim 33, further comprising a pharmaceutically acceptable carrier.
  • 35. A cell comprising the nucleic acid of claim 1.
  • 36. The cell of claim 35, wherein the cell is a stem cell.
  • 37. The cell of claim 35, wherein the cell is a mammalian cell.
  • 38. The cell of claim 37, wherein the cell is a human cell.
  • 39. A composition comprising the AAV vector of claim 29.
  • 40. The composition of claim 39, further comprising a pharmaceutically acceptable carrier.
  • 41. A method of correcting a gene defect in a cell, the method comprising contacting the cell with: the nucleic acid of claim 1.
  • 42. The method of claim 41, wherein the cell is a stem cell.
  • 43. The method of claim 41, wherein the cell is a mammalian cell.
  • 44. The method of claim 43, wherein the cell is a human cell.
  • 45. A method of treating a subject suffering from Duchenne muscular dystrophy, the method comprising administering to the subject a therapeutically effective amount of: the nucleic acid of claim 1.
  • 46. A method of treating a subject suffering from Duchenne muscular dystrophy, the method comprising administering to the subject: a first vector, wherein the first vector is the recombinant vector of claim 1, anda second vector, wherein the second vector encodes a nuclease.
  • 47. The method of claim 46, wherein the nuclease is a Type II, Type V-A, Type V-B, Type V-C, Type V-U, or Type VI-B nuclease.
  • 48. The method of claim 46, wherein the nuclease is a TAL nuclease, a meganuclease, or a zinc-finger nuclease.
  • 49. The method of claim 46, wherein the nuclease is a Cas9, Cas12a, Cas12b, Cas12c, Tnp-B like, Cas13a (C2c2), Cas13b, or Cas14 nuclease.
  • 50. The method of claim 49, wherein the nuclease is a Cas9 nuclease.
  • 51. The method of claim 50, wherein the Cas9 is a Streptococcus pyogenes or Streptococcus aureus Cas9.
  • 52. The method of claim 50, wherein the nuclease is a modified Cas9 nuclease.
  • 53. The method of claim 52, wherein the nuclease is a modified Streptococcus pyogenes Cas9 or a modified Streptococcus aureus Cas9.
  • 54. The method of claim 46, wherein the second vector is a plasmid.
  • 55. The method of claim 46, wherein the second vector is an expression vector.
  • 56. The method of claim 46, wherein the second vector is a viral vector.
  • 57. The method of claim 56, wherein the viral vector is a lentiviral vector, a retroviral vector, an adenoviral vector, or an adeno-associated virus (AAV) vector.
  • 58. The method of claim 57, wherein the viral vector is an adeno-associated virus (AAV) vector.
  • 59. The method of claim 58, wherein the serotype of the AAV vector is selected from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVRh74, AAV2i8, AAVRh10, AAV39, AAV43, AAVRh8, avian AAV, bovine AAV, canine AAV, equine AAV, and ovine AAV.
  • 60. The method of claim 46, wherein the second vector is a non-viral vector, wherein the non-viral vector comprises calcium phosphate, liposomes, nanoparticles, and/or lipid emulsions.
  • 61. The method of claim 46, wherein the administering induces a frameshift mutation in a target nucleic acid sequence in a cell of the patient.
  • 62. The method of claim 61, wherein the frameshift mutation comprises a deletion of at least one nucleotide, wherein the number of nucleotides deleted is not a multiple of 3.
  • 63. The method of claim 62, wherein the frameshift mutation comprises a deletion of 1, 2, 4, 5, 7, 8, 10, 11, 13, 14, 16, 17, 19 or 20 nucleotides.
  • 64. The method of claim 61, wherein the frameshift mutation comprises an insertion of at least one nucleotide, wherein the number of nucleotides inserted is not a multiple of 3.
  • 65. The method of claim 64, wherein the frameshift mutation comprises an insertion of 1, 2, 4, 5, 7, 8, 10, 11, 13, 14, 16, 17, 19 or 20 nucleotides.
  • 66. The method of claim 65, wherein the frameshift mutation comprises an insertion of 1 nucleotide.
  • 67. The method of claim 46, wherein the first vector and the second vector are administered simultaneously.
  • 68. The method of claim 46, wherein the first vector and the second vector are administered sequentially.
  • 69. The method of claim 46, wherein the first vector and the second vector are administered locally.
  • 70. The method of claim 46, wherein the first vector and the second vector are administered systemically.
  • 71. The method of claim 46, wherein the first vector and the second vector are administered by an oral, rectal, transmucosal, topical, transdermal, inhalation, intravenous, subcutaneous, intradermal, intramuscular, intra-articular, intrathecal, intraventricular, intravenous, intraperitoneal, intranasal, or intraocular route of administration.
  • 72. The method of claim 46, wherein the subject is greater than or equal to 18 years old.
  • 73. The method of claim 46, wherein the subject is less than 18 years old.
  • 74. The method of claim 73, wherein the subject is less than 2 years old.
  • 75. The method of claim 46, wherein the subject is a human.
  • 76. The method of claim 46, wherein the ratio of the first vector to the second vector is 1:1 to 1:100.
  • 77. The method of claim 46, wherein the ratio of the second vector to the first vector is 1:1 to 1:100.
  • 78. A combination therapy comprising; a first composition comprising a first vector comprising the nucleic acid of claim 1; anda second composition comprising a second vector comprising a nucleic acid that encodes a nuclease.
  • 79. The combination therapy of claim 78, wherein at least one of the first and the second composition comprises a pharmaceutically acceptable carrier.
  • 80. The combination therapy of claim 78, wherein the nuclease is a Type II, Type V-A, Type V-B, Type V-C, Type V-U, or Type VI-B nuclease.
  • 81. The combination therapy of claim 78, wherein the nuclease is a TAL nuclease, a meganuclease, or a zinc-finger nuclease.
  • 82. The combination therapy of claim 78, wherein the nuclease is a Cas9, Cas12a, Cas12b, Cas12c, Tnp-B like, Cas13a (C2c2), Cas13b, or Cas14 nuclease.
  • 83. The combination therapy of claim 82, wherein the nuclease is a Cas9 nuclease.
  • 84. The combination therapy of claim 83, wherein the Cas9 is a Streptococcus pyogenes or Streptococcus aureus Cas9.
  • 85. The combination therapy of claim 83, wherein the nuclease is a modified Cas9 nuclease.
  • 86. The combination therapy of claim 85, wherein the nuclease is a modified Streptococcus pyogenes Cas9 or a modified Streptococcus aureus Cas9.
  • 87. The composition of claim 33.
  • 88. The composition of claim 33.
  • 89. A mouse whose genome comprises (a) a deletion of exon 43 of the dystrophin gene resulting in an out of frame shift and a premature stop codon in exon 44, (b) a deletion of exon 45 resulting in an out of frame shift and premature stop codon in exon 46, or (c) a deletion of exon 52 resulting in an out of frame shift and premature stop codon in exon 53.
  • 90. The mouse of claim 89, further comprising a reporter gene located downstream of and in frame with exon 79 of the dystrophin gene, and upstream of a dystrophin 3′-UTR, wherein said reporter gene is expressed when exon 79 is translated in frame with exon 45, exon 47, or exon 54.
  • 91. The mouse of claim 90, wherein the reporter gene is luciferase.
  • 92. The mouse of claim 90, further comprising a protease coding sequence upstream of and in frame with said reporter gene, and downstream of and in frame with exon 79.
  • 93. The mouse of claim 92, wherein said protease is autocatalytic.
  • 94. The mouse of claim 93, wherein said protease is 2A protease.
  • 95. The mouse of claim 89, wherein the mouse is heterozygous for said deletion.
  • 96. The mouse of claim 89, wherein the mouse is homozygous for said deletion.
  • 97. The mouse of claim 89, wherein the mouse exhibits increased creatine kinase levels.
  • 98. The mouse of claim 89, wherein the mouse does not exhibit detectable dystrophin protein in heart or skeletal muscle.
  • 99. A method of producing the mouse of claim 89 comprising: (a1) contacting a fertilized oocyte with CRISPR/Cas9 elements and two single guide RNA (sgRNA) targeting sequences flanking exon 43, thereby creating a modified oocyte, wherein deletion of exon 43 by CRISPR/Ca9 results in an out of frame shift and a premature stop codon in exon 44;(a2) contacting a fertilized oocyte with CRISPR/Cas9 elements and two single guide RNA (sgRNA) targeting sequences flanking exon 45, thereby creating a modified oocyte, wherein deletion of exon 45 by CRISPR/Ca9 results in an out of frame shift and a premature stop codon in exon 46; or(a3) contacting a fertilized oocyte with CRISPR/Cas9 elements and two single guide RNA (sgRNA) targeting sequences flanking exon 52, thereby creating a modified oocyte, wherein deletion of exon 52 by CRISPR/Ca9 results in an out of frame shift and a premature stop codon in exon 53; and(b) transferring said modified oocyte into a recipient female.
  • 100. The method of claim 99, wherein said oocyte comprises a dystrophin gene having a reporter gene located downstream of and in frame with exon 79 of said dystrophin gene, and upstream of a dystrophin 3′-UTR, wherein said reporter gene is expressed when exon 79 is translated in frame with exon 45, exon 47, or exon 54.
  • 101. The method of claim 100, wherein the reporter gene is luciferase.
  • 102. The method of claim 100, further comprising a protease coding sequence upstream of and in frame with said reporter gene, and downstream of and in frame with exon 79.
  • 103. The method of claim 102, wherein said protease is autocatalytic.
  • 104. The method of claim 103, wherein said protease is 2A protease.
  • 105. The method of claim 99, wherein the mouse is heterozygous for said deletion.
  • 106. The method of claim 99, wherein the mouse is homozygous for said deletion.
  • 107. The method of claim 99, wherein the mouse exhibits increased creatine kinase levels.
  • 108. The method of claim 99, wherein the mouse does not exhibit detectable dystrophin protein in heart or skeletal muscle.
  • 109. An isolated cell obtained from the mouse of claim 89.
  • 110. The isolated cell of claim 109, further comprising a reporter gene located downstream of and in frame with exon 79 of the dystrophin gene, and upstream of a dystrophin 3′-UTR, wherein said reporter gene is expressed when exon 79 is translated in frame with exon 45, exon 47, or exon 54.
  • 111. The isolated cell of claim 110, wherein the reporter gene is luciferase.
  • 112. The mouse of claim 110, further comprising a protease coding sequence upstream of and in frame with said reporter gene, and downstream of and in frame with exon 79.
  • 113. The cell of claim 112, wherein said protease is autocatalytic.
  • 114. The cell of claim 113, wherein said protease is 2A protease.
  • 115. The cell of claim 109, wherein the cell is heterozygous for said deletion.
  • 116. The cell of claim 109, wherein the cell is homozygous for said deletion.
  • 117. A mouse produced by a method comprising the steps of: (a1) contacting a fertilized oocyte with CRISPR/Cas9 elements and two single guide RNA (sgRNA) targeting sequences flanking exon 43, thereby creating a modified oocyte, wherein deletion of exon 43 by CRISPR/Ca9 results in an out of frame shift and a premature stop codon in exon 44;(a2) contacting a fertilized oocyte with CRISPR/Cas9 elements and two single guide RNA (sgRNA) targeting sequences flanking exon 45, thereby creating a modified oocyte, wherein deletion of exon 45 by CRISPR/Ca9 results in an out of frame shift and a premature stop codon in exon 46; or(a3) contacting a fertilized oocyte with CRISPR/Cas9 elements and two single guide RNA (sgRNA) targeting sequences flanking exon 52, thereby creating a modified oocyte, wherein deletion of exon 52 by CRISPR/Ca9 results in an out of frame shift and a premature stop codon in exon 53; and(b) transferring said modified oocyte into a recipient female.
  • 118. A method of screening a candidate substance for DMD exon-skipping activity comprising: (a) contacting a mouse according to claim 1 with a candidate substance; and(b) assessing in frame transcription and/or translation of exon 79,
  • 119. The method of claim 118, wherein the mouse does not exhibit detectable dystrophin protein in heart or skeletal muscle.
  • 120. The method of claim 118, wherein the genome of the mouse further comprises a reporter gene located downstream of and in frame with exon 79 of the dystrophin gene, and upstream of a dystrophin 3′-UTR, wherein said reporter gene is expressed when exon 79 is translated in frame with exon 45, exon 47, or exon 54.
  • 121. The method of claim 120, wherein the reporter gene is luciferase.
  • 122. The method of claim 120, wherein the genome of the mouse further comprises a protease coding sequence upstream of and in frame with said reporter gene, and downstream of and in frame with exon 79.
  • 123. The method of claim 122, wherein said protease is autocatalytic.
  • 124. The method of claim 123, wherein said protease is 2A protease.
  • 125. The method of claim 118, wherein the mouse is heterozygous for said deletion.
  • 126. The method of claim 118, wherein the mouse is homozygous for said deletion.
  • 127. The method of claim 118, wherein the mouse exhibits increased creatine kinase levels.
  • 128. An isolated nucleic acid comprising a sequence of any one of SEQ ID NO: 1-72, 340-359, or 360-515.
  • 129. A double-stranded nucleic acid formed by hybridization of SEQ ID NO: 1 and 2, SEQ ID NO: 3 and 4, SEQ ID NO: 5 and 6, SEQ ID NO: 7 and 8, SEQ ID NO: 9 and 10, SEQ ID NO: 11 and 12, SEQ ID NO: 13 and 14, SEQ ID NO: 15 and 16, SEQ ID NO: 17 and 18, SEQ ID NO: 19 and 20, SEQ ID NO: 21 and 22, SEQ ID NO: 23 and 24, SEQ ID NO: 25 and 26, SEQ ID NO: 27 and 28, SEQ ID NO: 29 and 30, SEQ ID NO: 31 and 32, SEQ ID NO: 33 and 34, SEQ ID NO: 35 and 36, SEQ ID NO: 37 and 38, SEQ ID NO: 39 and 40, SEQ ID NO: 41 and 42, SEQ ID NO: 43 and 44, SEQ ID NO: 45 and 46, SEQ ID NO: 47 and 48, SEQ ID NO: 49 and 50, SEQ ID NO: 51 and 52, SEQ ID NO: 53 and 54, SEQ ID NO: 55 and 56, SEQ ID NO: 57 and 58, SEQ ID NO: 59 and 60, SEQ ID NO: 61 and 62, SEQ ID NO: 63 and 64, SEQ ID NO: 65 and 66, SEQ ID NO: 67 and 68, SEQ ID NO: 69 and 70, and SEQ ID NO: 71 and 72.
  • 130. An expression construct comprising a nucleic acid formed by hybridization of SEQ ID NO: 1 and 2, SEQ ID NO: 3 and 4, SEQ ID NO: 5 and 6, SEQ ID NO: 7 and 8, SEQ ID NO: 9 and 10, SEQ ID NO: 11 and 12, SEQ ID NO: 13 and 14, SEQ ID NO: 15 and 16, SEQ ID NO: 17 and 18, SEQ ID NO: 19 and 20, SEQ ID NO: 21 and 22, SEQ ID NO: 23 and 24, SEQ ID NO: 25 and 26, SEQ ID NO: 27 and 28, SEQ ID NO: 29 and 30, SEQ ID NO: 31 and 32, SEQ ID NO: 33 and 34, SEQ ID NO: 35 and 36, SEQ ID NO: 37 and 38, SEQ ID NO: 39 and 40, SEQ ID NO: 41 and 42, SEQ ID NO: 43 and 44, SEQ ID NO: 45 and 46, SEQ ID NO: 47 and 48, SEQ ID NO: 49 and 50, SEQ ID NO: 51 and 52, SEQ ID NO: 53 and 54, SEQ ID NO: 55 and 56, SEQ ID NO: 57 and 58, SEQ ID NO: 59 and 60, SEQ ID NO: 61 and 62, SEQ ID NO: 63 and 64, SEQ ID NO: 65 and 66, SEQ ID NO: 67 and 68, SEQ ID NO: 69 and 70, and SEQ ID NO: 71 and 72.
  • 131. The expression construct of claim 130, wherein said expression construct is a viral vector.
  • 132. A kit comprising one or more isolated nucleic acids of any one of SEQ ID NO: 1-72, 340-359, or 360-515.
  • 133. A method of correcting a dystrophin gene defect in exon 44, exon 46 or exon 53 of the DMD gene in a subject comprising contacting a cell in said subject with Cpf1 or Cas9 and a DMD guide RNA as defined in claim 42, resulting in selective skipping of a mutant DMD exon.
  • 134. The method of claim 133, wherein said cell is a muscle cell, a satellite cell, or an iPSC or iPSC-CM.
  • 135. The method of claim 133, wherein Cas9, Cpf1 and/or DMD guide RNA are provided to said cell through expression from one or more expression vectors coding therefor.
  • 136. The method of claim 135, wherein said expression vector is a viral vector.
  • 137. The method of claim 136, wherein said viral vector is an adeno-associated viral vector.
  • 138. The method of claim 135, wherein said expression vector is a non-viral vector.
  • 139. The method of claim 133, wherein Cas9, Cpf1 or Cas9 is provided to said cell as naked plasmid DNA or chemically-modified mRNA.
  • 140. The method of claim 133, further comprising contacting said cell with a single-stranded DMD oligonucleotide to effect homology directed repair.
  • 141. The method of claim 133, wherein Cpf1 or Cas9, DMD guide RNA and/or single-stranded DMD oligonucleotide, or expression vectors coding therefor, are provided to said cell in one or more nanoparticles.
  • 142. The method of claim 133, wherein said Cpf1 or Cas9, DMD guide RNA and/or single-stranded DMD oligonucleotide are delivered directly to a muscle tissue.
  • 143. The method of claim 142, wherein said muscle tissue is tibialis anterior, quadricep, soleus, diaphragm or heart.
  • 144. The method of claim 133, wherein said Cpf1 or Cas9, DMD guide RNA and/or single-stranded DMD oligonucleotide are delivered systemically.
  • 145. The method of claim 133, wherein said subject exhibits normal dystrophin-positive myofibers and/or mosaic dystrophin-positive myofibers containing centralized nuclei.
  • 146. The method of claim 133, wherein said subject exhibits a decreased serum CK level as compared to a serum CK level prior to contacting.
  • 147. The method of claim 133, wherein said subject exhibits improved grip strength as compared to a serum CK level prior to contacting.
  • 148. The method of claim 133, wherein the correction is permanent skipping of said mutant DMD exon.
  • 149. The method of claim 133, wherein the correction is permanent skipping of more than one mutant DMD exon.
  • 150. The method of claim 133, wherein the Cpf1 or Cas9 and/or DMD guide RNA are delivered to a human iPSC with an adeno-associated viral vector.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims priority to U.S. Provisional Application Ser. No. 62/854,131, filed May 29, 2019, and U.S. Provisional Application Ser. No. 62/688,003, filed Jun. 21, 2018, each of which is incorporated by reference herein in its entirety for all purposes.

FEDERAL FUNDING SUPPORT CLAUSE

This invention was made with government support under grant no. U54 HD 087351 awarded by National Institutes of Health. The government has certain rights in the invention.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2019/038398 6/21/2019 WO 00
Provisional Applications (2)
Number Date Country
62854131 May 2019 US
62688003 Jun 2018 US