Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CRISPR/CAS9-MEDIATED EXON-SKIPPING APPROACH FOR USH2A-ASSOCIATED USHER SYNDROME
Document Type and Number:
WIPO Patent Application WO/2019/183641
Kind Code:
A1
Abstract:
Compositions for use in treating subjects with USH2A-associated retinal and/or cochlear degeneration that result from mutations in exon 13 of the USH2A gene by deletion of exon 13 from the USH2A gene or transcripts, and methods of use thereof, as well as genetically modified animals and cells.

Inventors:
PIERCE ERIC A (US)
MARGULIES CARRIE MARIE (US)
PENDSE NACHIKET D (US)
GLOSKOWSKI SEBASTIAN W (US)
LIU QIN (US)
MAEDER MORGAN L (US)
Application Number:
PCT/US2019/023934
Publication Date:
September 26, 2019
Filing Date:
March 25, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MASSACHUSETTS EYE & EAR INFIRMARY (US)
EDITAS MEDICINE INC (US)
International Classes:
A61K38/46; A61K47/26; C12N7/00; C12N9/22; C12N15/113
Domestic Patent References:
WO2018026976A12018-02-08
WO1999057318A21999-11-11
WO2016141224A12016-09-09
WO2017100791A12017-06-15
WO2016049024A22016-03-31
WO2016004925A12016-01-14
WO2017124086A12017-07-20
WO2018009562A12018-01-11
Foreign References:
US20140256802A12014-09-11
US20160340661A12016-11-24
US20170073674A12017-03-16
US20170159052A12017-06-08
CA2986021A12016-11-24
US201862647578P2018-03-23
US201962794402P2019-01-18
US5288644A1994-02-22
US20040014095A12004-01-22
US20140060163W2014-10-10
US7206639B22007-04-17
US9901080B22018-02-27
US4873191A1989-10-10
Other References:
FUSTER-GARCIA ET AL.: "USH2A Gene Editing Using the CRISPR System", MOLECULAR THERAPY: NUCLEIC ACIDS, vol. 8, 15 September 2017 (2017-09-15), pages 529 - 541, XP055572855
MAERKER ET AL., HUM MOL GENET., vol. 17, no. 1, 1 January 2008 (2008-01-01), pages 71 - 86
LIU ET AL., PROC NATL ACAD SCI USA., vol. 104, no. 11, 13 March 2007 (2007-03-13), pages 4413 - 8
REINERS ET AL., HUM MOL GENET., vol. 14, no. 24, 15 December 2005 (2005-12-15), pages 3933 - 43
HUMAN MOLECULAR GENETICS, vol. 26, pages 1157 - 1172
"GenBank", Database accession no. NC_000001.11
VAN WIJK, E. ET AL.: "Identification of 51 novel exons of the Usher syndrome type 2A (USH2A) gene that encode multiple conserved functional domains and that are mutated in patients with Usher syndrome type II", AM J HUM GENET, vol. 74, no. 4, 2004, pages 738 - 44, XP055140764
GRATI, M. ET AL.: "Localization of PDZD7 to the stereocilia ankle-link associates this scaffolding protein with the Usher syndrome protein network", J NEUROSCI, vol. 32, no. 41, 2012, pages 14288 - 93
MATSUDA, T.C.L. CEPKO: "Controlled expression of transgenes introduced by in vivo electroporation", PROC NATL ACAD SCI USA, vol. 104, no. 11, 2007, pages 1027 - 32
YAN ET AL., J HUM GENET, vol. 54, no. 12, December 2009 (2009-12-01), pages 732 - 738
LEROY ET AL., EXP EYE RES, vol. 72, no. 5, May 2001 (2001-05-01), pages 503 - 9
CONSUGAR ET AL., GENET MED, vol. 17, no. 4, April 2015 (2015-04-01), pages 253 - 261
CHURCHGILBERT, PROC. NATL. ACAD. SCI. USA, vol. 81, 1988, pages 1991 - 1995
SANGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 74, 1977, pages 5463 - 5467
SCHAFER ET AL., NAT. BIOTECHNOL., vol. 15, 1995, pages 33 - 39
SHEFFIELD ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 2766 - 2770
UNDERHILL ET AL., GENOME RES, vol. 7, 1997, pages 996 - 1005
FLAVELL ET AL., CELL, vol. 15, 1978, pages 25
GEEVER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 78, 1981, pages 5081
RACA ET AL., GENET TEST, vol. 8, no. 4, 2004, pages 387 - 94
VAN DER PUTTEN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 82, 1985, pages 6148 - 1652
MYERS ET AL., SCIENCE, vol. 230, 1985, pages 1242
COKOL ET AL., EMBO REP, vol. 1, no. 5, 15 November 2000 (2000-11-15), pages 411 - 415
FREITASCUNHA, CURR GENOMICS, vol. 10, no. 8, December 2009 (2009-12-01), pages 550 - 557
HORVATH ET AL., J BACTERIOL, vol. 190, 2008, pages 1401 - 1412
RAN, F.A. ET AL.: "In vivo genome editing using Staphylococcus aureus Cas9", NATURE, vol. 520, no. 7546, 2015, pages 186 - 191, XP055484527, DOI: 10.1038/nature14299
ZHANG ET AL., MOL CELL, vol. 50, 2013, pages 488 - 503
ZETSCHE ET AL., CELL, vol. 163, 2015, pages 759 - 771
SCHUNDER ET AL., INT J MED MICROBIOL, vol. 303, 2013, pages 51 - 60
MAKAROVA ET AL., NAT REV MICROBIOL, vol. 13, 2015, pages 722 - 736
FAGERLUND ET AL., GENOME BIOL, vol. 16, 2015, pages 251
KLEINSTIVER ET AL., NAT BIOTECHNOL., vol. 33, no. 12, December 2015 (2015-12-01), pages 1293 - 1298
MUZYCZKA ET AL., CURR. TOPICS IN MICRO AND IMMUNOL., vol. 158, 1992, pages 97 - 129
MINGOZZIHIGH, NATURE REVIEWS GENETICS, vol. 12, 2011, pages 341 - 355
DEYLERUSSELL, CURR OPIN MOL THER, vol. 11, no. 4, August 2009 (2009-08-01), pages 442 - 447
ASOKAN ET AL., MOL THER, vol. 20, no. 4, April 2012 (2012-04-01), pages 699 - 708
PANG ET AL., VISION RESEARCH, vol. 48, no. 3, 2008, pages 377 - 385
KHANI ET AL., INVEST OPHTHALMOL VIS SCI., vol. 48, no. 9, September 2007 (2007-09-01), pages 3954 - 61
ALLOCCA ET AL., J. VIROL., vol. 81, no. 20, 2007, pages 11372 - 11380
THOMPSON ET AL., CELL, vol. 56, 1989, pages 313 - 321
LO, MOL. CELL. BIOL., vol. 3, 1983, pages 1803 - 1814
WILMUT ET AL., NATURE, vol. 385, 1997, pages 810 - 813
WAKAYAMA ET AL., NATURE, vol. 394, 1998, pages 369 - 374
CIBELLI ET AL., SCIENCE, vol. 280, 1998, pages 1256 - 1258
KALINEC ET AL., CELL BIOLOGY INTERNATIONAL, vol. 23, no. 3, 1999, pages 175 - 184
KELLEY ET AL., DEVELOPMENT, vol. 119, no. 4, 1993, pages 1041 - 1053
ZOU ET AL., HUM MOL GENET, vol. 24, no. 24, 15 December 2015 (2015-12-15), pages 6944 - 57
LU, B. ET AL.: "Cell transplantation to arrest early changes in an ush2a animal model", INVEST OPHTHALMOL VIS SCI, vol. 51, no. 4, 2010, pages 2269 - 76
NAT. BIOTECH., vol. 33, 2015, pages 187 - 197
NAT. METHODS, vol. 12, 2015, pages 237 - 243
HARTONG, D.T.E.L. BERSONT.P. DRYJA: "Retinitis pigmentosa", LANCET, vol. 368, no. 9549, 2006, pages 1795 - 809, XP025093439, DOI: 10.1016/S0140-6736(06)69740-7
SAIHAN, Z. ET AL.: "Update on Usher syndrome", CURR OPIN NEUROL, vol. 22, no. 1, 2009, pages 19 - 27
LENTZ, J.B. KEATS ET AL.: "Usher Syndrome Type II, in GeneReviews®", 1993, UNIVERSITY OF WASHINGTON
SANDBERG, M.A. ET AL.: "Disease course in patients with autosomal recessive retinitis pigmentosa due to the USH2A gene", INVEST OPHTHALMOL VIS SCI, vol. 49, no. 12, 2008, pages 5532 - 9
MAGUIRE, A.M. ET AL.: "Safety and efficacy of gene transfer for Leber's congenital amaurosis", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 358, no. 21, 2008, pages 2240 - 8, XP055027817, DOI: 10.1056/NEJMoa0802315
BAINBRIDGE, J.W. ET AL.: "Effect of gene therapy on visual function in Leber's congenital amaurosis", N.ENGL.J.MED., vol. 358, no. 21, 2008, pages 2231 - 2239, XP002718542, DOI: 10.1056/NEJMoa0802268
CIDECIYAN, A.V. ET AL.: "Human gene therapy for RPE65 isomerase deficiency activates the retinoid cycle of vision but with slow rod kinetics", PROC.NAT.ACAD.SCI., vol. 105, no. 39, 2008, pages 15112 - 7, XP055390820, DOI: 10.1073/pnas.0807027105
MAGUIRE, A.M. ET AL.: "Age-dependent effects of RPE65 gene therapy for Leber's congenital amaurosis: a phase 1 dose-escalation trial", LANCET, vol. 374, no. 9701, 2009, pages 1597 - 605, XP026742607, DOI: 10.1016/S0140-6736(09)61836-5
JACOBSON, S.G. ET AL.: "Gene therapy for leber congenital amaurosis caused by RPE65 mutations: safety and efficacy in 15 children and adults followed up to 3 years", ARCHIVES OF OPHTHALMOLOGY, vol. 130, no. 1, 2012, pages 9 - 24
BENNETT, J. ET AL.: "AAV2 gene therapy readministration in three adults with congenital blindness", SCIENCE TRANSLATIONAL MEDICINE, vol. 4, no. 120, 2012
BOWLES, D.E ET AL.: "Phase 1 gene therapy for Duchenne muscular dystrophy using a translational optimized AAV vector", MOLECULAR THERAPY : THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY, vol. 20, no. 2, 2012, pages 443 - 55, XP055152225, DOI: 10.1038/mt.2011.237
MACLACHLAN, T.K. ET AL.: "Preclinical safety evaluation of AAV2-sFLT01- a gene therapy for age-related macular degeneration", MOLECULAR THERAPY : THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY, vol. 19, no. 2, 2011, pages 326 - 34, XP055136302, DOI: 10.1038/mt.2010.258
NATHWANI, A.C ET AL.: "Adenovirus-associated virus vector-mediated gene transfer in hemophilia B", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 365, no. 25, 2011, pages 2357 - 65, XP055079598, DOI: 10.1056/NEJMoa1108046
SWIECH, L. ET AL.: "In vivo interrogation of gene function in the mammalian brain using CRISPR-Cas9", NAT BIOTECHNOL, vol. 33, no. 1, 2015, pages 102 - 6, XP055176807, DOI: 10.1038/nbt.3055
LIU, X. ET AL.: "Usherin is required for maintenance of retinal photoreceptors and normal development of cochlear hair cells", PROC NATL ACAD SCI U SA, vol. 104, no. 11, 2007, pages 4413 - 8, XP055470637, DOI: 10.1073/pnas.0610950104
ALLER, E ET AL.: "The USH2A c.2299delG mutation: dating its common origin in a Southern European population", EUR J HUM GENET, vol. 18, no. 7, 2010, pages 788 - 93
YAN, D. ET AL.: "Mutation analysis in the long isoform of USH2A in American patients with Usher Syndrome type II", J HUM GENET, vol. 54, no. 12, 2009, pages 732 - 8
KEMALADEWI, D.U.R.D. COHN: "Exon Snipping in Duchenne Muscular Dystrophy", TRENDS MOL MED, vol. 22, no. 3, 2016, pages 187 - 9, XP029434246, DOI: 10.1016/j.molmed.2016.01.007
LONG, C ET AL.: "Postnatal genome editing partially restores dystrophin expression in a mouse model of muscular dystrophy", SCIENCE, vol. 351, no. 6271, 2016, XP055575397, DOI: 10.1126/science.aad5725
NELSON, C.E. ET AL.: "In vivo genome editing improves muscle function in a mouse model of Duchenne muscular dystrophy", SCIENCE, vol. 351, no. 6271, 2016, XP055675964, DOI: 10.1126/science.aad5143
TABEBORDBAR, M. ET AL.: "In vivo gene editing in dystrophic mouse muscle and muscle stem cells", SCIENCE, vol. 351, no. 6271, 2016, pages 407 - 11, XP055676162, DOI: 10.1126/science.aad5177
YOUNG, C.S. ET AL.: "A Single CRISPR-Cas9 Deletion Strategy that Targets the Majority of DMD Patients Restores Dystrophin Function in hiPSC-Derived Muscle Cells", CELL STEM CELL, vol. 18, no. 4, 2016, pages 533 - 40, XP029496784, DOI: 10.1016/j.stem.2016.01.021
KALINEC, F. ET AL.: "Establishment and characterization of conditionally immortalized organ of corti cell lines", CELL BIOL INT, vol. 23, no. 3, 1999, pages 175 - 84, XP071979600, DOI: 10.1006/cbir.1998.0339
MILLAN, J.M. ET AL.: "An update on the genetics of usher syndrome", J OPHTHALMOL, 2011, pages 417217
LENASSI, E. ET AL.: "A detailed clinical and molecular survey of subjects with nonsyndromic USH2A retinopathy reveals an allelic hierarchy of disease-causing variants", EUR J HUM GENET, vol. 23, no. 10, 2015, pages 1318 - 27
SUZUKI, K. ET AL.: "In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration", NATURE, vol. 540, no. 7631, 2016, pages 144 - 149, XP055664441, DOI: 10.1038/nature20565
ZOU, J ET AL.: "Whirlin replacement restores the formation of the USH2 protein complex in whirlin knockout photoreceptors", INVEST OPHTHALMOL VIS SCI, vol. 52, no. 5, 2011, pages 2343 - 51
ZOU, J. ET AL.: "Deletion of PDZD7 disrupts the Usher syndrome type 2 protein complex in cochlear hair cells and causes hearing loss in mice", HUM MOL GENET, vol. 23, no. 9, 2014, pages 2374 - 90, XP055470539, DOI: 10.1093/hmg/ddt629
DAVIS, E.E. ET AL.: "TTC21B contributes both causal and modifying alleles across the ciliopathy spectrum", NAT GENET, vol. 43, no. 3, 2011, pages 189 - 96, XP055080874, DOI: 10.1038/ng.756
ZHANG, Q. ET AL.: "Knockdown of ttc26 disrupts ciliogenesis of the photoreceptor cells and the pronephros in zebrafish", MOL BIOL CELL, vol. 23, no. 16, 2012, pages 3069 - 78
WANG, H. ET AL.: "One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering", CELL, vol. 153, no. 4, 2013, pages 910 - 8, XP028538358, DOI: 10.1016/j.cell.2013.04.025
INUI, M. ET AL.: "Rapid generation of mouse models with defined point mutations by the CRISPR/Cas9 system", SCI REP, vol. 4, 2014, pages 5396
MASHIKO, D. ET AL.: "Feasibility for a large scale mouse mutagenesis by injecting CRISPR/Cas plasmid into zygotes", DEV GROWTH DIFFER, vol. 56, no. 1, 2014, pages 122 - 9, XP055242547, DOI: 10.1111/dgd.12113
LIU, Q.J. ZUOE.A. PIERCE: "The retinitis pigmentosa 1 protein is a photoreceptor microtubule-associated protein", J NEUROSCI, vol. 24, no. 29, 2004, pages 6427 - 36
CHENG, A.W. ET AL.: "Multiplexed activation of endogenous genes by CRISPR-on, an RNA-guided transcriptional activator system", CELL RES, vol. 23, no. 10, 2013, pages 1163 - 71
OUSTEROUT, D.G ET AL.: "Multiplex CRISPR/Cas9-based genome editing for correction of dystrophin mutations that cause Duchenne muscular dystrophy", NAT COMMUN, vol. 6, 2015, pages 6244, XP055575568, DOI: 10.1038/ncomms7244
PLATT, R.J ET AL.: "CRISPR-Cas9 knockin mice for genome editing and cancer modeling.", CELL, vol. 159, no. 2, 2014, pages 440 - 55, XP055523070, DOI: 10.1016/j.cell.2014.09.014
SENIS, E. ET AL.: "CRISPR/Cas9-mediated genome engineering: an adeno-associated viral (AAV) vector toolbox", BIOTECHNOL J, vol. 9, no. 11, 2014, pages 1402 - 12, XP002737562, DOI: 10.1002/biot.201400046
TSAI, S.Q. ET AL.: "GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases", NAT BIOTECHNOL, vol. 33, no. 2, 2015, pages 187 - 97, XP055555627, DOI: 10.1038/nbt.3117
TSAI, S.Q. ET AL.: "Open-source guideseq software for analysis of GUIDE-seq data", NAT BIOTECHNOL, vol. 34, no. 5, 2016, pages 483, XP055555630, DOI: 10.1038/nbt.3534
JELCICK, A.S. ET AL.: "Genetic variations strongly influence phenotypic outcome in the mouse retina", PLOS ONE, vol. 6, no. 7, 2011, pages e21858
LIU, Q.A. SAVELIEVE.A. PIERCE: "The severity of retinal degeneration in Rplh gene-targeted mice is dependent on genetic background", INVEST OPHTHALMOL VIS SCI, vol. 50, no. 4, 2009, pages 1566 - 74
VIJAYAKUMAR SDEPREUX FFJODELKA FM ET AL.: "Rescue of peripheral vestibular function in Usher syndrome mice using a splice-switching antisense oligonucleotide", HUM MOL GENET, vol. 26, no. 18, 15 September 2017 (2017-09-15), pages 3482 - 3494
SLIJKERMAN RWVACHE CDONA M ET AL.: "Antisense Oligonucleotide-based Splice Correction for USH2A-associated Retinal Degeneration Caused by a Frequent Deep-intronic Mutation", MOL THER NUCLEIC ACIDS, vol. 5, no. 10, 1 November 2016 (2016-11-01), pages e381, XP055750198, DOI: 10.1038/mtna.2016.89
ANNEMIEKE AARTSMA-RUS ET AL.: "Theoretic applicability of antisense-mediated exon skipping for Duchenne muscular dystrophy", HUM MUTAT, vol. 30, no. 3, March 2009 (2009-03-01), pages 293 - 9, XP002541505, DOI: 10.1002/humu.20918
BEROUD C ET AL.: "Multi exon skipping leading to an artificial DMD protein lacking amino acids from exons 45 through 55 could rescue up to 63% of patients with Duchenne muscular dystrophy", HUM MUTAT, vol. 28, no. 2, February 2007 (2007-02-01), pages 196 - 202
See also references of EP 3768303A4
Attorney, Agent or Firm:
DEYOUNG, Janice Kugler (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A nucleic acid comprising sequences encoding a Cas9 protein, and a first gRNA, and a second gRNA, wherein the first and second gRNAs are targeted to sequences flanking exon 13 of an usherin (USH2A) gene of the subject, preferably wherein the target sequence of the first gRNA is in the 3’ 1500 base pairs (bp) of intron 12, and the target sequence of the second gRNA is in the 5’ l500bp of intron 13 of the USH2A gene.

2. The nucleic acid of claim 1, wherein the first gRNA comprises a target sequence shown in Table 1 or 6a, and/or wherein the second gRNA comprises a target sequence shown in Table 2 or 6b.

3. A nucleic acid comprising sequences encoding a Cas9 protein, and a gRNA

targeted to a splice acceptor site for exon 13 of an USH2A gene of the subject.

4. The nucleic acid of claim 3, wherein the gRNA comprises a target sequence

shown in Table 3.

5. The nucleic acid of any of the preceding claims, wherein the nucleic acid encodes S. aureus Cas9.

6. The nucleic acid of claim 5, wherein the Cas9 comprises a nuclear localization signal, optionally a C-terminal nuclear localization signal and/or an N-terminal nuclear localization signal; and/or wherein the sequences encoding Cas9 comprises a polyadenylation signal.

7. The nucleic acid of any of claims 1-4, wherein the gRNA is a unimolecular S. aureus gRNA comprising SEQ ID NO:7 or SEQ ID NO: 8 of WO 2018/026976, or the corresponding two-part modular S. aureus gRNA, wherein the crRNA component comprises the underlined section and the tracrRNA component comprises the double underlined section of SEQ ID NO:7 or SEQ ID NO:8 of WO 2018/026976.

8. The nucleic acid of claims 1 to 4, which comprises a viral delivery vector.

9. The nucleic acid of claim 8, wherein the viral delivery vector comprises a promoter for Cas9, preferably a CMV, EFS, or hGRKl promoter.

10. The nucleic acid of claim 8, wherein the viral delivery vector comprises an adeno- associated virus (AAV) vector.

11. The nucleic acid of claims 8-10, which comprises:

(i) a first guide RNA comprising a targeting domain sequence selected from the group listed in Table 1 or 6a and a second guide RNA comprising a targeting domain sequence selected from the group listed in Table 2 or 6b, or a single guide RNA comprising a targeting domain sequence selected from the group listed in Table 3;

(ii) a first and a second inverted terminal repeat sequence (ITR); and

(iii) a promoter for driving expression of the Cas9 selected from the group consisting of a CMV, an EFS, or an hGRKl promoter.

12. The nucleic acid of any of claims 1 to 11, for use in therapy.

13. The nucleic acid of any of claims 1 to 11, for use in preparation of a medicament.

14. The nucleic acid of any of claims 1 to 11, for use in a method of treating a subject who has a condition associated with a mutation in exon 13 of USH2A gene.

15. The nucleic acid for the use of claim 13, wherein the condition is Usher Syndrome type 2 or autosomal recessive retinitis pigmentosa (arRP).

16. The nucleic acid for the use of claim 13 or 14, wherein the AAV vector is

delivered to a retina of a subject by injection, such as by subretinal injection, or is delivered to the inner ear of a subject by injection, preferably through the round window.

17. A transgenic non-human mammal, preferably a mouse, wherein the genome of the mouse comprises a mouse USH2A gene lacking exon 12 or a mutation in a splice acceptor site for exon 12 of the USH2A gene, and wherein the cells of the mouse express an usherin protein lacking exon 12.

18. A transgenic non-human mammal, preferably a mouse, wherein the genome of the mouse comprises a human USH2A gene lacking exon 13 or a mutation in a splice acceptor site for exon 13 of the USH2A gene, and wherein the cells of the mouse express an usherin protein lacking exon 13.

19. A cell, tissue, or organ obtained from the transgenic non-human mammal of claim 17 or 18.

20. An isolated cell, wherein the genome of the cell comprises a human USH2A gene lacking exon 13 or a mutation in a splice acceptor site for exon 13 of the USH2A gene, and wherein the cells express a human usherin protein lacking exon 13, and wherein the cells do not express a functional mouse usherin protein.

21. The isolated cell of claim 20, wherein the cell is a cultured mouse cochlear cell.

22. The isolated cell of claim 20, wherein the cultured mouse cochlear cell is an Oc- Kl cell.

23. An isolated human usherin protein lacking exon 13, preferably comprising SEQ ID NO:2.

24. An isolated nucleic acid encoding the human usherin protein of claim 23.

25. A method of altering the genome of a cell, the method comprising using CRISPR editing to form a first double strand break within intron 12 of the human USH2A gene and a second double strand within intron 13 of the human USH2A gene.

26. The method of claim 25, wherein the cell is a cell of the eye or inner ear of a mammal.

27. The method of claim 25, wherein the first double strand break is generated using a gRNA comprising a sequence selected from Table 1 and the second double strand break is generated using a gRNA comprising a sequence selected from Table 2.

28. The method of claim 25, wherein the first double strand break is formed between nucleotides 216,232,137 to 216,246,584 of chromosome 1 and the second double strand break is formed between nucleotides 216,247,227 and 216,250,902 of chromosome 1.

29. The method of claims 25-28, wherein the first and second double strand breaks are repaired by NHEJ in a manner that results in removal of exon 13 of an USH2A gene on chromosome 1.

30. The method of claims 25-28, wherein the step of forming the first strand break comprises contacting the cell with a first gRNA which comprises a targeting domain sequence selected from Table 1 and the step of forming the second strand break comprises contacting the cell with a second gRNA which comprises a targeting domain sequence selected from Table 2.

31. The method of claims 25-30, wherein the first and/or second gRNA is configured to form a complex with a Cas9 molecule.

32. A method of altering a genome of a cell, comprising using CRISPR to form a first double strand break between nucleotides 216,248,383 to 216,248,639 of chromosome 1 and to form a second double strand break between nucleotides 216,245,292 and 216,246,542 of chromosome 1, wherein the first and second double strand breaks are repaired by NHEJ in a manner that results in the removal of exon 13 of the USH2A gene on chromosome 1.

33. The method of claim 32, wherein the step of forming the first strand break

comprises contacting the cell with a first gRNA selected from Table 6a and the step of forming the second strand break comprises contacting the cell with a second gRNA selected from Table 6b.

34. The method of claim 33, wherein the first and second gRNAs selected from

Tables 6a and 6b are configured to form a first and second complex with a Cas9 molecule, respectively.

35. The method of claims 25-34, wherein the cell is from a subject suffering from Usher syndrome type 2A.

36. The method of claims 25-35, wherein the cell is a retinal cell or a photoreceptor cell.

37. The method of claim 36, wherein the photoreceptor cell is a cone photoreceptor cell or a cone cell, a rod photoreceptor cell or a rod cell or a macular cone photoreceptor cell.

38. A method of altering a cell, comprising contacting the cell with a recombinant viral particle comprising:

a nucleotide sequence encoding a first gRNA molecule comprising a targeting domain sequence selected from Table 1;

a nucleotide sequence encoding a second gRNA molecule comprising a targeting domain sequence selected from Table 2; and

a nucleotide sequence encoding a Cas9 molecule;

wherein said viral particle is capable of delivery to a non-dividing cell, and wherein said contacting results in removal of exon 13 of human USH2A gene.

39. The method of claim 38, wherein the viral particle is an adeno-associated virus (AAV) viral particle.

40. The nucleic acid of any of claims 1-11, or the method of claims 25-39, wherein the first gRNA and the second gRNA are Inl2_307 and Ih13_318; Inl2_307 and Inl3_322; Inl2_307 and Inl3_323; Inl2_307 and Ih13_327; Ih12_307 and Ih13_328; Ih12_321 and Ih13_318; Ih12_321 and Ih13_322; Ih12_321 and Ih13_323; Ih12_321 and Ih13_327; Ih12_321 and Ih13_328.

Description:
CRISPR/Cas9-Mediated Exon-Skipping Approach for USH2A- Associated Usher Syndrome

CLAIM OF PRIORITY

This application claims the benefit of U.S. Provisional Patent Application Serial Nos. 62/647,578, filed on March 23, 2018, and 62/794,402, filed on January 18, 2019. The entire contents of the foregoing are hereby incorporated by reference. TECHNICAL FIELD

Described herein are compositions for use in treating subjects with USH2A- associated retinal and/or cochlear degeneration that result from mutations in exon 13 of the USH2A gene by deletion of exon 13 from the USH2A gene or transcripts, and methods of use thereof, as well as genetically modified animals and cells. BACKGROUND

The USH2A gene encodes the transmembrane protein Usherin. Usherin localizes mainly at the periciliary region of mammalian photoreceptors and at the stereocilia or hair bundle of the inner ear hair cells (see, e.g., Maerker et al, Hum Mol Genet. 2008 Jan 1;17(1):71-86; Liu et al., Proc Natl Acad Sci U S A. 2007 Mar 13; 104(1 l):44l 3-8). The Usherin protein has a large extracellular domain that is proposed to interact with basement membrane collagen IV and fibronectin via laminin domains (see, e.g., Maerker et al., 2008; Reiners et al., Hum Mol Genet. 2005 Dec 15 ; 14(24) : 3933 -43) . Usherin also interacts with other proteins of USH1 and USH2 complex to form Usher networks (Human Molecular Genetics, 26, 1157-1172).

Mutations in USH2A are the most common cause of both Usher syndrome type II and autosomal recessive retinitis pigmentosa (arRP), accounting for approximately 17% of the recessive RP cases [1, 2]. The impairment of both vision and hearing in Usher syndrome results in a reduced ability of the individual to perceive, communicate, and extract vital information from the environment

[3] Longitudinal regression analysis has showed that the disease course for patients with USH2A mutations can be rapidly progressive, particularly with respect to losing visual field and mobility [4] SUMMARY

The c.2299delG mutation in exonl3 of the USH2A gene is a single basepair deletion that results in a frameshift and premature stop codon, truncating the protein at exon 132 and truncates protein causing ciliary defects. Exon 13 encodes amino acids 723-936, which span 4 of 8 Laminin EGF-like domains in the protein. As not all of these domains appear to be necessary for proper protein function, complete removal of exon 13 can be used to correct the disease phenotype by restoring the proper reading frame of the gene. Exons 12 and 14 are in frame with each other so deletion of exon 13 by a dual-cut approach, in which one gRNA directs a double-strand break to intron 12 and a second gRNA directs a double-strand break to intron 13, is hypothesized to lead to direct splicing of exon 12 to exon 14, thus generating an in- frame coding sequence lacking several of the Laminin EGF-like domains.

Alternatively, disrupting the exon 13 splice acceptor site using a single gRNA, would provide similar results. As the protein lacking exon 13 retains functionality, this approach could also be applied to other exon 13 mutations, e.g., as known in the art, e.g., as shown in Table A.

Provided herein are nucleic acids comprising sequences encoding a Cas9 protein, and a first gRNA, and a second gRNA, wherein the first and second gRNAs are targeted to sequences flanking exon 13 of an usherin ( USH2A ) gene of the subject, preferably wherein the target sequence of the first gRNA is in the 3’ 1500 base pairs (bp) of intron 12, and the target sequence of the second gRNA is in the 5’ l500bp of intron 13 of the USH2A gene. In some embodiments, the first gRNA comprises a target sequence shown in Table 1 or 6A, and/or wherein the second gRNA comprises a target sequence shown in Table 2 or 6B. In some embodiments, the gRNAs comprise Inl2_307 with Ih13_318; Inl2_307 with Inl3_322; Inl2_307 with

Inl3_323; Inl2_307 with Ih13_327; Ih12_307 with Ih13_328; Ih12_321 with Ihΐ 3 318; Ih12_321 with Ih13_322; Ih12_321 with Ih13_323; Ih12_321 with Ih13_327; or Ih12_321 with Ih13_328.

Also provided herein are nucleic acids comprising sequences encoding a Cas9 protein, and a gRNA targeted to a splice acceptor site for exon 13 of an USH2A gene of the subject. In humans, the splice acceptor site is TAGG where TAG is in intron 12 and G is in exon 13. In some embodiments, the gRNA comprises a target sequence shown in Table 3. In some embodiments, the nucleic acid encodes S. aureus Cas9, preferably wherein the nucleic acid comprises a Cas9 coding sequence according to SEQ ID NO: 10 or encodes a Cas9 comprising the sequence of SEQ ID NO: 11 of WO

2018/026976.

In some embodiments, the sequences encoding Cas9 comprises a nuclear localization signal, e.g., a C-terminal nuclear localization signal and/or an N-terminal nuclear localization signal; and/or wherein the sequences encoding Cas9 comprises a polyadenylation signal.

In some embodiments, the gRNA is a unimolecular S. aureus gRNA comprising SEQ ID NO:7 or SEQ ID NO: 8 of WO 2018/026976, or the

corresponding two-part modular S. aureus gRNA, wherein the crRNA component comprises the underlined section and the tracrRNA component comprises the double underlined section of SEQ ID NO:7 or SEQ ID NO:8 of WO 2018/026976.

In some embodiments, the nucleic acid comprises a viral delivery vector. In some embodiments, the viral delivery vector comprises a promoter for Cas9, preferably a CMV, EFS, or hGRKl promoter. In some embodiments, the viral delivery vector comprises an adeno-associated virus (AAV) vector.

In some embodiments, the nucleic acid comprises: (i) a first guide RNA comprising a targeting domain sequence selected from the group listed in Table 1 or 6a and a second guide RNA comprising a targeting domain sequence selected from the group listed in Table 2 or 6b, or a single guide RNA comprising a targeting domain sequence selected from the group listed in Table 3; (ii) a first and a second inverted terminal repeat sequence (ITR); and (iii) a promoter for driving expression of the Cas9 selected from the group consisting of a CMV, an EFS, or an hGRKl promoter. In some embodiments, the gRNAs comprise Inl2_307 with Inl3_3l8; Inl2_307 with Inl3_322; Inl2_307 with Inl3_323; Inl2_307 with Ih13_327;

Ih12_307 with Ih13_328; Ih12_321 with Ih13_318; Ih12_321 with Ih13_322;

Ih12_321 with Ih13_323; Ih12_321 with Ih13_327; or Ih12_321 with Ih13_328.

Also provided are the nucleic acids described herein for use in therapy, for use in preparation of a medicament; and/or for use in a method of treating a subject who has a condition associated with a mutation in exon 13 of USH2A gene.

In some embodiments, the condition is Usher Syndrome type 2 or autosomal recessive retinitis pigmentosa (arRP). In some embodiments, the AAV vector is delivered to a retina of a subject by injection, such as by subretinal injection, or is delivered to the inner ear of a subject by injection, e.g., through the round window.

Also provided herein are transgenic non-human mammal, e.g., a mouse, wherein the genome of the mouse comprises a mouse USH2A gene lacking exon 12 or a mutation in a splice acceptor site for exon 12 of the USH2A gene, and wherein the cells of the mouse express an usherin protein lacking exon 12; and/or wherein the genome of the mouse comprises a human USH2A gene lacking exon 13 or a mutation in a splice acceptor site for exon 13 of the USH2A gene, and wherein the cells of the mouse express an usherin protein lacking exon 13.

Also provided herein are cells, tissue, or organ (e.g., an eye or cochlea) obtained from the transgenic non-human mammals described herein.

Also provided herein are isolated cells, wherein the genome of the cells comprises a human USH2A gene lacking exon 13 or a mutation in a splice acceptor site for exon 13 of the USH2A gene, and wherein the cells express a human usherin protein lacking exon 13, and wherein the cells do not express a functional mouse usherin protein.

In some embodiments, the cell is a cultured mouse cochlear cell. In some embodiments, the cultured mouse cochlear cell is an Oc-Kl cell.

In addition, provided herein is an isolated human usherin protein lacking exon 13, e.g., comprising SEQ ID NO:2, and a nucleic acid encoding the isolated human usherin protein.

In some embodiments, a CRISPR-Cas9 method of altering a cell described herein comprises forming a first double strand break within intron 12 of the human USH2A gene and a forming a second double strand within intron 13 of the human USH2A gene. In various embodiments described herein, the first double strand break is generated using a gRNA targeting domain sequence selected from Table 1 and the second double strand break is generated using a gRNA targeting domain sequence selected from Table 2.

In some embodiments described herein, a CRISPR-Cas9 method of altering a cell is described, which method comprises the step of forming a first double strand break between nucleotides 216,232,137 to 216,246,584 of chromosome 1 and the step of forming a second double strand break between nucleotides 216,247,227 and 216,250,902 of chromosome 1, wherein the first and second double strand breaks are repaired by NHEJ in a manner that results in the removal of exon 13 of the USH2A gene on chromosome 1. In some embodiments, the step of forming the first strand break comprises contacting the cell with a gRNA which comprises a targeting domain sequence selected from Table 1 and the step of forming the second strand break comprises contacting the cell with a gRNA which comprises a targeting domain sequence selected from Table 2. In various embodiments, a gRNA is configured to form a complex with a Cas9 molecule.

In further embodiments, a CRISPR-Cas9 method of altering a cell is described, which method comprises the step of forming a first double strand break between nucleotides 216,248,383 to 216,248,639 of chromosome 1 and the step of forming a second double strand break between nucleotides 216,245,292 and

216,246,542 of chromosome 1, wherein the first and second double strand breaks are repaired by NHEJ in a manner that results in the removal of exon 13 of the USH2A gene on chromosome 1. In some embodiments, the step of forming the first strand break comprises contacting the cell with a gRNA selected from Table 6a and the step of forming the second strand break comprises contacting the cell with a gRNA selected from Table 6b. In various embodiments, the gRNAs selected from Tables 6a and 6b are configured to form a first and second complex with a Cas9 molecule, respectively.

In various embodiments described herein, the cell is from a subject suffering from Usher syndrome type 2A. In some embodiments, the cell is a retinal cell or a photoreceptor cell. In some embodiments, the photoreceptor cell is a cone photoreceptor cell or a cone cell, a rod photoreceptor cell or a rod cell or a macular cone photoreceptor cell.

In some embodiments, a method of altering a cell comprises contacting the ceil with a recombinant viral particle comprising:

(a) a nucleotide sequence encoding a first gRNA molecule comprising a targeting domain sequence selected from Table 1;

(b) a nucleotide sequence encoding a second gRN A molecule comprising a

targeting domain sequence selected from Table 2; and

(c) a nucleotide sequence encoding a Cas9 molecule;

wherein said viral particle is capable of delivery to a non-dividing cell, and wherein said contacting results in removal of exon 13 of human USH2A gene.

In some embodiments, the viral particle is an AAV viral particle. 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 invention belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.

Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.

DESCRIPTION OF DRAWINGS

FIGs. 1A-1D: Expression of Ush2a in OC-kl cells. 1A. RT-PCR assay shows the expression of Ush2a and other cilia proteins, including Whm, Vlgrl, Sept7, Sept2 and Ehdl, in the OC-kl cells. IB. Three experimental replicates of RT-PCR for Ush2a gene in OC-kl cells. 1C. Ush2a protein detected by anit-Ush2a antibody in OC-l cells in green. Primary cilia in the OC-kl cells were labeled with acetylated alpha tubulin in red. ID. Merged images of 1C, showing that Ush2a protein is localized at the base of primary cilia.

FIGs. 2A-2D: Generation of null Ush2a cell model in OC-kl cells. 2A.

Schematic illustration of sgRNA for targeting the exon 5 of Ush2a gene on mouse chromosome 1 (SEQ ID NO:3). 2B. The percentage of in-frame (15%) and out-of- flame (85%) indels introduced by NHEJ in cells transfected with U6-sgRNA and CAG-SpCas9-P2A-GFP plasmids. 2C. Examples of T7E1 assay for individual cell clones after transfection with Cas9/sgRNA. 2D. Allele sequences at the target region of Ush2a gene in clone 4, 17 and J (SEQ ID NOs:4-ll).

FIGs. 3A-3D: Ciliogenesis in wild-type and Ush2a null OC-K1 cell lines. Immunostaining for ciliary markers for clone 4 (3 A), 17 (3B) and J (3C). Ac-Tub appears in red stains the ciliary axoneme, ush2a appears in green that co-localizes at the base of cilia. The length of cilia in clone 17 and clone J is significantly shorter (3D)

FIGs. 4A-E: Expression of USH2A-AExl3 cDNA rescues the ciliogenesis in Ush2a null cells. Ush2a null cells from clone J transfected with mouse full-length Ush2a cDNA (4B), human wild-type full length USH2A cDNA (4C) and human USH2A- \Ex 13 cDNAs (4D). Non-transfected cell used as control (4A). Cilia were labeled with Ac-tubulin in red and the expression of Ush2a is detected by C-term anti- Ush2a antibody in green. 4E. The cilia length was significantly increased in cells transfected with wild-type and human USH2A- \Ex 13 cDNAs as compared to the knockout.

FIGs. 5A-D: Generation of Ush2a-AExl2 mouse lines using

CRISPR/Cas9. 5A. Schematic illustration of approach for knocking out exon 12 in mouse Ush2a gene. 5B. sgRNA sequences to target intron 11 (11 A (SEQ ID NO: 12), 11B (SEQ ID NO: 13) and 11C (SEQ ID NO: 14)) and intron l2 (l2A (SEQ ID

NO: 15), 12C (SEQ ID NO: 16) and 12D (SEQ ID NO: 17)). 5C. Cleavage efficiencies of selected sgRNAs on a DNA template derived from Ush2a genomic region surrounding exon 12. 5D. Schematic illustration of deletion of exonl2 and surrounding intronic sequence in 9 Ush2a-AExl2 found mice.

FIGs. 5E-K: Phenotypic rescue by Ush2a- \Exl2. 5E, both the w ild type and exon l2-skippped Ush2a proteins were localized at the transition zone of photoreceptor sensory cilia in DEc12/DEc12, \Ex 12/ko. and wt mice. 5F, Cochleas isolated from P3 AExl2/AExl2 and wt mice and stained for phalloidin (top panel), and Ush2a, FM1-43, and phalloidin. 5(7, Inner and outer hair cell structures in

AExl2/AExl2, AExl2/ko, and wt mice on staining with Ush2a, as compared with the knockout. 5H, Auditory brain stem recordings showing restoration of the ABR response in the Ush2a ko/ko mice by the Ush2a-Exl2 allele as compared to the knockout. 51, Phalloidin staining showed disrupted bundles in the ko/ko mice, but not in AExl2/AExl2, AExl2/ko, or wt mice. 5J, Arrows indicate the accumulation of GFAP protein in ko retina but not in AExl2/AExl2, AExl2/ko, or wt retina 5K,

Normal cone opsin localization in AExl2/AExl2 and AEx 12/ko mice, as compared to ko/ko mice.

FIG. 6. Schematic illustration of two approaches to USH2A gene editing.

Exons 12 and 14 are in frame with each other, so the goal of both approaches is to generate USH2A mRNA without exon 13 and thereby restore reading frame. The skipping of exon 13 results in the formation of a novel domain composed of the n- terminus of Laminin EGF-like (LE) domain repeat 4 and the C-terminus of LE repeat 8 FIG. 7. The splice acceptor sequence is well-conserved across humans (SEQ ID NO: 18), African Green monkeys (SEQ ID NO: 19), and cynomolgus monkeys (SEQ ID NO: 19). The target site of SaCas9-KKH sgRNAs for disrupting the splicing acceptor of exon 13 in human USH2A gene.

FIG. 8 schematically depicts a gRNA used in certain embodiments of the disclosure (SEQ ID NO:2l).

FIG. 9. Schematic illustration of an exemplary AAV vector to deliver SaCas9 and gRNAs. U6, human U6 promoter; hGRKl, human photoreceptor-specific rhodopsin kinase promoter; SV40 SD/SA, SV40 intron sequence; NLS, nuclear localization signal; pA, polyadenylation signal.

FIGs. 10A-B. Screening of top dual gRNA pairs and single splice acceptor gRNA. 10A: Editing of top 16 gRNA pairs for complete removal of USH2A exon 13 as determined by a ddPCR assay. 10B: Total editing of identified single gRNA that fall near the USH2A exon 13 splice acceptor.

FIGs. 11A-D. Editing of USH2A human cells and measurement of USH2A delta exon 13 transcripts. 11A: Total editing of CRL-5923 cells with single gRNA targeting the exon 13 splice acceptor. 11B: Delta 13 USH2 A transcripts compared to WT USH2A after editing with single gRNA as shown in 10A. 11C: Percent deletion of exon 13 after editing of CRL-5923 cells with the dual gRNA approach as determined by ddPCR. 11D. Delta 13 USH2 A transcripts as a fraction of WT USH2A transcripts after editing with dual gRNAs as shown in 11C.

FIG. 12. Percent expression of delta 13 USH2A transcripts in human retinal explants after editing with dual guide approach delivered as an AAV5.

DETAILED DESCRIPTION

Despite the success of clinical and pre-clinical studies of AAV mediated gene augmentation therapy for multiple genetic types of inherited retinal degeneration [5- 13], developing gene therapy for the USH2A form of arRP has been challenging, because the large size of the USH2A coding sequence (CDSl5602bp, 5202aa) far exceeds the packaging capacity of commonly used AAV viral delivery vectors. The present methods overcome these translational barriers by using a Cas9 gene editing approach for USH2A associated arRP [14, 15] The CRISPR/Cas system is capable of maintaining the edited gene under its endogenous regulatory elements by directly altering the genomic DNA, thereby avoiding ectopic expression and abnormal gene production that may occur with conventional viral-mediated gene augmentation therapies [14, 15]

The usherin protein encoded by USH2A (GenBank Acc No. NC_00000l .11, Reference GRCh38.p7 Primary Assembly, Range 215622894-216423396, complement; SEQ ID NO: 1) is a transmembrance protein anchored in the

photoreceptor plasma membrane (van Wijk, E., et al, Am J Hum Genet, 2004. 74(4): p. 738-44; Grati, M., et al, J Neurosci, 2012. 32(41): p. 14288-93). Its extracellular portion, which accounts for over 96% of the length of the protein and projects into the periciliary matrix, is thought to have an important structural and a possible signaling role for the long-term maintenance of photoreceptors (van Wijk, E., et al., Am J Hum Genet, 2004. 74(4): p. 738-44; Grati, M., et al., J Neurosci, 2012. 32(41): p. 14288- 93). Two isoforms of USH2A have been described. Isoform b (GenBank Acc. No. NM_206933.2 (transcript) and NP_9968l6.2 (protein)) is most abundantly expressed in retina and is used as the canonical, standard sequence in the literature and in this application. Usherin is a protein with a high degree of homologous domain structures (Liu, X., et al, Proc Natl Acad Sci U S A, 2007. 104(11): p. 4413-8). Intracellularly, a PDZ domain has been identified to bind whirlin, whereas extracellularly, several domains are present and in most cases in a repetitive fashion, includinglO Laminin EGF-like (LE) domains and 35 Fibronectin type 3 (FN3) domains. These repetitive domains comprise over 78% of the protein structure combined. The most common mutation c.2299delG, p.Glu767fs in USH2A gene, which causes approximately 15%- 30% of USH2A cases is USA [19, 20], is located in exon 13 that encodes LE domain 5 (aa 747-794) (Liu, X., et al, Proc Natl Acad Sci U S A, 2007. 104(11): p. 4413-8). Given the high degree of repetitive regions in usherin, it was hypothesized that an usherin protein that lacks one or more of the repetitive domains would retain partial or complete structural integrity and function, such that the abbreviated USH2A can serve as a therapeutic strategy for Usher syndrome type II and autosomal recessive retinitis pigmentosa (arRP) by skipping the mutant exon in USH2A gene.

As shown herein, Ush2a lacking exon 12 and with exons 11 and 13 fused in frame is expressed and localized correctly in the mouse retina and cochlea. When the Ush2a-AExl2 allele was expressed on an Ush2a null background, the Ush2a-AExl2 protein appeared to rescue the impaired hair cell structure and auditory function as shown by ABR, as compared to mice and also showed early signs of at least partial rescue of retinal phenotype. Without wishing to be bound by theory, this data supports the use of the present compositions and methods to restore sight and/or hearing, e.g., at least partially restore sight and/or hearing, in a subject who has Usher syndrome, e.g., associated with a mutation in exon 13 of USH2A gene. Thus a CRISPR/Cas9-based exon-skipping gene editing strategy to restore the reading frame of USH2A by deleting exon 13 holds therapeutic potential for the treatment of USH2A patients.

In one embodiment, an Ush2A nucleic acid molecule includes a nucleotide sequence that is at least about 85% or more identical to the entire length of SEQ ID NO: l. In some embodiments, the nucleotide sequence is at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 1.

To determine the percent identity of two amino acid sequences, or of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). The length of a reference sequence aligned for comparison purposes is at least 80% of the length of the reference sequence, and in some embodiments is at least 90% or 100%. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. In another embodiment, the percent identity of two amino acid sequences can be assessed as a function of the conservation of amino acid residues within the same family of amino acids (e.g., positive charge, negative charge, polar and uncharged, hydrophobic) at corresponding positions in both amino acid sequences (e.g., the presence of an alanine residue in place of a valine residue at a specific position in both sequences shows a high level of conservation, but the presence of an arginine residue in place of an aspartate residue at a specific position in both sequences shows a low level of conservation).

For purposes of the present invention, the comparison of sequences and determination of percent identity between two sequences can be accomplished using a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.

Methods of Treatment

CRISPR/Cas-based exon-skipping has been successfully used for restoring the expression of functional dystrophin and dystrophic muscle function in the Duchene muscular dystrophy mouse model [21-25] The methods described herein include methods for the treatment of disorders associated with mutations in exon 13 of the USH2A gene. Exemplary mutations, including 12 nonsense or frameshift mutations and 7 missense mutations on exon 13 (LOVD database), as shown in Table A, such as the most common missense mutation c.2276G>T.

Table A. Reported mutations on exon 13 of USH2A gene

In some embodiments, the disorder is Usher syndrome, e.g., type 2 Usher syndrome. Subjects with type 2 Usher syndrome typically have moderate to severe hearing loss at birth, and vision that becomes progressively impaired starting in adolescence. In some embodiments, the disorder is autosomal recessive retinitis pigmentosa (arRP). Generally, the methods include administering a therapeutically effective amount of a genome editing system as described herein, to a subject who is in need of, or who has been determined to be in need of, such treatment. The term "genome editing system" refers to any system having RNA-guided DNA editing activity. Genome editing systems of the present disclosure include at least two components adapted from naturally occurring CRISPR systems: a gRNA and an RNA-guided nuclease. These two components form a complex that is capable of associating with a specific nucleic acid sequence in a cell and editing the DNA in or around that nucleic acid sequence, for example by making one or more of a single- strand break (an SSB or nick), a double-strand break (a DSB) and/or a base substitution. See, e.g., WO2018/026976 for a full description of genome editing systems.

As used in this context, to“treat” means to ameliorate at least one symptom of the disorder associated with mutations in exon 13 of the USH2A gene. Often, these mutations result in hearing loss and/or loss of sight; thus, a treatment comprising administration of a therapeutic gene editing system as described herein can result in a reduction in hearing impairment and/or visual impairment; a reduction in the rate of progression of hearing loss and/or vision loss; and/or a return or approach to normal hearing and/or vision. Hearing and vision can be tested using known methods, e.g., electroretinogram, optical coherence tomography, videonystagmography, and audiology testing. The methods can be used to treat any subject (e.g., a mammalian subject, preferably a human subject) who has a mutation in exon 13 of the USH2A gene, e.g., the c.2299delG mutation or c.2276G>T mutation, e.g., in one or both alleles of USH2A. As used herein, an“allele” is one of a pair or series of genetic variants of a polymorphism (also referred to as a mutation) at a specific genomic location. As used herein,“genotype” refers to the diploid combination of alleles for a given genetic polymorphism. A homozygous subject carries two copies of the same allele and a heterozygous subject carries two different alleles. Methods for identifying subjects with such mutations are known in the art; see, e.g., Yan et al., J Hum Genet. 2009 Dec; 54(12): 732-738; Leroy et al, Exp Eye Res. 2001 May;72(5):503-9; or

Consugar et al, Genet Med. 2015 Apr;l7(4):253-26l. For example, gel

electrophoresis, capillary electrophoresis, size exclusion chromatography, sequencing, and/or arrays can be used to detect the presence or absence of the allele or genotype. Amplification of nucleic acids, where desirable, can be accomplished using methods known in the art, e.g., PCR. In one example, a sample (e.g., a sample comprising genomic DNA), is obtained from a subject. The DNA in the sample is then examined to identify or detect the presence of an allele or genotype as described herein. The allele or genotype can be identified or determined by any method described herein, e.g., by Sanger sequencing or Next Generation Sequencing (NGS). Since the exon 13 is 643bp in size, thus the genotyping of the patients with exon 13 mutations is simple and straight forward using Sanger sequencing or NGS. Other methods can include hybridization of the gene in the genomic DNA, RNA, or cDNAto a nucleic acid probe, e.g., a DNA probe (which includes cDNA and oligonucleotide probes) or an RNA probe. The nucleic acid probe can be designed to specifically or preferentially hybridize with a particular mutation (also referred to as a polymorphic variant).

Other methods of nucleic acid analysis can include direct manual sequencing (Church and Gilbert, Proc. Natl. Acad. Sci. USA 81 : 1991-1995 (1988); Sanger et al, Proc. Natl. Acad. Sci. USA 74:5463-5467 (1977); Beavis et al, U.S. Pat. No.

5,288,644); automated fluorescent sequencing; single-stranded conformation polymorphism assays (SSCP) (Schafer et al, Nat. Biotechnol. 15:33-39 (1995)); clamped denaturing gel electrophoresis (CDGE); two-dimensional gel electrophoresis (2DGE or TDGE); conformational sensitive gel electrophoresis (CSGE); denaturing gradient gel electrophoresis (DGGE) (Sheffield et al, Proc. Natl. Acad. Sci. USA 86:232-236 (1989)); denaturing high performance liquid chromatography (DHPLC, Underhill et al., Genome Res. 7:996-1005 (1997)); infrared matrix-assisted laser desorption/ionization (IR-MALDI) mass spectrometry (WO 99/57318); mobility shift analysis (Orita et al, Proc. Natl. Acad. Sci. USA 86:2766-2770 (1989)); restriction enzyme analysis (Flavell et al, Cell 15:25 (1978); Geever et al, Proc. Natl. Acad. Sci. USA 78:5081 (1981)); quantitative real-time PCR (Raca et al, Genet Test 8(4):387-94 (2004)); heteroduplex analysis; chemical mismatch cleavage (CMC) (Cotton et al., Proc. Natl. Acad. Sci. USA 85:4397-4401 (1985)); RNase protection assays (Myers et al, Science 230: 1242 (1985)); use of polypeptides that recognize nucleotide mismatches, e.g., E. coli mutS protein; allele-specific PCR, and combinations of such methods. See, e.g., Gerber et al, U.S. Patent Publication No. 2004/0014095 which is incorporated herein by reference in its entirety.

In certain aspects, the present disclosure provides AAV vectors encoding CRISPR/Cas9 genome editing systems, and on the use of such vectors to treat USH2A associated disease. Exemplary AAV vector genomes are schematized in FIG. 9, which illustrates certain fixed and variable elements of these vectors: inverted terminal repeats (ITRs), one or two gRNA sequences and promoter sequences to drive their expression, a Cas9 coding sequence and another promoter to drive its expression (an exemplary construct for use in the Method Two described herein would be the same as that illustrated in FIG. 9, but with only 1 gRNA and U6). Each of these elements is discussed in detail herein. Although Fig. 9 shows a single vector used to deliver a Cas9 and two gRNAs, in some embodiments a plurality of vectors are used, e.g., wherein one vector is used to deliver Cas9, and another vector or vectors is used to deliver one or more gRNAs (e.g., one vector for one gRNA, one vector for two gRNAs, or two vectors for each of two gRNAs). RNA-guided nucleases/Cas9

Various RNA-guided nucleases can be used in the present methods, e.g., as described in WO 2018/026976. In some embodiments, the RNA-guided nuclease used in the present methods and compositions is a S. aureus Cas9 or a S. pyogenes cas9. In some embodiments of this disclosure a Cas9 sequence is modified to include two nuclear localization sequences (NLSs) (e.g., PKKKRKV (SEQ ID NO:22) at the C- and N-termini of the Cas9 protein, and a mini-polyadenylation signal (or Poly -A sequence). An exemplary NLS is SV40 large T antigen NLS (PKKKRRV (SEQ ID NO:23)) and nucleoplasmin NLS (KRPAATKKAGQAKKKK (SEQ ID NO:24)). Other NLSs are known in the art; see, e.g., Cokol et al, EMBO Rep. 2000 Nov 15; l(5):4l 1—415; Freitas and Cunha, Curr Genomics. 2009 Dec; 10(8): 550-557. An exemplary polyadenylation signal is

T AGC A AT A A AGGAT C GTTT ATTTT C ATT GGAAGC GT GT G TTGGTTTTTTGATCAGGCGCG (SEQ ID NO:25)). Exemplary S. aureus Cas9 sequences (both nucleotide and peptide) are described in Table 4 of WO

2018/026976, e.g., SEQ ID NOs 10 and 11 therein.

Guide RNAs

In some embodiments, the gRNAs used in the present disclosure can be unimolecular or modular, as described below. An exemplary unimolecular S. aureus gRNA is shown in FIG. 8, and exemplary DNA and RNA sequences corresponding to unimolecular S. aureus gRNAs are shown below:

DNA:

[N 16-24] GTTTTAGTACTCTGgaaaCAGAATCTACTAAAACAAGGC AA

AATGCCGTGTTTATCTCGTCAACTTGTTGGCGAGATTTTTT (SEQ ID NO: 26) and

RNA:

[Ni6-24]GUUUUAGUACUCUGgaaaCAGAAUCUACUAAAACAAGGC

AAAAUGCCGUGUUUAUCUCGUCAACUUGUUGGCGAGAUUUUUU (SEQ ID NO: 27).

DNA:

[N 16-24] GTTATAGTACTCTGgaaaCAGAATCTACTATAACAAGGCAA

AATGCCGTGTTTATCTCGTCAACTTGTTGGCGAGATTTTTT (SEQ ID NO: 28) and

RNA:

[Ni6-24]GUUAUAGUACUCUGGaaaCAGAAUCUACUAUAACAAGG

C A AAAuGCC GU GUUU AU CUCGU C A ACUU GUU GGCGAGAUUUUUU (SEQ ID NO:29).

It should be noted that, while figure 8 depicts a targeting domain of 20 nucleotides, the targeting domain can have any suitable length. As indicated by the “N16-24” notation in the sequences above, gRNAs used in the various embodiments of this disclosure preferably include targeting domains of between 16 and 24 (inclusive) bases in length at their 5’ ends, and optionally include a 3’ U6 termination sequence as illustrated.

The gRNA in FIG. 8 is depicted as unimolecular, but in some instances modular guides can be used. In the exemplary unimolecular gRNA sequences above, a 5’ portion corresponding to a crRNA (bold) is connected by a GAAA linker (lower case) to a 3’ portion corresponding to a tracrRNA (double underlined). Skilled artisans will appreciate that two- part modular gRNAs can be used that correspond to the bold and double underlined sections.

Either one of the gRNAs presented above can be used with any of targeting sequences in tables 1-3, and two gRNAs in a pair do not necessarily include the same backbone sequence. Additionally, skilled artisans will appreciate that the exemplary gRNA designs set forth herein can be modified in a variety of ways, which are described below or are known in the art; the incorporation of such modifications is within the scope of this disclosure. Method One: Dual-gRNA Deletion of Exon 13

Described herein are two approaches for treating subjects with mutations in exon 13 of USH2A. The first makes use of dual-gRNAs for deletion of exon 13. Two gRNAs (one in intron 12, one in intron 13) are used in combination to cut out a segment of DNA including exon 13. In addition to deleting this segment, it may also be inverted and reinserted. In the present studies, inversion of the exon was seen as commonly as deletion, and the inverted version was equally functional; without wishing to be bound by theory, the rearrangement may remove the functional splice sites, so the protein still lacks exon 13 and thus corrects the phenotype.

In some embodiments, this approach uses Staphylococcus aureus Cas9 (SaCas9) and corresponding gRNAs. SaCas9 is one of several smaller Cas9 orthologues that are suited for viral delivery (Horvath et al., J Bacteriol 190, 1401 - 1412 (2008); Ran et al, Nature 520, 186-191 (2015); Zhang et al., Mol Cell 50, 488- 503 (2013)). The wild type recognizes a longer NNGRRT PAM that is expected to occur once in every 32 bps of random DNA; or the alternative NNGRRA PAM. Preferably, the 5’ base of each gRNA is a G, and the protospacer is length 20, 21 or 22 nucleotides, and the target sequence falls in the 3’ l500bp of intron 12 or 5’ l500bp of intron 13. The methods can include using two gRNAs, one that targets intron 12, and one that targets intron 13. The genomic coordinates of introns 12 and 13 are provided in the following table.

Tables 1 and 2 provide exemplary sequences for the gRNAs in exons 12 and

13, respectively. Note that in the sequences provided herein, the actual gRNA would have U in place of T.

Table 1. Exemplary Guide RNAs for approach 1: Intron 12

Table 2. Exemplary Guide RNAs for approach 1: Intron 13

In some embodiments of these methods, any of the intron 12 gRNAs in Table 1 can be used with any of the intron 13 gRNAs in Table 2, though certain combinations may be more suitable for certain applications. It should be noted, notwithstanding the use of“first” and“second” as nomenclature for gRNAs, that any guide in a pair, in intron 12 or intron 13, can be placed in either one of the gRNA coding sequence positions illustrated in FIG. 9.

In some embodiments, one of the combinations of gRNAs in the following table is used; each row shows a preferred combination (e.g., Inl2_307 with

Inl2_3l8).

In any of the methods described herein, the engineered CRISPR from

Prevotella and Francisella 1 (Cpfl, also known as Casl2a) nuclease can also be used, e.g., as described in Zetsche et al, Cell 163, 759-771 (2015); Schunder et al, Int J Med Microbiol 303, 51-60 (2013); Makarova et al., Nat Rev Microbiol 13, 722-736 (2015); Fagerlund et al, Genome Biol 16, 251 (2015). Unlike SpCas9, Cpfl/Casl2a requires only a single 42 -nt crRNA, which has 23 nt at its 3’ end that are

complementary to the protospacer of the target DNA sequence (Zetsche et al., 2015). Furthermore, whereas SpCas9 recognizes an NGG PAM sequence that is 3’ of the protospacer, AsCpfl and LbCpl recognize TTTN PAMs that are found 5’ of the protospacer (Id.). In some embodiments, the Casl2a is, e.g., Acidaminococcus sp. BV3L6 Cpfl (AsCpfl, UniProt U2UMQ6.1) or Lachnospiraceae bacterium ND2006 (LbCpfl, UniProt A0A182DWE3.1), with corresponding gRNAs.

Method Two: Single gRNA Deletion of Exon 13 Splice Acceptor

The second approach makes use of a single gRNA for destruction of the exon

13 splice acceptor. Non-homologous end joining (NHEJ)-mediated indels destroy the splice acceptor, thus preventing exon 13 from being spliced into mRNA. Preferably, this method uses Staphylococcus aureus wild type or KKH variant SaCas9 (See Kleinstiver et al, Nat Biotechnol. 2015 Dec; 33(12): 1293-1298; WO 2016/141224) or Casl2a and corresponding gRNAs. Table 3 provides exemplary target sites in the splice acceptor.

Table 3. Guide RNAs for approach 2:

AA V Delivery Systems

The methods include delivery of a CRISPR/Cas9 genome editing system, including a Cas9 nuclease and one or two guide RNAs, to a subject in need thereof. The delivery methods can include, e.g., viral delivery, e.g., preferably using an adeno- associated virus (AAV) vector that comprises sequences encoding the Cas9 and guide RNA(s). Adeno-associated virus is a naturally occurring defective virus that requires another virus, such as an adenovirus or a herpes virus, as a helper virus for efficient replication and a productive life cycle. (For a review see Muzyczka et al, Curr.

Topics in Micro and Immunol.158:97-129 (1992)). AAV vectors efficiently transduce various cell types and can produce long-term expression of transgenes in vivo. AAV vectors have been extensively used for gene augmentation or replacement and have shown therapeutic efficacy in a range of animal models as well as in the clinic; see, e.g., Mingozzi and High, Nature Reviews Genetics 12, 341-355 (2011); Deyle and Russell, Curr Opin Mol Ther. 2009 Aug; 11(4): 442-447; Asokan et al., Mol Ther. 2012 April; 20(4): 699-708. AAV vectors containing as little as 300 base pairs of AAV can be packaged and can produce recombinant protein expression. For example, AAV2, AAV5, AAV2/5, AAV2/8 and AAV2/7 vectors have been used to introduce DNA into photoreceptor cells (see, e.g., Pang et al, Vision Research 2008, 48(3):377-385; Khani et al, Invest Ophthalmol Vis Sci. 2007 Sep;48(9):3954-6l;

Allocca et al, J. Virol. 2007 81(20): 11372-11380). In some embodiments, the AAV vector can include (or include a sequence encoding) an AAV capsid polypeptide described in PCT/US2014/060163; for example, a virus particle comprising an AAV capsid polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, and 17 of PCT/US2014/060163, and a Cas9 sequence and guide RNA sequence as described herein. In some embodiments, the AAV capsid polypeptide is an Anc80 polypeptide, e.g., Anc80L27; Anc80L59;

Anc80L60; Anc80L62; Anc80L65; Anc80L33; Anc80L36; or Anc80L44. In some embodiments, the AAV incorporates inverted terminal repeats (ITRs) derived from the AAV2 serotype. Exemplary left and right ITRs are presented in Table 6 of WO 2018/026976. It should be noted, however, that numerous modified versions of the AAV2 ITRs are used in the field, and the ITR sequences shown below are exemplary and are not intended to be limiting. Modifications of these sequences are known in the art, or will be evident to skilled artisans, and are thus included in the scope of this disclosure.

Cas9 expression is driven by a promoter known in the art. In some embodiments, expression is driven by one of three promoters: cytomegalovirus (CMV), elongation factor- 1 (EFS), or human g-protein receptor coupled kinase- 1 (hGRKl), which is specifically expressed in retinal photoreceptor cells. Nucleotide sequences for each of these promoters are provided in Table 5 of WO 2018/026976. Modifications of these sequences may be possible or desirable in certain applications, and such modifications are within the scope of this disclosure.

Expression of the gRNAs in the AAV vector is driven by a promoter known in the art. In some embodiments, a polymerase III promoter, such as a human U6 promoter. An exemplary U6 promoter sequence is presented below:

AAGGTCGGGCAGGAAGAGGGCCTATTTCCCATGATTCCTTCATATTTGCAT

ATACGATACAAGGCTGTTAGAGAGATAATTAGAATTAATTTGACTGTAAA

CACAAAGATATTAGTACAAAATACGTGACGTAGAAAGTAATAATTTCTTG

GGTAGTTTGCAGTTTTAAAATTATGTTTTAAAATGGACTATCATATGCTTA CCGTAACTTGAAAGTATTTCGATTTCTTGGCTTTATATATCTTGTGGAAAG GACGAAACACC (SEQ ID NO: 188).

In some embodiments, the nucleic acid or AAV vector shares at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or greater sequence identity with one of the nucleic acids or AAV vectors recited above

The AAV genomes described above can be packaged into AAV capsids (for example, AAV5 capsids), which capsids can be included in compositions (such as pharmaceutical compositions) and/or administered to subjects. An exemplary pharmaceutical composition comprising an AAV capsid according to this disclosure can include a pharmaceutically acceptable carrier such as balanced saline solution (BSS) and one or more surfactants (e.g., Tween 20) and/or a thermosensitive or reverse-thermosensitive polymer (e.g., pluronic). Other pharmaceutical formulation elements known in the art may also be suitable for use in the compositions described here.

Compositions comprising AAV vectors according to this disclosure can be administered to subjects by any suitable means, including without limitation injection, for example, subretinal injection or injection through the round window. The concentration of AAV vector within the composition is selected to ensure, among other things, that a sufficient AAV dose is administered to the retina or inner ear of the subject, taking account of dead volume within the injection apparatus and the relatively limited volume that can be safely administered. Suitable doses may include, for example, lxlO 11 viral genomes (vg)/mL, 2xlO n viral genomes (vg)/mL, 3xl0 n viral genomes (vg)/mL, 4xlO n viral genomes (vg)/mL, 5xl0 n viral genomes (vg)/mL, 6xlO n viral genomes (vg)/mL, 7xlO n viral genomes (vg)/mL, 8xl0 n viral genomes (vg)/mL, 9xlO n viral genomes (vg)/mL, lxlO 12 vg/mL, 2xl0 12 viral genomes

(vg)/mL, 3xl0 12 viral genomes (vg)/mL, 4xl0 12 viral genomes (vg)/mL, 5xl0 12 viral genomes (vg)/mL, 6xl0 12 viral genomes (vg)/mL, 7xl0 12 viral genomes (vg)/mL, 8xl0 12 viral genomes (vg)/mL, 9xl0 12 viral genomes (vg)/mL, lxlO 13 vg/mL, 2xl0 13 viral genomes (vg)/mL, 3xl0 13 viral genomes (vg)/mL, 4xl0 13 viral genomes (vg)/mL, 5xl0 13 viral genomes (vg)/mL, 6xl0 13 viral genomes (vg)/mL, 7xl0 13 viral genomes

(vg)/mL, 8xl0 13 viral genomes (vg)/mL, or 9xl0 13 viral genomes (vg)/mL. Any suitable volume of the composition may be delivered to the subretinal or cochlear space. In some instances, the volume is selected to form a bleb in the subretinal space, for example 1 microliter, 10 microliters, 50 microliters, 100 microliters, 150 microliters, 200 microliters, 250 microliters, 300 microliters, etc.

Any region of the retina may be targeted, though the fovea (which extends approximately 1 degree out from the center of the eye) may be preferred in certain instances due to its role in central visual acuity and the relatively high concentration of cone photoreceptors there relative to peripheral regions of the retina. Alternatively or additionally, injections may be targeted to parafoveal regions (extending between approximately 2 and 10 degrees off center), which are characterized by the presence of all three types of retinal photoreceptor cells. In addition, injections into the parafoveal region may be made at comparatively acute angles using needle paths that cross the midline of the retina. For instance, injection paths may extend from the nasal aspect of the sclera near the limbus through the vitreal chamber and into the parafoveal retina on the temporal side, from the temporal aspect of the sclera to the parafoveal retina on the nasal side, from a portion of the sclera located superior to the cornea to an inferior parafoveal position, and/or from an inferior portion of the sclera to a superior parafoveal position. The use of relatively small angles of injection relative to the retinal surface may advantageously reduce or limit the potential for spillover of vector from the bleb into the vitreous body and, consequently, reduce the loss of the vector during delivery. In other cases, the macula (inclusive of the fovea) can be targeted, and in other cases, additional retinal regions can be targeted, or can receive spillover doses.

For delivery to the inner ear, injection to the cochlear duct, which is filled with high potassium endolymph fluid, could provide direct access to hair cells. However, alterations to this delicate fluid environment may disrupt the endocochlear potential, heightening the risk for injection- related toxicity. The perilymph-filled spaces surrounding the cochlear duct, scala tympani and scala vestibuli, can be accessed from the middle ear, either through the oval or round window membrane (RWM). The RWM, which is the only non-bony opening into the inner ear, is relatively easily accessible in many animal models and administration of viral vector using this route is well tolerated. Administration through the oval window or across the tympanic membrane can also be used. See, e.g., W02017100791 and US7206639.

For pre-clinical development purposes, systems, compositions, nucleotides and vectors according to this disclosure can be evaluated ex vivo using a retinal explant system, or in vivo using an animal model such as a mouse, rabbit, pig, nonhuman primate, etc. Retinal explants are optionally maintained on a support matrix, and AAV vectors can be delivered by injection into the space between the photoreceptor layer and the support matrix, to mimic subretinal injection. Tissue for retinal explanation can be obtained from human or animal subjects, for example mouse.

Explants are particularly useful for studying the expression of gRNAs and/or Cas9 following viral transduction, and for studying genome editing over

comparatively short intervals. These models also permit higher throughput than may be possible in animal models, and can be predictive of expression and genome editing in animal models and subjects. Small (mouse, rat) and large animal models (such as rabbit, pig, nonhuman primate) can be used for pharmacological and/or toxicological studies and for testing the systems, nucleotides, vectors and compositions of this disclosure under conditions and at volumes that approximate those that will be used in clinic. Because model systems are selected to recapitulate relevant aspects of human anatomy and/or physiology, the data obtained in these systems will generally (though not necessarily) be predictive of the behavior of AAV vectors and compositions according to this disclosure in human and animal subjects.

Genetically modified Animals and Cells Lacking Exon 13

Also provided herein are non-human genetically modified animals comprising a mutation in exon 13 (or the equivalent exon, for example, exon 12 in the mouse) in the USH2A gene. Such animals are useful as models of disease, e.g., of Usher syndrome or arRP, for studying the function and/or activity of USH2A protein and for identifying and/or evaluating potential therapeutic compounds for treating conditions associated with mutations in exon 13 of the USH2A gene. As used herein, a “genetically modified animal” is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a modified gene. Other examples of genetically modified animals include non-human primates, sheep, dogs, cows, goats, chickens, amphibians, and the like.

The genetically modified animals can have a complete deletion of exon 13, an inversion of exon 13, or a mutation that disrupts the exon 13 splice acceptor site, integrated into or occurring in the genome of the cells of a genetically modified animal (e.g., in one or both alleles of the gene in the genome). In preferred embodiments, the animal has had both endogenous USH2A alleles replaced with a human USH2A gene, or has had part of both endogenous USH2A alleles containing the relevant exon and flanking intronic regions replaced with a human USH2A exon 13 and flanking intronic regions, with a complete deletion of exon 13, an inversion of exon 13, or a mutation that disrupts the exon 13 splice acceptor site.

Methods for making genetically modified animals are known in the art; see, e.g., WO2016049024; WO201604925; WO2017124086; W02018009562; and US9,90l,080. Such techniques include, without limitation, pronuclear microinjection (See, e.g., US4,873,l9l), retrovirus mediated gene transfer into germ lines (Van der Putten et al, Proc. Natl. Acad. Sci. USA, 82:6148-1652 (1985)), gene targeting into embryonic stem cells (Thompson et al, Cell 56:313-321 (1989)), electroporation of embryos (Lo, Mol. Cell. Biol., 3: 1803-1814 (1983)), and in vitro transformation of somatic cells, such as cumulus or mammary cells, followed by nuclear transplantation (Wilmut et al, Nature, 385:810-813 (1997); and Wakayama et al, Nature, 394:369- 374 (1998)); these methods can be modified to use CRISPR as described herein. For example, fetal fibroblasts can be genetically modified using CRISPR as described herein, and then fused with enucleated oocytes. After activation of the oocytes, the eggs are cultured to the blastocyst stage. See, for example, Cibelli et al, Science, 280: 1256-1258 (1998)

A founder animal can be identified based upon the presence of a mutation in exon 13 of USH2A in its genome and/or expression of USH2A mRNA lacking exon 13 in tissues or cells of the animals. A genetically modified founder animal can then be used to breed additional animals carrying the modified gene. Moreover, animals carrying a modified encoding an Ush2A protein lacking exon 13 can further be bred to Ush2a knockout animals.

The invention also includes a population of cells isolated from an animal as described herein, as well as primary or cultured cells, e.g., isolated cells, engineered to include a mutation in exon 13 of human USH2A gene or a deletion of exon 12 in the mouse Ush2a gene. The cells can have a complete deletion of exonl3, an inversion of exon 13, or a mutation that disrupts the exon 13 splice acceptor site, integrated into or occurs in the genome of the cells. The cells can be from any mammal, e.g., a human or non-human mammal, or other animal.

Further provided herein are nucleic acids (e.g., isolated nucleic acids) that comprise or encode an USH2A mRNA that lacks exon 13, e.g., that have a complete deletion of exonl3, an inversion of exon 13, or a mutation that disrupts the exon 13 splice acceptor site, as well as expression and delivery vectors (including viral and non-viral vectors) comprising the nucleic acids, and usherin proteins lacking exon 13. Preferably the sequences are generated using a human USH2A sequence, but they can also be generated from other mammals, including mouse (mRNA: NM_02l408.3; syntenic exon: 12) rat (mRNA: NM_00l 302219.1; syntenic exon 13); chimpanzee (mRNA: XM_0l 6938662.1; syntenic exon: 12); Cynomolgus macaque (macaca fasicularis, mRNA: XM_005540847.2 or XM_005540848.1) and african green monkey (chlorocebus sabeus, mRNA: XM_007988447. l). EXAMPLES

The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.

Example 1. USH2A In vitro Model

This Examiner describes the generation of an USH2A knockout cell line. 1. Characterization of OC-kl cells.

OC-kl cells were selected as a model systemfor this study. OC-kl cells are derived from mouse cochlea (Kalinec et al. (1999) Cell biology international

23(3): l75-l84; Kelley et al. (1993) Development 119(4): 1041-1053). These are tri allelic, expressing Ush2a protein and its interacting proteins such as Whm, Vlgrl in the Usher 2 complex (Figures 1 A, 1B), making these cells particularly appropriate for these studies. The preliminary results illustrated that the OC-kl wild type cells stably expressed the Ush2a protein at the base of primary cilia. (Figures 1C, 1D).

2. Generation of Ush2A knockout cell line in OC-kl cells

CRISPR/Cas9 technology was used to create the Ush2a null cell model in OC- kl cells. Guide RNA 5’-GGAATGCAGTACTGCTGAACGG-3’ (SEQ ID NO:3) for wild-type SpCas9 was designed to target the exon 5 of mouse Ush2a gene (Figure 2A). U6-sgRNA and CAG-SpCas9-P2A-GFP plasmids were CO-transfected into OC- kl cells using lipofectamine 3000. Two days post-transfection, the GFP positive cells were collected by sorting, plated at low density, and grown to mature colonies. Post genomic DNA isolation from these cells, the region spanning the sgRNA target site was PCR amplified and deep sequencing analysis was performed to get insights into the frequency of NHEJ-induced insertion-deletions (Indels) in Ush2a alleles. A cleavage efficiency of -69% was observed with a wide variety of indels with 85% of out-of-frame indels (Figure 2B). A total of 328 clones were screened with T7E1 assay (Figure 2C) and found that the clone 4 obtained from first attempt of transfections resulted in a mixed pattern of indels including in-frame, out-of-frame and uncut alleles (Figure 2D). In order to knockout all three Ush2a alleles in a single clone, clone 4 was re-transfected with the same SpCas9-sgRNA pair and serial dilution of the culture was performed. Of many different single clones analyzed with both T7E1 and NGS analysis, clone J was confirmed to be completely null for Ush2a (two alleles with 7bp and remaining one allele with lbp out of frame deletions, based on ratios of NGS paired reads) as shown in Figure 2D. Other clones, for example clone 17, showed in-frame deletion for one allele and out-of-frame deletion for other two alleles.

3. Characterization of Ush2A knockout cell lines

Ush2a is expressed at the base of cilia. This experiment investigated whether depletion of Ush2a would affect ciliogenesis. Cells were serum starved and stained for Ush2a and acetylated alpha-tubulin, a ciliary marker. As illustrated in figure 3, clone 4 and clone 17 produced cilia as shown by Ace-tubulin staining (Figure 3 A and 3B). They also expressed Ush2a protein at base of cilium, detected by anti-C-term Ush2a antibody (Zou et al, Hum Mol Genet. 2015 Dec 15 ;24(24): 6944-57) . The null clone J failed to produce any detectable cilia (Figure 3C). However, a stunted basal body look-like structure was observed in clone J. Cilia length was further measured from the basal body to the tip of cilia using an acetylated tubulin antibody. (Figure 3D, n=50). Significant shortening of cilia in clone 17 and J, compared to the wild-type cells was observed, indicating that ciliogenesis is hampered in the Ush2a null OC-kl cells.

4. Expression of USH2A-AExl3 cDNA rescued the ciliogenesis in Ush2a null cells

In order to determine whether the Ush2a protein that lacks portion of the reparative Laminin EGF like domain (encoded by exon 13) will retain partial or complete biologic function of USH2A protein, full-length and USH2A- \Ex 13 cDNAs were transfected into the Ush2a null line and their effect on ciliogenesis was evaluated. It was observed that both the mouse and human wild-type full length USH2A cDNAs were able to rescue ciliogenesis (Figure 4B and 4C). In addition, the expression of human USH2A- \Ex 13 was able to increase the length of cilia to 63% of the wild-type cilia (Figure 4D and 4E). These results indicate that the product of the aberrant USH2A- \Ex 13 retains at least partial biological function of USH2A.

Example 2. Targeted Ush2a-AExl2 Mouse Model

To further assess the cell-based findings in Ush2a null cells in vivo, Ush2a- AExl2 mouse lines were generated using CRISPR/Cas9 technology. Pairs of sgRNAs that target the flanking intron 11(11 A, 11B and 11C) and intron 12 (12A, 12C and 12D) (Figure 5A, 5B) were designed. All guides were synthesized and in vitro transcribed and tested for cleavage efficiency using an in-vitro cleavage assay. Figure 3C illustrates the selected sgRNAs are active and cleaved PCR template derived from Ush2a genomic region surrounding exon 12 with efficiencies of 67.3% to 98.2%. All three pairs of sgRNAs were microinjected together with SpCas9 protein into the pronuclei of mouse zygotes to generate the Ush2a-AExl2 mouse lines in the Genome Modification Facility at Harvard University. A total of 40 pups were obtained from the pronuclear injections. With initial genotyping and sequencing, it was confirmed that 29 mice (71%) carry deletion of exon 12 and flanking introns with different sizes. Sanger sequence verified 9 founders of mice (Figure 5D). The male founders were further backcrossed with C57BL6/J females and Fl generations were obtained.

The phenotype of the resulting Ush2a-AExl 2 mouse lines was characterized histologically and functionally. The localization of the Ush2a- \Ex 12 protein and its interaction with other Usher2 complex proteins at two months of age was determined by immunostaining, and showed that both the wild type and exon l2-skippped Ush2a proteins were localized at the transition zone of photoreceptor sensory cilia in AExl2/AExl2, AExl2/ko, and wt mice (Figure 5E).

The therapeutic potential of Ush2a-AExl2 was evaluated by transferring this Ush2a-AExl2 allele onto an Ush2a null background in Ush2a ~/~ knockout mice (Liu, X., et al, Proc Natl Acad Sci U S A, 2007. 104(11): p. 4413-8.) The phenotypes observed in these Ush2a ~/~ mice include progressive disruption of inner hair cells in the cochlea after 4 months of age and diminished inner hair cells at 7 months of age; a detectable accumulation of GFAP and mis-localization of cone opsin at 3 months of age; gradual outer nuclear layer thinning and photoreceptor abnormalities after 10 months of age; 50% loss of photoreceptors and 60% or greater reduction of ERG amplitudes for a- and b- waves by the age of 20 months (Liu, X., et al., Proc Natl Acad Sci U S A, 2007. 104(11): p. 4413-8; Lu, B., et al, Invest Ophthalmol Vis Sci, 2010. 51(4): p. 2269-76.

Cochleas were isolated from P3 Mi 12 Mi 12 and wt mice and stained for phalloidin (top panel), and Ush2a, FM1-43, and phalloidin (Figure 5F), showing essentially normal morphology in the Mix 12 Mix 12 mice. Inner and outer hair cell structures were grossly normal in Mix 12 Mix 12. AExl2/ko, and wt mice on staining with Ush2a, as compared with the knockout (Figure 5G). Auditory brain stem recordings showed improved hearing as determined by ABR in the Ush2a ko/ko mice by the Ush2a-Exl2 allele (Figure 5H). Phalloidin staining showed disrupted bundles in the ko/ko mice, but not in AExl2/AExl2, AExl2/ko, or wt mice (Figure 51).

In the retina, abnormal accumulation of GFAP in Ush2a ko/ko mice was reduced in the AExl2Zko mice at 3 months of age, and cone opsin localization was normalized in A ExJ2/kn mice at 3 months of age, as compared to ko/ko mice (Figures 5J-K).

These results showed that the protein encoded by Ush2a-AExl2 allele rescued the cochlear and retinal phenotypes observed in Ush2a ~/~ knockout mice.

Example 3. Humanized USH2A Mouse Models

A humanized USH2A mouse models is developed. Exon 12 of the mouse Ush2a gene, along with up to l500bp of the flanking introns, is replaced with the syntenic human exon 13 and up to l500bp of flanking introns. Two models are generated - one in which the wildtype human exon 13 is used and one in which the human exon 13 contains the c.2299delG mutation.

The expectation is that the mouse containing the wildtype human exon 13 will be phenotypically normal. This is supported by the high level of similarity between the amino acids encoded by mouse exon 12 and human exon 13 (74.8% exact sequence identity match and 86.9% sequence similarity match based on amino acid properties).

The expectation is that the mouse containing the c.2299delG mutation will exhibit an Usher Syndrome disease phenotype. As in Usher Syndrome patients, the c.2299delG mutation will result in a frameshift and premature stop codon, leading to a prematurely truncated, and non-functional protein. This is therefore expected to mimic the phenotype of the Ush2a knockout mouse ( Ush2a ~/~ ) as described above The humanized Ush2a mouse models enable pharmacology PK/PD studies with human USH2A-targeted therapeutic guides. In addition, they enable

demonstration of correction of disease phenotype. Lead gRNAs, along with Cas9 will be packaged in AAV. An example of what the configuration of this vector could look like is given in FIG. 9. Several AAV serotypes could be used, for example, AAV5, AAV8, AAV9 and AAV-Anc80 have all been demonstrated to show strong tropism for photoreceptors lul of AAV will be delivered to mice subretinally at doses ranging from 1E+11 to 1E+13 viral genomes/mL. Mice may be treated at different ages to assess ability to reverse pathology at various stages in disease. Mice will be evaluated at several time points to assess functional and structural rescue of retinal and cochlear phenotypes. Molecular analysis will determine expression levels of Cas9 and gRNAs as well as quantify targeted gene editing rates. ERG will measure photoreceptor function in the retina. Optical coherence tomography (OCT) and histology will examine retinal structure. Example 4. sgRNAs for exonl3 skipping in humanized USH2A mice

A comprehensive list of sgRNAs for SaCas9, SpCas9, their variants, and Cpfl were generated to target the flanking intron 12 and 13 in the humanized USH2A mice. Those guides are individually screened in the human cell lines. Optimal pairs of sgRNAs are further evaluated for skipping the exon 13 in the humanized mice.

Table 4. Guide RNAs for flanking Intronic sequences of humanized exon 13

Example 5: Screening of gRNA

Guide RNAs were screened within human USH2A intron 12 and intron 13 to find the best cutting gRNAs. To this end, ability of 41 gRNAs within intron 12 and 72 gRNAs within intron 13 to generate indels in HEK293 cells was evaluated. The cells were transfected with RNPs, gDNA was isolated 48 hours later and subjected to PCR amplification and high-throughput sequencing and analysis to determine the editing rates for each gRNA (Table 5).

Following this initial screen, a second screen focused on 2 intron 12 gRNAs and 8 intron 13 gRNAs which were identified from the initial screen. All the possible gRNA combinations were screened to determine which worked together to give the highest loss of exon 13. U20S cells were transfected with plasmids that expressed S. aureus Cas9 and the gRNAs of interest and gDNA was isolated 48 hours later for analysis. Editing was determined with a ddPCR assay that measures the presence or absence of USH2A exon 13. Results are shown in Fig. 10A (N=3) for all gRNA combinations as well as negative controls (GFP).

For the single gRNA approach, where the aim is to disrupt the exon 13 splice acceptor, gRNA that cut near the exon 13 splice acceptor were identified (Table 3). The ability of these gRNA to cut and form indels in U20S cells was tested through plasmid transfection of CRISPR Cas9 or Cpfl and the associated gRNA. The genomic DNA was extracted and subject to PCR amplification and sequencing to determine the percentage of indels (Fig. 10B).

Table 5

Example 6: Specificity of Top gRNA

The specificity of the top cutting gRNA for the dual gRNA approach was assessed using three different analyses. First, an in silico screen was conducted to identify all sites in the human genome where the particular guide could potentially cut, allowing for up to 3 mismatches or gaps in the protospacer sequence (Tables 6a and 6b).

Next, two different unbiased screens to identify off-target cut sites were completed. Guide-Seq was performed to assess the number and location of all editing events that occurred following treatment of cells with RNPs containing the one of the top gRNA. Guide-Seq was performed in primary human T cells after activation and expansion of the cells. The cells were nucleofected with RNPs and a short double- stranded oligo (Nat. Biotech. 2015, 33: 187-197). gDNA was isolated, sheared, and adapters for PCR amplification were added before PCR amplification. DNA sequences adjacent to the Guide-Seq oligo were aligned to the genome to identify the location where the double-strand oligo was inserted.

Finally, Digenome-Seq was used as a second unbiased method to locate off- target cut sites. In this method, purified genomic DNA is mixed with RNP in a cell- free system (Nat. Methods 2015, 12: 237-243). The DNA is then isolated, undergoes high-throughput sequencing, and is aligned to the human genome to identify locations where the DNA was cut (Tables 6a and 6b).

Table 6a

Table 6b

Example 7: Human model of USH2A editing and exon 13 skipping in cells

An RTddPCR assay to measure the amount of delta exon 13 USH2A transcript relative to WT USH2A was established. The WT assay amplifies the RNA junction between exons 13 and 14 while the delta 13 assay amplifies the junction between exon 12 and exon 14, which will only occur if exon 13 is precisely skipped (Fig.

11A). Another transcript ubiquitously expressed from Chromosome 1 (Clorf43) was used as a reference in this assay. The assay was validated with plasmids to check the linearity against expected inputs.

The ability of the assay to detect delta exon 13 USH2A transcripts after CRISPR/Cas9 mediated editing was tested in human CRL-5923 cells, which have been shown to express USH2A. CRL-5923 cells were transfected with plasmids expressing Cas9 and gRNA for the single guide approach and DNA and RNA were isolated from the cells 4 days after transfection. The DNA was assessed for genomic editing by high-throughput sequencing. RNA was used in the RTddPCR assay to measure the level of WT and delta exon 13 USH2A transcripts (Figs. 11B-C).

Editing and delta 13 USH2A expression was compared with two top pairs of gRNAs. CRL-5923 cells were nucleofected with RNPs containing the two gRNA and then DNA and RNA were isolated 4 days later (6 biological replicates). Loss of genomic USH2A exon 13 was measured by a ddPCR assay (Fig. 11D) and levels of WT and delta exon 13 USH2A transcripts were measured by RTddPCR (Fig. 11E).

Example 8: Editing of USH2A in human retinal explants

AAV5 vectors were cloned and produced to express inl2_32l and inl3_322 as depicted in Fig. 9. Retinal explant punches (3 millimeters) were taken from cadaver eyes within 5 hours post-mortem and individually cultured on membranes. 10 uL of AAV at a titer of 5el3 vg/mL was added between the neural retina and the membrane such that it created a viral bleb under the retinal tissue. Tissue was incubated for 28 days at which point the punches were collected. Each punch was split in half, one half for gDNA and the other half for RNA isolation. PCR amplification and sequencing showed that the punches treated with AAV had between 5 - 40% total editing, while untreated control punches had background levels of editing. RNA was used for

RTddPCR analysis to determine whether editing had caused production of USH2A delta exon 13 transcripts. Analysis showed that between 14-30% of USH2A transcripts were lacking exon 13 (Fig. 12). SEQUENCES

Hu ma n USH2A m RNA lacking exon 13: (exon 12 in bold, exon 14 dou ble u nderlined)

TGTTTG CTCTG C AG A ATACTTTACCTG G G C ACCCAAGTCTT CCTT CCAG CATTCCTG CTG CTA CAGCCTATTTGCTGAGTAACCAGGGGTTACAGCAGCGTTGCCAGGCAACGAGGGACAGCG GTCCTGTTGAAGAGCCATTTGTCACACTGAGGGGACTGGTTGAAATGCAATAAAGAAATG A TACCAG CAG CT ACT CAT GT CTT CG CCATTG CTA AG A ACGTCGTT G GTATT ACCTT ACTCTG AG AACGT GTCTGCAGTTTCCAG AA AAT G G AGTATCG CAACAT CACTT AAAGT ACCCT G CTT CAA AGTATTGCTGGCAAGTGGCGTGGGCCTGATTATTTATTTAGAAATGCTTTATCAGGAGGA G A ATG CTTTTTT GT A A AC AT G AATTG CCC AGTTCTTTC ATTG G G CTCTG G CTT CTT GTTT CAG G T C ATT G A A ATGTTG AT CTTT G CCT ATPT G CTT C A AT ATCCTT GACTGAGTCACGAGGT CTTTT CCCAAGG CTG G AG A ACGTG G G AGCTTT CAAG A AAGTTTCCAT CGT G CCAACCCAAG CAGTA TGTGGACTCCCAGACCGAAGCACTTTTTGTCACAGCTCTGCTGCTGCTGAAAGTATTCAG TT CTGTACCCAG CG GTTTTGTATTCAG G ATTG CCCAT ACAG AT CTT CACACCCTACCT ACACT G C CCTTTTCTCAG CAG G CCTCAGTAG CTG CAT CACACCAG ACAAG A AT G ATCTG CATCCTAACG CCC ATAG C A ATT CT G C A AGTTTTATTTTT G G A A AT C AC A AG AG CTG CTTTT CTT CT CCT CCTT C TCCAAAGCTGATGGCATCATTTACCTTAGCTGTATGGCTGAAACCTGAGCAACAAGGTGT AA TGTGTGTT ATAG AAAAGACAGTAG ATGGGCAGATTGTGTTCAAACTTACAAT ATCTG AG AA AG AG ACC AT GTTTTATT ATCG C AC AGTA A AT G GTTT G C A ACCT CCA AT A A A AGTA AT G AC AC TGGGGAGAATTCTTGTGAAGAAATGGATTCATCTTAGTGTGCAGGTGCATCAGACAAAAA T CAG CTT CTTT ATCAATGGCGTGGAGAAGGAT CAT AC ACCTTT C A ATG CAAG A ACT CTA AGTG GTTCAATT ACAG ATTTT G CAT CT G GT ACT GT G CAAATAGG ACAG AGTTT AAAT G GTTTAG AG CAGTTTGTCGGAAGAATGCAAGATTTTCGATTATACCAAGTGGCACTTACAAACAGAGAG A TTCTG G A AGTCTT CTCTG G AG AT CTT CT CAG ATT G CAT G CCC A AT C AC ATT GCCGTTGCCCTG GCAGCCACCCGCGGGTCCACCCTTTGGCACAGCGGTACTGCATTCCTAATGATGCAGGAG A CACAGCTGATAATAGAGTGTCACGGTTGAATCCTGAAGCCCATCCTCTCTCTTTTGTCAA TG AT AAT G ATGTTG GT ACTT CAT G GGTTTCAA AT GT GTTTACAAACATT ACACAGCTT AAT CAA GGAGTGACTATTTCAGTTGATTTGGAAAATGGACAGTATCAGGTGTTTTATATTATCATT CA GTTCTTTAGTCCACAACCAACGGAAATAAGGATTCAAAGGAAGAAGGAAAATAGTTTAGA T TGGGAGGACTGG C A AT ATTTT G CC AG G A ATT GTG GTG CTTTT G G AAT G A A A A AC A AT G G AG ATTT G G A A A A ACCT G ATT CTGT C A ACT GT CTT CAG CTTT CC A ATTTTACT CC AT ATT CCCGTG G CAATGTCACATTT AGC AT CCT G ACACCTGG ACCAAATT AT CGTCCT G G AT ACAAT AACTT CT A T AAT ACCCCAT CT CTT CAAG AGTT CGTAAAAGCCACGCAA AT AAG GTTTCATTTT CATGG G C AGTACTATACAACTGAGACTGCTGTTAACCTCAGACACAGATATTATGCAGTGGACGAAA TC ACCATTAGTGGGAGATGTCAGTGCCATGGTCATGCCGATAACTGCGACACAACAAGCCAG C CAT ATAG ATG CCTCTG CTCCC AG G AG AG CTT C ACT G A AG G ACTT C ATT GTGATCGCTG CTT G CCT CTTT AT AAT G AC A AG CCTTT CCGCCAAGGTGAT C A AGTTT ACG CTTT C A ATT GTA A ACCT TGTCAATG CA ACAG CCATTCCA AAAG CTG CCATT ACAACAT CT CT GTAG ACCCATTT CCTTTT GAGCACTTCAGAGGGGGAGGAGGAGTTTGTGATGATTGTGAGCATAACACTACAGGAAGG AACTGTGAGCTGTGCAAGGATTACTTTTTCCGACAAGTTGGTGCAGATCCTTCGGCCATA GA T GTTT GCA AACCCT GTG ACTGTG AT ACAGTT G G CACT AG AAAT G GTAG CATT CTTT GTG ATC AGATTGGAGGACAGTGTAATTGTAAGAGACACGTGTCTGGCAGGCAGTGCAATCAGTGC CAGAATGGATTCTACAATCTACAAGAGTTGGATCCTGATGGCTGCAGTCCCTGTAACTGC A ATACCTCTGGGACAGTGGATGGAGATATTACCTGTCACCAAAATTCAGGCCAGTGCAAGT G CAA AG C A A ACGTTATT G GTTTTT AT ATTT CT CC AG G C A AT GCCACTGGCTGCCTGCCATGC TCATGCCATACAACTGGTGCAGTTAATCACATCTGTAATAGCCTGACTGGTCAGTGTGTT TG

CCAAGATGCTTCCATTGCTGGGCAACGTTGTGACCAATGCAAAGACCATTACTTTGG ATTTG ATCCTCAGACTGGAAGATGTCAGCCTTGTAATTGTCATCTCTCAGGAGCCTTGAATGAAA CC TGTCACTTGGTCACAGGCCAGTGTTTCTGTAAACAATTTGTCACTGGCTCAAAGTGTGAT GC TTGT GTT CCCAGT G CAAGCCACTT G G AT GTCAACAAT CT ATT G GGTTG CAGCA AAACTCCAT TCCAGCAACCTCCGCCCAGAGGACAAGTTCAAAGTTCTTCTGCTATCAATCTCTCCTGGA GT CC ACCTG ATT CT CCA A AT G CCC ACTG G CTT ACTT AC AGTTTACT C AG G G AT G GTTTT G A A AT C T ACACA ACAG AG G AT CAATACCCAT ACAGTATT CAATACTT CTTAG ACACAG ACCT GTTACC ATATACCAAATATTCCTATTACATTGAGACCACCAATGTGCATGGTTCAACAAGGAGTGT AG CTGTCACTTACAAGACAAAACCAGGGGTCCCAGAGGGAAACTTGACTTTAAGTTATATCA TT CCT ATT G G CTC AG ACTCTGTG AC ACTT ACCT G G AC A AC ACTCT C A A AT C A AT CT G GTCCC AT A GAGAAATATATTTTGTCCTGTGCCCCTTTGGCTGGTGGTCAGCCATGTGTTTCCTACGAA GG T CAT G AA ACCT CAGCTACCAT CT G G AAT CTGGTTCCATTT G CCAAGTACG ATPTT CT GT ACA G GCGTGTACTAG CG GG G G CTGTTTAC ACAG CTTG CCCATT ACAGTG ACCACAG CCCAG GCC CCT CCCCA AAG ACT AAGT CCACCT AAG ATGCAG AA AAT CAGTTCT ACAG AA CTT CAT GTAG A ATGGTCTCCACCAGCGGAACTAAATGGAATAATTATAAGATATGAACTATACATGAGAAG A CTGAGATCTACTAAAGAAACCACATCTGAGGAAAGTCGAGTTTTTCAGAGCAGTGGTTGG C T CAGTCCT CATT CATTT GTAG AAT CG GCCA AT G A AAAT GCATT A AAACCT CCT CA AACAAT G ACAACCATCACTGGCTTGGAGCCATACACCAAGTATGAGTTCAGAGTCTTAGCTGTGAAT AT GGCTGGAAGTGTGTCTTCTGCCTGGGTCTCAGAAAGAACGGGAGAATCAGCACCTGTATT C ATG AT CCCT CCTT CAGTCTTT CCCCT CT CTT CGTACT CT CT CAAT AT CT CCT G GG AG A AGCCAG CAG AT AAT GTTACA AG AG G A AAAGTT GTG G GGTAT G ACAT CAAT AT G CTTT CTG AACAAT C ACCT CAACAGT CT ATT CCCATGG CGTTTT CACAG CTGTTG CACACTG CT AAAT CCCAAG AACT ATCTTACACTGTAG AAG G ACTG AAACCTTATAG GATATATG AGTTTACTATTACTCTCTG CAA TTCAGTTGGTTGTGTGACCAGTGCTTCGGGAGCAGGACAAACTTTAGCAGCAGCACCAGC A CAACTG AG G CCACCT CT G GTTAAAG G AAT CAACAGCAC AACAAT CCAT CTT AAGT G GTTTCC ACCTGAAGAACTGAATGGACCCTCTCCTATATATCAGCTGGAAAGGAGAGAGTCATCTCT AC CAG CT CTG ATG ACC A CG ATG ATG A A AG G AAT CCGTTTC AT AG G AAAT G G GTATT GTA A ATTT CCC AG CTCC ACTC ACCC AGTC A ATAC AG ACTT C ACTG G CATT A AG G CC AG CTTT CG A AC A A A AGTG CCTG A AG GTTTG ATTGTCTTTG CAGCATCACCTG G CAATCAGG AAG AGTATTTTG CAC TTCAGTTGAAGAAGGGACGTCTTTATTTTCrmTGATCCTCAGGGGTCACCAGTGGAAGTA ACTAC A ACT AAT G ATC ATG G CAA AC A AT ATAGTG AT G G A A A AT G G CAT G AAAT A ATT G CTA TTAG G CAT C AG G CTTTT G G CCA A AT C ACTCTG G ATG G G ATATATAC AG GTTCCTCTG CCAT C CTGAATGGTAGTACTGTTATTGGAGATAACACAGGAGTCTTTCTGGGAGGGCTCCCGCGA A GTTATACCATCCTCAGGAAGGATCCTGAGATAATCCAAAAAGGTTTTGTGGGCTGTCTCA AG GATGTACATTTTATGAAGAATTACAATCCGTCAGCTATTTGGGAACCTCTGGATTGGCAG AG TTCTG A AG A AC A A AT C A ACGT GTATAACAGCTGGGAGGGATGTCCCG CTT CATT AAAT GAG GGAGCTCAGTTCCTAGGAGCAGGGTTCCTGGAACTTCATCCATATATGTTTCATGGTGGA AT G AACTTTG AG ATTT CCTTT A AGTT CAG AACT G ACC AATT AAAT G G ATT G CTT CTTTT CGTTTA TAACAAAGATGG ACCTG ATTTTCTTGCTATGGAGCTGAAAAGTGGAATATTGACCTTCCGGT TAAATACCAGTCTTGCCTTTACACAAGTGGATCTATTGCTGGGGCTATCCTATTGTAATG GA A AGTG G AAT A AAGT CATT ATT A A A A AG G A AG G CT CTTT CAT AT CAG CAAGTGTG A ATG G AC TGATGAAGCATGCATCGGAGTCCGGAGACCAGCCACTGGTGGTGAATTCACCAGTTTATG T GGGAGGAATCCCACAGGAACTGCTGAACTCTTATCAACATTTGTGTTTGGAACAAGGTTT C GGTGGTTGCATGAAGGATGTTAAATTTACACGGGGTGCTGTCGTTAACTTGGCATCTGTG TC CAGCGGTGCTGTCAGAGTCAATCTGGATGGATGCCTATCAACTGACAGTGCTGTTAACTG C AGGGGAAATGACTCCATCCTGGTTTACCAGGGAAAAGAGCAGAGTGTTTACGAGGGTGGT CTCCAGCCTTTTACAGAATACCTGTATCGAGTGATAGCCTCGCATGAAGGAGGTTCAGTA TA TAGTGATTGGAGTCGAGGACGTACAACAGGAGCAGCTCCACAAAGTGTGCCAACTCCCTC A AGAGTCCGCAGCTTAAATGGATACAGCATTGAGGTGACCTGGGATGAACCTGTTGTCAGA G GTGT A ATT G AG AAGTACATT CT G A AAG CCT AT AGTG AG G ACAG CACCCGTCC ACCCCG CAT GCCCTCTGCCAGTGCTG A ATTT GTCA AT AC A AG C A ACCT C AC AG G CAT ATT G AC AG G CTT G C T ACCCTT C A A A A ACT AT G C AGTA ACCCTA ACTG CTTG CACTTTG GCTGG CTGTACTG AG AG C T CACATGCATT G AACAT CT CT ACT CCACA AG AAG CCCCACA AG AGGTTCAG CCACCAGT AG C CAA AT CCCTT CCCAGTTCTTT G CTG CTCTCCTG G A ACCCACCC AAAA AG GCA AATG GTATTAT AACTCAGTACTGTTTATACATGGATGGGAGGCTGATCTATTCAGGCAGTGAGGAGAACTA C ACAGTCACAG ATTTAG CAGT ATTT ACACCCCACCAGTTT CT ACT AAGT G CATG CACACATGT G GG CT GTACAA ACAGTTCCTGG GTCCT ACT GT ACACAG CACAG CTG CCACCAG AACACGTG GATT CCCCAGTTCT G ACTGTCCT G G ATT CT AG A ACT AT ACACAT ACAGTGG AA ACAACCA AG AAAAATAAGTGGGATTCTGGAACGCTATGTATTATATATGTCAAACCATACACATGATTT TA CAATTTGG AGTGTCAT CT AT AACAGTACAG AACTTTT CCAG G AT CAT ATG CT ACAAT ACGTTT TACCTG GT A AT A A AT AT CT CAT C A AG CTG G G AG CTT GCACAGGTGGTGGGTGCACAGTGAG TGAGGCCAGTGAGGCCCTAACTGACGAGGACATACCCGAAGGCGTGCCAGCCCCCAAAGC CC ACT CAT ATT C ACCTG ACT CCTTT AATGTCTCCTGGACTGAGCCTG A AT AT CCG A AT G GTGT T AT CACG AGTTAT G G ATT AT AT CT AG ATGGTAT ATT AAT CCACA ATT CCT CAG AACT CAG CT A TCGTG CTT ACG G ATTT G CT CCTT G G AGTTTAC ATT CCTT CAGAGTCCAAG CAT G C ACG G CC A AAGGTTGTGCTCTGGGCCCACTGGTGGAAAATCGAACTCTAGAAGCTCCTCCTGAAGGAA C AGTAAATGTGTTTGTCAAAACACAGGGATCCCGGAAAGCCCACGTGAGGTGGGAAGCACC TTTTCGCCCTAATGGACTCTTAACACACTCAGTCCTTTTCACTGGGATATTCTATGTAGA CCC AGTAG GT AAT AACTACACCCTT CT G A AT GTCACAAA AGTCAT GTACAGCG G AG AAG AG ACA A A CCTTT G G GTG CT CAT CG ATG G G CTG GTT CCTTTTACC A ACT AT ACT GTAC A AGT G AAT ATT TCAA ATAG CC AAG GCAG CTTG ATAACTG ATCCTATAACA ATTG CAATG CCTCC AG G AG CTCC AG ATG G CGTG CTG CCTCCC AG G CTTTC ATCTG CC ACTCC A ACC AGTCTTC AG GTTGTCTG GT CT ACACCAG CT CGT AAT AACG CT CCT G GCTCT CCCAG AT ACCAACT CCAG AT GAGGTCTGGC G ACT CCACCCATGG ATTT CTAG AGTTATTTT CCA AT CCTT CTGCAT CGTT AAG CTATG AAGTG AGTG AT CT CCA ACCGTACAC AG AGT AT AT GTTT CG GTT G GTTGCCT CCA AT G G ATTT G G CAG T G CACATAGTT CTT G G ATTCCATT CATG ACCG CAG AG G ACAA ACCTGG ACCT GTAGTT CCT C CG ATT CTT CTG GAT GTG AAGTCAAG AAT GAT GTTG GTCACCT G G CAGC AT CCT AG AAAAT CC AAT G G G GTT ATT ACCC ATT AT A AC ATTT AT CT AC AT G G CCGTCT ATACTT GAGAACTCCTGGA AATGTCACTAATTGCACAGTGATGCATTTACACCCATACACTGCCTATAAGTTTCAGGTA GA AG CCTG C ACTT CAA A AG GAT GTT CCCTTT CACCAGAGTCCCAGACTGTATGGACACTCCCAG GGGCACCGGAAGGGATCCCAAGTCCAGAGCTGTTCTCTGATACTCCAACATCTGTGATTA TA T CTT G G CA ACCCCCT ACCCACCCCAATGG CTT G GT G GAG A ATTT CACAATT GAG AG AAG AGT CAAAGGAAAGGAAGAAGTTACTACCCTGGTGACTCTCCCGAGGAGTCATTCCATGAGGTT T ATT G AC A AG ACTT CT G CT CTT AG CCC AT G G AC A A A AT AT G AAT ATCGGGTACTGATGAGCAC TCTTCATGGAGGCACAAACAGCAGTGCTTGGGTAGAAGTTACCACAAGACCCTCACGACC T GCTGGGGTGCAGCCACCTGTGGTGACAGTGCTGGAACCCGATGCAGTCCAGGTCACTTGG AAACCCCCACTCATCCAGAACGGAGACATACTTAGCTATGAGATTCACATGCCTGACCCT CA CAT CACTTT A ACCAAT GTG ACTT CCG CAGT GTT AAGT CAA AAAGTTACT CAT CTG ATT CCTTT C ACT A ATT ATT CT GTC ACC ATT GTTG CTT G CTC AG G GG GTAATGG GTACCTTG G AG G GTG CA CAG AG AGTTTACCT ACCT AT GTT ACCACT CACCCCACCGTACCT CAG AAT GTT G GCCCATT GT CTGT G ATTCCACT AAGT G A AT CAT AT GTT GT G ATTT CTT G GCAACCACCAT CCA AG CCAAAT GGACCTAATTTGAGATATGAGCTTCTGAGACGTAAAATCCAGCAGCCACTTGCATCAAAT CC CCCAG AAGATTTAAATCGGTGGCACAAT ATTT ATTCAGGAACTCAGTGGCTTTATGAAGATA AGGGTCTTAGCAGGTTTACAACCTATGAATATATGCTCTTCGTACACAACAGTGTGGGTT TT ACACCGAGCCGAGAAGTGACTGTGACAACGTTAGCTGGTCTTCCAGAGAGAGGAGCCAAT CT CACT G CG AGT GTCCTT A ACCACACAG CCAT CGACGTGAGGTGGGCT A AACCAACT GTT CA AG ACCTAC A AG GTG A AGTTG A AT ATT AC AC ACrTTTTT G G AGTTCTG CTACCT C A A ACG ACT CTCTAA AAATCTTG CCAG AT GTAAACT CT CATGTCATT G G CCACCTA AAG CCAAACACAG AG T ATT G G AT CTTT AT CT CTGTCTT CAAT G G AGTCCACAG CATCA ACAGTG CAG G ACTTCATGCA ACCACTTGCGATGGGGAGCCTCAGGGCATGCTTCCTCCAGAGGTTGTCATCATCAACAGT A CAG CTGTACGTGTCATCTG G ACATCTCCTTCA AACCCA AATG GTGTTGTCACTG AGTATTCTA TCTATGTAAATAATAAGCTCTACAAGACTGGAATGAATGTGCCTGGGTCGTTTATTCTGA GA G ACCTGTCT CCCTT CACT AT CT AT G AC ATT CAGGTTGAAGTCTG C AC A AT ATATG CCTG CGTG AA AAG CAAT G G AACCCAA ATT ACCACT GT G G A AG ACACT CCAAGTG AT ATACCA ACACCCA C A ATT CGTG G CAT C ACTT C A AG AT CT CTT C A A ATT G ATT GGGTGTCTCCACGGAAG CCA A AT G G CAT C ATT CTT G G ATATG ATCT CCT ATG G A A A AC AT G GTAT CCAT G CG CT A A A ACT C A A A A GTTAGTGCAGGATCAGAGTGATGAGCTCTGCAAGGCAGTGAGGTGTCAAAAACCTGAATC T ATCTGTG G ACACATTTG CT ATT CTT CTG A AG CTAAG GTTTGTTGTA ACGG AGTG CTCTATAAC CCCAAGCCTGGACATCGCTGTTGTGAAGAAAAGTATATCCCGTTTGTTCTGAATTCTACT GG AGTTT GTTGTGGTGGCCG A AT AC AG G AG G C AC A ACC A A AT CAT C AGTG CTG CTCTG G GTAT TACG CTAG A ATT CTACC AG GTG A AGTATG CTGTCC AG ATG A AC AG C AC A ATCG G GTTTCT GT TGGCATTGGTGATTCCTGCTGTGGCAGAATGCCGTACTCCACCTCAGGAAACCAGATTTG CT GTGCTGGGAGGCTTCATGATGGCCATGGCCAGAAGTGCTGTGGCAGACAGATTGTGAGCA ACGATTTAGAGTGTTGTGGTGGAGAAGAAGGAGTGGTGTACAATCGCCTTCCAGGTATGT T CTGTTGTGGGCAGG ATT ATGTG A AT ATGTC AG AT ACC AT ATG CTG CT CAG CTT CCAGTGGAG AGTCTAAAGCACATATTAAAAAGAATGACCCGGTGCCAGTAAAATGCTGTGAGACTGAAC T TATTCCAAAGAGCCAGAAATGCTGTAATGGAGTTGGATATAATCCTTTGAAATATGTTTG CT CT G AC A AG ATTT C A ACT G G A AT G ATG ATG AAG G A A ACC A A AG AGTGCAGGATCCTCTGCCC AG CATCTATG G AAG CCACAG AACATT GT GG CAG GT GTG ACTT CAACTTT ACCAG CCACATTT GCACTGTGATAAGAGGGTCTCACAATTCCACAGGGAAGGCATCAATTGAAGAAATGTGTT C ATCTGCCGAAGAAACCATTCATACAGGGAGTGTAAACACGTACTCTTACACAGATGTGAA C CTCAAGCCCTACATGACATATGAGTACAGGATTTCTGCCTGGAACAGCTATGGGCGAGGA C TCAGCAAAGCTGTGAGAGCCAGAACAAAAGAAGATGTGCCTCAAGGAGTGAGTCCCCCTA CGTGGACCAAAATAGACAATCTTGAAGATACAATTGTCTTAAACTGGAGAAAACCTATAC A AT CA AAT G GT CCT ATT ATTT ACT ACAT CCTT CTT CG AAAT GG AATT G A ACGTTTTCG G G G AAC AT CACTG AG CTT CT CTG AT AA AG AGG G AATT CAACCATTT CAG G AAT ATT CAT AT CAG CTG A AAGCTTG CACG GTTG CTGG CTGTGCCACCAGTAG CAAG GTAGTTG CAG CTACTACCC AAG G AGTTCCGGAGAGCATCCTGCCACCAAGCATCACAGCCCTAAGTGCAGTGGCTCTGCATCT G AG CTG G AGTGTCCCTG AG AA ATCAAACGG CGTCATTAAAG AGTACCAG ATCAGG CAG GTTG G G A AAG GTCTC AT CCACACTG ACACCACTG ACAG G AG ACAG CATACG GTCACAG GTCTCCA G CC ATAC ACC A ACTAC AG CTTC ACTCTTAC AG CTTGTAC ATCTG CTG G GTG C ACTTC A AG CG AGCCTTTTCTAGGTCAGACACTGCAGGCAGCTCCTGAAGGAGTTTGGGTGACACCTCGAC A C ATT AT CAT C A ATT CTACA AC AGT G G AATT AT ATT G G AGTCTG CC AG A A A AG CCC A AT G G CC TCGTTTCTCAATATCAATTGAGTCGTAATGGAAACTTGCTTTTCCTGGGTGGCAGTGAGG AG CAG AATTT CACT GAT A AAAACCT G G AG CCCAAT AG CAG ATACACTT ACAAGTT AG AAGTCA AAA CTGG AGGTGGCAGCAGTGCTAGTG ATG ATTACATTGTTCAAACACCTATGTCAACACC AG AAG AAAT CT AT CCT CCAT AT AAT AT CACAGT AATT GG GCCTT ATT CT AT ATTT GTAGCTT G GATACCACCAGGGATCCTCATCCCCGAAATTCCTGTGGAGTACAATGTCTTACTCAATGA TG G A AGTGT A AC ACCTCTG G CCTT CT CCGTTG GTC AT CAT CAAT CC ACCCTT CTG G A A A ATTT G A CTCCATTCACACAGTATGAGATAAGGATACAAGCATGTCAAAATGGAAGTTGTGGAGTTA G C AGTAG G AT GTTT GTC A A A AC ACCT G A AG C AG CCCC A AT G G AT CTT A ATT CTCCT GTTCTT A AG G CACTG GG GTCAG CTTG CATAG AG ATTAAGTG G ATGCCACCTG A AAAACC AAATG G AAT CAT CAT C A ACT ACTTT ATTT ACAGACGCCCTGCTGG C ATT GAAGAGGAGTCT GTTTTATTT GT CTG GTCAG A AG GAG CCCTTG AATTTATGG AT G AAGG AG ACACCCT GAG G CCTTTCACACTC TACGAATATCGGGTCAGAGCCTGTAACTCCAAGGGTTCAGTGGAGAGTCTGTGGTCATTA A C AC A A ACTCTG G A AG CTCCACCTC A AG ATTTTCC AG CTCCTTG G G CTC A AG CCACGAGTGCT CATT CAGTTCT GTTG AATT GG ACAA AG CCAG AAT CT CCCA AT G G CATT AT CT CCCATT ACCGT GTGGTCT ACCAG G AG AG ACCCG ACG AT CCT ACATTT AACAG CCCT ACCGT G CAT G CTTT CAC AGTGAAGGGAACAAGCCATCAAGCCCACCTGTACGGGTTAGAACCATTCACAACATATCG C ATT GGTGTTGTGGCTG C A A ACC AT G C AG G AG A A ATPT A AG CCCTT G G ACTCTG ATT C A A AC CTT AG AAT CTT CCCC A AGT G G ACTG AG A A ACTTT ATAGTAG AACAG A A AG AG AAT G G CCG G G CATT G CT ACT ACAGT G GT CAG AACCT AT G AG A ACCAAT G GTGTG ATT AAG ACATACAAC A TCTTCAGTGACGGGTTCCTGGAGTACTCTGGTTTGAATCGTCAGTTTCTCTTCCGCCGCC TGG AT CCTTT C ACTCTCTAC AC ACTG ACCCTG G AG G CCTG CACC AG AG C AG GTTGTG C AC ACTCG G CG CCT CAG CCT CTGTG G ACAG AT G AAGCCCCT CCAG ACT CT CAG CTG G CTCCTACTGTCC A CTCTGT G A AGT CCACCAGT GTTG AG CTG AGCTG GTCTG AG CCT GTTAACCCA AAT G G AAAA AT AATT CG CTATG A AGTG ATT CG C AG ATG CTT CG AG G G A A A AG CTT G G G G A A AT CAG ACG A TCCAGGCCG ACG AGAAAATTGTTTTCA CAG AAT AT AA CACTG AAAGGAATACATTTATGTAT AATGACACAGGTTTGCAACCATGGACGCAGTGTGAATATAAAATCTACACTTGGAATTCA G CTGGGCATACCTGTAGCTCTTGGAATGTGGTGAGGACATTGCAAGCACCTCCAGAAGGTC T CT CT CCA CCT GTG ATAT CCT AT GTTT CT AT G A AT CCCCAAA AACT G CTG ATTT CCT G G AT CCC ACCAGAACAGTCTAATGGTATTATCCAGTCCTATAGGCTTCAAAGGAATGAAATGCTCTA TC CTTTTAGCTTTGATCCTGTGACTTTCAATTACACTGATGAAGAGCTTCTTCCTTTTTCCA CCTA TAG CTATG C ACT CCA AG CCTG CACGAGTGGAGG ATG CTCCACCAG C A A ACCC ACC AG C AT C ACA ACTCTG G AGG CTGCTCCATCAG A AGTC AG CCCTCCAG ATCTTTGG G CCGTCAGTG CCAC T CAA AT G A AT GTATGTTG GT CACCGCCCACAGT G CA AAAT G G AAAG ATT ACT AA AT ATTT AG TTAG ATATG AT AAT A A AG AGT CCCTT GCTGGCCAGGGCCTGTGCCTGCT G GTTTCCC ACCT G CAGCCTTACTCTCAGTATAACTTCTCCCTTGTAGCCTGCACGAATGGAGGTTGCACAGCT AG TGTGT CAA A AT CTG CCTG G AC A AT GGAGGCCCTGCCAG AG A AC AT G G ACTCT CCA AC ATT G CAAGT CACAGG CT CAG A AT CAATAG AA AT CACCTGG AA ACCT CCAAG AA ACCCAAAT G G CC AGATCAGAAGTTATGAACTTAGGAGGGATGGAACCATTGTATATACAGGCTTGGAAACAC G CTATCGTGATTTTACTCTCACCCCAGGTGTGGAGTATAGCTACACAGTAACTGCCAGCAA CA GCCAAGGGGGTATTTTGAGTCCTCTTGTCAAAGATCGAACCAGCCCCTCAGCACCCTCAG G GATGGAACCTCCAAAATTGCAGGCCAGGGGTCCTCAGGAGATCTTAGTGAACTGGGACCC T CC AGTG AG A AC A A AT G GTG ATAT CAT C A ATT ATACCCT CTT CAT CCGTG A ACT ATTT G AAAG AG A A ACT A A A AT C ATAC AC AT A A AC AC A ACT CAT AATT CTTTT G GTATG C AGT CAT ATATAGT AAACCAGCTGAAGCCATTTCACAGGTATGAAATACGAATTCAAGCGTGCACCACCCTGGG A TGTG CATCAAGTG ACTG G ACATTCATACAG ACCCCTG AG ATTG CACCTTTG ATG CAACCCCC TCC AC ATCTG G AG GTAC A A ATG G CTCC AG G AG G ATTCC AG CCAACT GTTT CT CTTTT GTG G A C AG G ACCG CTG C AG CCA A AT G G A A A AGTTTT GT ATT ACG AATT ATAC AG A AG AC A A AT AG C AACTCAGCCTAG A A A AT CCA AT CCAGTCCTA ATCTATA ACG G A AG CT C A AC AT CTTTT ATAG ATTCCGAACTATTGCCTTTCACAGAGTATGAGTATCAGGTCTGGGCAGTGAATTCTGCAG GA AAAGCCCCCAGTAGCTGGACATGGTGCAGAACCGGGCCAGCCCCACCAGAAGGTCTCAGA G CCCCCACGTTCCAT GTG AT CT CTT CT ACCCAAGCAGT G GT CA ACAT CAGT G CCCCTGG G A A GCCCAACGGGATCGTCAGTCTCTACAGGCTGTTCTCCAGCAGCGCCCATGGGGCTGAGAC A GTG CTATCCG A AG G CAT G G CC ACCC AG CAG ACTCT CC AT G G CCTT C A AG CCTT C ACTA ACTA CTCTATTGGAGTAGAGGCCTGCACCTGCTTCAACTGTTGCAGCAAAGGACCGACAGCTGA A CTG AG A ACCC AT CCTG CCCC ACCCTC AG G ACTGTCCTCT CCAC A A AT CGGGACGCTGGCCTC AAGG ACG G CCT CCTT CCG GTGG AGTCCCCCCATGTTCCCCAAT G GTGT CATT CACAG CT AT G AACTCCAATT CCACGT G G CTTGCCCT CCTG ACT CAG CCCT CCCCT GTACT CCCAGCCAAAT AG AA ACAAAGT ACACG G G G CTGG G G CAG A AAG CCAGCCTTG G GG GTCTCCAG CCCTACACCA CATACAAG CTG AG AGTG GTG G CACACAACG AGGTG GG CAGTACG G CTTCCG AGTG G ATCA GTTTCACCACCCAA AAAG A ATT G CCT CAGTACCG AG CCCCATTTT CG GTGG ACAGCA ATTT G TCTGTGGTGTGTGTGAACTGGAGTGACACCTTCCTCCTGAACGGCCAACTGAAGGAGTAC G TGTTAACCGACGGAGGGCGACGCGTGTACAGCGGCTTGGACACCACCCTCTACATACCGA G AACG G CG G AC AAAACCTT CTTTTT CCAG GTCAT CT G CACG ACTG ACG AAG G A AGTGTTAAG ACG CCGTTG ATCCA ATATG ATACCTCTACTG G ACTT G G CTT G GTCCT A AC A ACTCCTG G G A A AAAGAAGGGATCGCGGAGCAAAAGCACAGAGTTCTACAGCGAGCTGTGGTTCATAGTGTT AATGG CG ATG CTGG G CTTG ATCTTGTTG G CCATTTTT CT GT CCCT G ATACT ACAAAG A AAAA TCCACAAAGAGCCATATATCAGAGAAAGACCTCCCTTGGTACCTCTTCAGAAGAGGATGT CT CCATTG AAT GTTT ACCCACCG G G G G A A A ACC ATATG GGGTTAG CCG ATACC A A A ATTCCCC G GTCTG G G AC ACCTGTG AGTATCCGC AG CA ACCG G AGTG CATGTGTCCTG CG CATCCCG AG T C A A A ACC A A ACC AG CCT AACCT ACT CCCAGG GTTCT CTT CACCG CAG CGTCAG CCAG CTCA TGGACATTCAAGACAAGAAAGTCTTGATGGACAACTCACTGTGGGAAGCCATCATGGGCC A CAACAGTGGACTGTATGTGGATGAAGAGGACCTGATGAACGCCATCAAGGATTTCAGCTC A GTGACTAAGGAACGCACCACATTCACAGACACCCACCTGTAAAGGATGGAAACCCAGAAG A CGTAACCCT G G AAT G CAAG GTCT GCACCCATTT CCT CCTGG GTTAT CACT CACACAT CAT AAA TG CT G A A A AG CC ATT GTTT ATT AT CCT AT A ATT CTTT AAAG A A AT G ATG ACT GTTTTT G AAAG TGTTCCTTCCTAATAGAGGTCTAAGAAATGATATTTTTCTCATCTTAAATGAGAGAGAAT ATT CAT AT G A A A AT ACTT G ATTT G CT CTT ATTTT GTAG A AG ACAAAG A AGTAT GTA ATT GT C ACTT GGTTCTGTTTGGCAGTGATGCTCCTGGTTAACTGAATAATCAGTGGCAATTTCAAGATGG CT CACAGTT GTTAG A AGTAGT A AGTTAGTT ACT G G CT CAAA AAT GATT CT GTTG AAAG GAT GTC ACTGCTGTTCATTTCTATCTGCCATTTCTGTCAGGGTTGACACAATCCTGCAAGAATAGT TAT TCTAATGATCACAGCTGCTAAATGAATCCCAAACTTTGCACCAGGTCGACAAACTTTTCT GA AG GTTCT ATTT ATTT ACC AT AC ATAG G GTTACTT ACC A A ACTTTTT GACAAGGCTGAAGGTTC T ATTT ATTT ACAAT ACAT AG GGTTACT CACCA AACTTTTTG ACAAGG CAACACAT AACTTACA CAT A A AT GTCTCT GTTCTT G CATTT ATG AATPT CCAAAA AT CT AAG G AGT A AACAG CTT ATT TATACATTTTGAGGAGAAAACAAAGTGTTTCACTAGGAACACCTCTACTTGAACCAATGT TT TT ATTT CAT AT ATTTT ATAGTTTTG AAACT AGTTTCT CAT A AAATT CT GTCAATT CACTG AAT A TC AG AG AAT ACTG AC AT CTT C AACCT AG C AC ATTT C A A AT G G A A ACT ACTGTTCT ATTT G C A A TATTAG G CTG CGTGAA ATTTT AAAAG G A AAAAT GT AT CT GTTCCTT CTAGCATT AACAT AT AT ACAT GTAG AG AC AAG ACT AT ACCT ATGTGTATATATATGTAT AT CAT GTATAT ATT ACT CTG C ACTATATCCCTTCTTTTTGGAGAACTAGCCATTATTTTAGCCACAGAATCAGTAAGAACA GAT GATATGCAACAGTACCAATTACGGTTCAAAAATGTCTGTCACCTGCTCTAGTTGGATTAC AA AGTCATT G GT G AA AGT CCT ATG G CAAG AAA AATTTT CTTGCAA AT CAT CCACAT AAAAT CAG AT ATTT A A ATTT GTT CTT CAT G G A A A AC AG AGTA AG A A A ACCT CTT GTCTTCCTT CAT CCTT A AAGGTCTTTGTGACCCCAGG AAAAT ATTGACTCTGTCTAACACACAATAGTCACAATACTTTT TGTGAATCTACAACCAGAGACAGGCAAAAACTTGTAAAGTAAGGGATAGTCTTACTTATT CT G CCTG AAAACAAT GT ATTACCCCAGGGCCCAACAGTAAAAG ATT GTGG ACTTTTT G G GTATT GAG ATTT CAT CTAG CTCTGTG AG AG AG C AG CTCCTC AG ACTG ACC A ACT CCTAG AC A A AGTT TG CCAACC ATAAGTGTCAA AAG CACAG G CCAGTATTAAG C AG A AGTTCT ACC ACCTT ATT AG A ACTG CT AT A A AC A A A AG CAT CTG A A AT A ATT GTGCACATCTGGCAGTGACTGTAG AAAAT ACG A A AT AT AT ATTT CTCG CC A AGTTTTT AT ACTTT CT G A A AT G A A A ACAT AG G ATT G ACTAG TTTACTG GTTTTT ATT CCC AT ATG CCG ATT CT G G G ACA ATAA AGTTGTTTA AAG CTG G CACAA ATA AG C ATTA ACC A AG G CTGTGTCC ACCTTCTGTG AG CT ACTTA AG GTATATAG G A A AG G A GTGGTCACAA ACTTGC AT CCT AAT CCTT G GT G G ACT CTT CT AAG AAT ACAGTTT G CTAGTCAC AAAGAATAGTCTACAAATATGCTTTGCTAGGTTCAGAAGATTGAGTTTATCCTGATTTTT GA AA AATT AACCAG GTAT CTTT AT CACT GTGT ATTTTT CCAAG CACAGTAT AA AATTTT AACA AC G C AC A A A A A A ATAC AG AACTG C AG G G G ATTTT AT CTT G G AT CATT AT CCATTT AAT CAT CT A ATT AG AC AT G A ACTCAGTTAG CTG AAT C ATTT AC ATTTT G ACT CC ATAG CTT AG G G C AG AC A GAAGCCTGTATGGCTTCTGCCCAGAACTCTGTCCCCTGCTACATGTCTAAGTTTACTTGT ATT T ATTT C AG AG A AG A ACTCTA AG ATGTTG CTTT G CT ACTTT A AGTG GT ATT G CGT G CCA AG CC T CT ATT AT AC A A ACC AT G C AG ACTCG CCTCTAG AG ATT CT G ATT CGGTTGATCTGGGGTGTG TGGCTGAGGCATCAGTACTTTTTAAAGCTTCCAGGTGTTCTAATGTTGAGACCCACTGAT GT TCC AC A ATCTG G A AG A A AT CAT GTAC AG G A ATA ATATG CT ATG C AC AG GGACTATG CTCCTT G G CT C ACCCCTT CT CCCTT AT A A AC A AT GAG C AGTTCTT G ATG A A CCT CTTT A A ATTT A A AT C T CCTG ACT CACATTTT ACCAATT GTACATGCC ACATT CT CAG CTT ACG AACTACCAT GTTTT GT T ATT CTT AAT AT CAACT GTTT G GT A AG AGT ACAGTT GTTTTT AT ACACT CT A AG A AAT GT GTT T AT AAT CT ACTGTAATTT CCACT AAAT G G AACCCAAAT ATT AAT GTTATG GTACCATAT ACT G AT GTAA AAAT CAT G CTGG CAT CCAT G A ACACACCG GTAAAT A AAACATAGT CCA AGTGG AA G AATT CATT A ATA AG G A ACTTTT AATT ATGT C AC A A AT G A ATAGTT G GTTTCCA AT G C AC A A AT AT CAT GT AA ACT AAT CT AA AG ATGGTTT G CTT AAT AAAT ATTT G A AT GTG ACC (SEQ ID NO:l)

Translation of this mRNA is expected to result in expression of human Usherin protein lacking part of laminin EGF-like domain 4, all of domains 5, 6 and 7 and part of domain 8:

(Partial domain 4 in bold, partial domain 8 double underlined!

MNCPVLSLGSGFLFQVIEMLIFAYFASISLTESRGLFPRLENVGAFKKVSIVPTQAVCGL PDRSTFC

HSSAAAESIQFCTQRFCIQDCPYRSSHPTYTALFSAGLSSCITPDKNDLHPNAHSNS ASFIFGNHK

SCFSSPPSPKLMASFTLAVWLKPEQQGVMCVIEKTVDGQIVFKLTISEKETMFYYRT VNGLQPPI

KVMTLGRILVKKWIHLSVQVHQTKISFFINGVEKDHTPFNARTLSGSITDFASGTVQ IGQSLNGLE

QFVGRMQDFRLYQVALTNREILEVFSGDLLRLHAQSHCRCPGSHPRVHPLAQRYCIP NDAGDT

ADNRVSRLNPEAHPLSFVNDNDVGTSWVSNVFTNITQLNQGVTISVDLENGQYQVFY IIIQFFS

PQPTEIRIQRKKENSLDWEDWQYFARNCGAFGMKNNGDLEKPDSVNCLQLSNFTPYS RGNVT

FSILTPGPNYRPGYNNFYNTPSLQEFVKATQIRFHFHGQYYTTETAVNLRHRYYAVD EITISGRCQ

CHGHADNCDTTSQPYRCLCSQESFTEGLHCDRCLPLYNDKPFRQGDQVYAFNCKPCQ CNSHSK

SCHYNISVDPFPFEHFRGGGGVCDDCEHNTTGRNCELCKDYFFRQVGADPSAIDVCK PCDCDT

VGTRNGSILCDQIGGQCNCKRHVSGRQCNQCQNGFYNLQELDPDGCSPCNCNTSGTV DGDIT

CHQNSGQCKCKANVIGFYISPGNATGCLPCSCHTTGAVNHICNSLTGQCVCQDASIA GQRCDQ

CKDHYFGFDPQTGRCQPCNCHLSGALNETCHLVTGQCFCKQFVTGSKCDACVPSASH LDVNNL

LGCSKTPFQQPPPRGQVQSSSAINLSWSPPDSPNAHWLTYSLLRDGFEIYTTEDQYP YSIQYFLD

TDLLPYTKYSYYIETTNVHGSTRSVAVTYKTKPGVPEGNLTLSYIIPIGSDSVTLTW TTLSNQSGPIE

KYILSCAPLAGGQPCVSYEGHETSATIWNLVPFAKYDFSVQACTSGGCLHSLPITVT TAQAPPQR

LSPPKMQKISSTELHVEWSPPAELNGIIIRYELYMRRLRSTKETTSEESRVFQSSGW LSPHSFVESA

NENALKPPQTMTTITGLEPYTKYEFRVLAVNMAGSVSSAWVSERTGESAPVFMIPPS VFPLSSYS

LNISWEKPADNVTRGKVVGYDINMLSEQSPQQSIPMAFSQLLHTAKSQELSYTVEGL KPYRIYEF

TITLCNSVGCVTSASGAGQTLAAAPAQLRPPLVKGINSTTIHLKWFPPEELNGPSPI YQLERRESSL

PALMTTMMKGIRFIGNGYCKFPSSTHPVNTDFTGIKASFRTKVPEGLIVFAASPGNQ EEYFALQL KKGRLYFLFDPQGSPVEVTTTNDHGKQYSDGKWHEIIAIRHQAFGQITLDGIYTGSSAIL NGSTVI

GDNTGVFLGGLPRSYTILRKDPEIIQKGFVGCLKDVHFMKNYNPSAIWEPLDWQSSE EQINVYN

SWEGCPASLNEGAQFLGAGFLELHPYMFHGGMNFEISFKFRTDQLNGLLLFVYNKDG PDFLAM

ELKSGILTFRLNTSLAFTQVDLLLGLSYCNGKWNKVIIKKEGSFISASVNGLMKHAS ESGDQPLVV

NSPVYVGGIPQELLNSYQHLCLEQGFGGCMKDVKFTRGAVVNLASVSSGAVRVNLDG CLSTDS

AVNCRGNDSILVYQGKEQSVYEGGLQPFTEYLYRVIASHEGGSVYSDWSRGRTTGAA PQSVPTP

SRVRSLNGYSIEVTWDEPVVRGVIEKYILKAYSEDSTRPPRMPSASAEFVNTSNLTG ILTGLLPFKN

YAVTLTACTLAGCTESSHALNISTPQEAPQEVQPPVAKSLPSSLLLSWNPPKKANGI ITQYCLYMD

GRLIYSGSEENYTVTDLAVFTPHQFLLSACTHVGCTNSSWVLLYTAQLPPEHVDSPV LTVLDSRTI

HIQWKQPRKISGILERYVLYMSNHTHDFTIWSVIYNSTELFQDHMLQYVLPGNKYLI KLGACTGG

GCTVSEASEALTDEDIPEGVPAPKAHSYSPDSFNVSWTEPEYPNGVITSYGLYLDGI LIHNSSELSY

RAYGFAPWSLHSFRVQACTAKGCALGPLVENRTLEAPPEGTVNVFVKTQGSRKAHVR WEAPFR

PNGLLTHSVLFTGIFYVDPVGNNYTLLNVTKVMYSGEETNLWVLIDGLVPFTNYTVQ VNISNSQ

GSLITDPITIAMPPGAPDGVLPPRLSSATPTSLQVVWSTPARNNAPGSPRYQLQMRS GDSTHGF

LELFSNPSASLSYEVSDLQPYTEYMFRLVASNGFGSAHSSWIPFMTAEDKPGPVVPP ILLDVKSR

M M LVTWQH P RKSN G VITH YN I YLHG RLYLRTPG NVTN CTVM H LH PYTAYKFQVEACTSKGCSL

SPESQTVWTLPGAPEGIPSPELFSDTPTSVIISWQPPTHPNGLVENFTIERRVKGKE EVTTLVTLPR

SHSMRFIDKTSALSPWTKYEYRVLMSTLHGGTNSSAWVEVTTRPSRPAGVQPPVVTV LEPDAV

QVTWKPPLIQNGDILSYEIHMPDPHITLTNVTSAVLSQKVTHLIPFTNYSVTIVACS GGNGYLGG

CTESLPTYVTTHPTVPQNVGPLSVIPLSESYVVISWQPPSKPNGPNLRYELLRRKIQ QPLASNPPE

DLNRWHNIYSGTQWLYEDKGLSRFTTYEYMLFVHNSVGFTPSREVTVTTLAGLPERG ANLTASV

LNHTAIDVRWAKPTVQDLQGEVEYYTLFWSSATSNDSLKILPDVNSHVIGHLKPNTE YWIFISVF

NGVHSINSAGLHATTCDGEPQGMLPPEVVIINSTAVRVIWTSPSNPNGVVTEYSIYV NNKLYKT

GMNVPGSFILRDLSPFTIYDIQVEVCTIYACVKSNGTQITTVEDTPSDIPTPTIRGI TSRSLQIDWVS

PRKPNGIILGYDLLWKTWYPCAKTQKLVQDQSDELCKAVRCQKPESICGHICYSSEA KVCCNGVL

YNPKPGHRCCEEKYIPFVLNSTGVCCGGRIQEAQPNHQCCSGYYARILPGEVCCPDE QHNRVSV

GIGDSCCGRMPYSTSGNQICCAGRLHDGHGQKCCGRQIVSNDLECCGGEEGVVYNRL PGMFC

CGQDYVNMSDTICCSASSGESKAHIKKNDPVPVKCCETELIPKSQKCCNGVGYNPLK YVCSDKIS

TGMMMKETKECRILCPASMEATEHCGRCDFNFTSHICTVIRGSHNSTGKASIEEMCS SAEETIHT

GSVNTYSYTDVNLKPYMTYEYRISAWNSYGRGLSKAVRARTKEDVPQGVSPPTWTKI DNLEDTI

VLNWRKPIQSNGPIIYYILLRNGIERFRGTSLSFSDKEGIQPFQEYSYQLKACTVAG CATSSKVVAA

TTQGVPESILPPSITALSAVALHLSWSVPEKSNGVIKEYQIRQVGKGLIHTDTTDRR QHTVTGLQP

YTNYSFTLTACTSAGCTSSEPFLGQTLQAAPEGVWVTPRHIIINSTTVELYWSLPEK PNGLVSQYQ

LSRNGNLLFLGGSEEQNFTDKNLEPNSRYTYKLEVKTGGGSSASDDYIVQTPMSTPE EIYPPYNIT

VIGPYSIFVAWIPPGILIPEIPVEYNVLLNDGSVTPLAFSVGHHQSTLLENLTPFTQ YEIRIQACQNG

SCGVSSRMFVKTPEAAPMDLNSPVLKALGSACIEIKWMPPEKPNGIIINYFIYRRPA GIEEESVLF

VWSEGALEFMDEGDTLRPFTLYEYRVRACNSKGSVESLWSLTQTLEAPPQDFPAPWA QATSAH

SVLLNWTKPESPNGIISHYRVVYQERPDDPTFNSPTVHAFTVKGTSHQAHLYGLEPF TTYRIGVV

AANHAGEILSPWTLIQTLESSPSGLRNFIVEQKENGRALLLQWSEPMRTNGVIKTYN IFSDGFLEY

SGLNRQFLFRRLDPFTLYTLTLEACTRAGCAHSAPQPLWTDEAPPDSQLAPTVHSVK STSVELSW

SEPVNPNGKIIRYEVIRRCFEGKAWGNQTIQADEKIVFTEYNTERNTFMYNDTGLQP WTQCEYK

IYTWNSAGHTCSSWNVVRTLQAPPEGLSPPVISYVSMNPQKLLISWIPPEQSNGIIQ SYRLQRNE

MLYPFSFDPVTFNYTDEELLPFSTYSYALQACTSGGCSTSKPTSITTLEAAPSEVSP PDLWAVSAT

QMNVCWSPPTVQNGKITKYLVRYDNKESLAGQGLCLLVSHLQPYSQYNFSLVACTNG GCTASV

SKSAWTMEALPENMDSPTLQVTGSESIEITWKPPRNPNGQIRSYELRRDGTIVYTGL ETRYRDFT

LTPGVEYSYTVTASNSQGGILSPLVKDRTSPSAPSGMEPPKLQARGPQEILVNWDPP VRTNGDII

NYTLFIRELFERETKIIHINTTHNSFGMQSYIVNQLKPFHRYEIRIQACTTLGCASS DWTFIQTPEIA PLMQPPPHLEVQMAPGGFQPTVSLLWTGPLQPNGKVLYYELYRRQIATQPRKSNPVLIYN GSS

TSFIDSELLPFTEYEYQVWAVNSAGKAPSSWTWCRTGPAPPEGLRAPTFHVISSTQA VVNISAP

GKPNGIVSLYRLFSSSAHGAETVLSEGMATQQTLHGLQAFTNYSIGVEACTCFNCCS KGPTAELR

THPAPPSGLSSPQIGTLASRTASFRWSPPMFPNGVIHSYELQFHVACPPDSALPCTP SQIETKYT

GLGQKASLGGLQPYTTYKLRVVAHNEVGSTASEWISFTTQKELPQYRAPFSVDSNLS VVCVNWS

DTFLLNGQLKEYVLTDGGRRVYSGLDTTLYIPRTADKTFFFQVICTTDEGSVKTPLI QYDTSTGLGL

VLTTPGKKKGSRSKSTEFYSELWFIVLMAMLGLILLAIFLSLILQRKIHKEPYIRER PPLVPLQKRMS

PLNVYPPGENHMGLADTKIPRSGTPVSIRSNRSACVLRIPSQNQTSLTYSQGSLHRS VSQLMDIQ

DKKVLMDNSLWEAIMGHNSGLYVDEEDLMNAIKDFSSVTKERTTFTDTHL (SEQID NO:2)

REFERENCES

1. Hartong, D.T., E.L. Berson, and T.P. Dryja, Retinitis pigmentosa. Lancet, 2006.368(9549): p.1795-809.

2. Saihan, Z., et al, Update on Usher syndrome. Curr Opin Neurol, 2009. 22(1): p.19-27.

3. Lentz, J. and B. Keats, Usher Syndrome Type II, in GeneReviews®, R.A. Pagon, et al., Editors.1993, University of Washington, Seattle: Seattle WA.

4. Sandberg, M.A., et al, Disease course in patients with autosomal recessive retinitis pigmentosa due to the USH2A gene. Invest Ophthalmol Vis Sci, 2008.49(12): p.5532-9.

5. Maguire, A.M., et al, Safety and efficacy of gene transfer for Leber's congenital amaurosis. The New England journal of medicine, 2008.358(21): p.2240- 8

6. Bainbridge, J.W., et al., Effect of gene therapy on visual function in Leber's congenital amaurosis. N.Engl.J.Med., 2008.358(21): p.2231-2239.

7. Cideciyan, A.V., et al, Human gene therapy for RPE65 isomerase deficiency activates the retinoid cycle of vision but with slow rod kinetics.

Proc.Nat.Acad.Sci., 2008.105(39): p.15112-7.

8. Maguire, A.M., et al, Age-dependent effects of RPE65 gene therapy for Leber's congenital amaurosis: a phase 1 dose-escalation trial. Lancet, 2009.

374(9701): p.1597-605.

9. Jacobson, S.G., et al, Gene therapy for leber congenital amaurosis caused by RPE65 mutations: safety and efficacy in 15 children and adults followed up to 3 years. Archives of Ophthalmology, 2012.130(1): p.9-24.

10. Bennett, J., et al., AAV2 gene therapy readministration in three adults with congenital blindness. Science translational medicine, 2012.4(120): p. !20ral5. 11. Bowles, D.E., et al, Phase 1 gene therapy for Duchenne muscular dystrophy using a translational optimized AAV vector. Molecular therapy : the journal of the American Society of Gene Therapy, 2012. 20(2): p. 443-55.

12. Maclachlan, T.K., et al, Preclinical safety evaluation of AAV2- sFLTOl- a gene therapy for age-related macular degeneration. Molecular therapy : the journal of the American Society of Gene Therapy, 2011. 19(2): p. 326-34.

13. Nathwani, A.C., et al, Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. The New England journal of medicine, 2011. 365(25): p. 2357-65.

14. Ran, F. A., et al., In vivo genome editing using Staphylococcus aureus

Cas9. Nature, 2015. 520(7546): p. 186-91.

15. Swiech, L., et al, In vivo interrogation of gene function in the mammalian brain using CRISPR-Cas9. Nat Biotechnol, 2015. 33(1): p. 102-6.

16. van Wijk, E., et al, Identification of 51 novel exons of the Usher syndrome type 2A (USH2A) gene that encode multiple conserved functional domains and that are mutated in patients with Usher syndrome type II. Am J Hum Genet, 2004. 74(4): p. 738-44.

17. Grati, M., et al, Localization of PDZD7 to the stereocilia ankle-link associates this scaffolding protein with the Usher syndrome protein network. J Neurosci, 2012. 32(41): p. 14288-93.

18. Liu, X., et al, Usherin is required for maintenance of retinal photoreceptors and normal development of cochlear hair cells. Proc Natl Acad Sci U S A, 2007. 104(11): p. 4413-8.

19. Aller, E., et al, The USH2A c.2299delG mutation: dating its common origin in a Southern European population. Eur J Hum Genet, 2010. 18(7): p. 788-93.

20. Yan, D., et al, Mutation analysis in the long isoform of USH2A in American patients with Usher Syndrome type II. J Hum Genet, 2009. 54(12): p. 732- 8

21. Kemaladewi, D.U. and R.D. Cohn, Exon Snipping in Duchenne Muscular Dystrophy. Trends Mol Med, 2016. 22(3): p. 187-9.

22. Long, C., et al., Postnatal genome editing partially restores dystrophin expression in a mouse model of muscular dystrophy. Science, 2016. 351(6271): p. 400-3. 23. Nelson, C.E., et al, In vivo genome editing improves muscle function in a mouse model of Duchenne muscular dystrophy. Science, 2016. 351(6271): p. 403-7.

24. Tabebordbar, M., et al, In vivo gene editing in dystrophic mouse muscle and muscle stem cells. Science, 2016. 351(6271): p. 407-11.

25. Young, C.S., et al, A Single CRISPR-Cas9 Deletion Strategy that Targets the Majority of DMD Patients Restores Dystrophin Function in hiPSC- Derived Muscle Cells. Cell Stem Cell, 2016. 18(4): p. 533-40.

26. Kalinec, F., et al., Establishment and characterization of conditionally immortalized organ of corti cell lines. Cell Biol Int, 1999. 23(3): p. 175-84.

27. Millan, J.M., et al, An update on the genetics of usher syndrome. J Ophthalmol, 2011. 2011: p. 417217.

28. Lenassi, E., et al, A detailed clinical and molecular survey of subjects with nonsyndromic USH2A retinopathy reveals an allelic hierarchy of disease- causing variants. Eur J Hum Genet, 2015. 23(10): p. 1318-27.

29. Suzuki, K., et al., In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration. Nature, 2016. 540(7631): p. 144-149.

30. Zou, J., et al, Whirlin replacement restores the formation of the USH2 protein complex in whirlin knockout photoreceptors. Invest Ophthalmol Vis Sci, 2011. 52(5): p. 2343-51.

31. Zou, J., et al, Deletion of PDZD7 disrupts the Usher syndrome type 2 protein complex in cochlear hair cells and causes hearing loss in mice. Hum Mol Genet, 2014. 23(9): p. 2374-90.

32. Davis, E.E., et al, TTC21B contributes both causal and modifying alleles across the ciliopathy spectrum. Nat Genet, 2011. 43(3): p. 189-96.

33. Zhang, Q., et al, Knockdown of ttc26 disrupts ciliogenesis of the photoreceptor cells and the pronephros in zebrafish. Mol Biol Cell, 2012. 23(16): p. 3069-78.

34. Wang, H., et al, One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell, 2013. 153(4): p.

910-8.

35. Inui, M., et al, Rapid generation of mouse models with defined point mutations by the CRISPR/Cas9 system. Sci Rep, 2014. 4: p. 5396. 36. Mashiko, D., et al, Feasibility for a large scale mouse mutagenesis by injecting CRISPR/Cas plasmid into zygotes. Dev Growth Differ, 2014. 56(1): p. 122- 9.

37. Lu, B., et al, Cell transplantation to arrest early changes in an ush2a animal model. Invest Ophthalmol Vis Sci, 2010. 51(4): p. 2269-76.

38. Liu, Q., J. Zuo, and E.A. Pierce, The retinitis pigmentosa 1 protein is a photoreceptor microtubule-associated protein. J Neurosci, 2004. 24(29): p. 6427-36.

39. Cheng, A.W., et al, Multiplexed activation of endogenous genes by CRISPR-on, an RNA-guided transcriptional activator system. Cell Res, 2013. 23(10): p. 1163-71.

40. Ousterout, D.G., et al, Multiplex CRISPR/Cas9-based genome editing for correction of dystrophin mutations that cause Duchenne muscular dystrophy. Nat Commun, 2015. 6: p. 6244.

41. Platt, R.J., et al., CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell, 2014. 159(2): p. 440-55.

42. Senis, E., et al, CRISPR/Cas9-mediated genome engineering: an adeno-associated viral (AAV) vector toolbox. Biotechnol J, 2014. 9(11): p. 1402-12.

43. Tsai, S.Q., et al, GUIDE-seq enables genome-wide profiling of off- target cleavage by CRISPR-Cas nucleases. Nat Biotechnol, 2015. 33(2): p. 187-97.

44. Tsai, S.Q., et al., Open-source guideseq software for analysis of

GUIDE-seq data. Nat Biotechnol, 2016. 34(5): p. 483.

45. Matsuda, T. and C.L. Cepko, Controlled expression of transgenes introduced by in vivo electroporation. Proc Natl Acad Sci U S A, 2007. 104(3): p. 1027-32.

46. Jelcick, A.S., et al, Genetic variations strongly influence phenotypic outcome in the mouse retina. PLoS One, 2011. 6(7): p. e2l858.

47. Liu, Q., A. Saveliev, and E.A. Pierce, The severity of retinal degeneration in Rplh gene-targeted mice is dependent on genetic background. Invest Ophthalmol Vis Sci, 2009. 50(4): p. 1566-74.

48. Vijayakumar S, Depreux FF, Jodelka FM, et al. Rescue of peripheral vestibular function in Usher syndrome mice using a splice-switching antisense oligonucleotide. Hum Mol Genet. 2017 Sep 15;26(18):3482-3494. 49. Slijkerman RW, Vache C, Dona M, et al. Antisense Oligonucleotide- based Splice Correction for USH2A-associated Retinal Degeneration Caused by a Frequent Deep-intronic Mutation. Mol Ther Nucleic Acids. 2016 Nov l;5(l0):e38l.

50. Annemieke Aartsma-Rus, et al. Theoretic applicability of antisense- mediated exon skipping for Duchenne muscular dystrophy. Hum Mutat. 2009 Mar;30 (3):293-9.

51. Beroud C, et al. Multi exon skipping leading to an artificial DMD protein lacking amino acids from exons 45 through 55 could rescue up to 63% of patients with Duchenne muscular dystrophy. Hum Mutat. 2007 Feb;28(2): 196-202.

OTHER EMBODIMENTS

It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.