PreprintPDF Available

Targeted genome editing restores auditory function in adult mice with progressive hearing loss caused by a human microRNA mutation

Authors:
Preprints and early-stage research may not have been peer reviewed yet.

Abstract

Mutations in microRNA-96 (MIR96) cause dominant delayed onset hearing loss DFNA50 without treatment. Genome editing has shown efficacy in hearing recovery by intervention in neonatal mice, yet editing in the adult inner ear is necessary for clinical applications. Here, we developed an editing therapy for a C>A point mutation in the seed region of the Mir96 gene, Mir9614C>A associated with hearing loss by screening gRNAs for genome editors and optimizing Cas9 and sgRNA scaffold for efficient and specific mutation editing in vitro. By AAV delivery in pre-symptomatic (3-week-old) and symptomatic (6-week-old) adult Mir9614C>A mutant mice, hair cell on-target editing significantly improved hearing long-term, with an efficacy inversely correlated with injection age. We achieved transient Cas9 expression without the evidence of AAV genomic integration to significantly reduce the safety concerns associated with editing. We developed an AAV-sgmiR96-master system capable of targeting all known human MIR96 mutations. As mouse and human MIR96 sequences share 100% homology, our approach and sgRNA selection for efficient and specific hair cell editing for long-term hearing recovery lays the foundation for future treatment of DFNA50 caused by MIR96 mutations.
Targeted genome editing restores auditory function in adult mice with 1
progressive hearing loss caused by a human microRNA mutation 2
3
Wenliang Zhu1,2,8, Wan Du1,2,8, Arun Prabhu Rameshbabu1,2, Ariel Miura Armstrong1,2, 4
Stewart Silver1,2, Yehree Kim1,2, Wei Wei1,2, Yilai Shu3,4,5, Xuezhong Liu6, Morag A 5
Lewis7, Karen P. Steel7, Zheng-Yi Chen1, 2 * 6
7
1 Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech 8
and Hearing Bioscience and Technology and Program in Neuroscience, Harvard 9
Medical School, Boston, USA. 10
2 Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, USA. 11
3 ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key 12
Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, 13
Fudan University, Shanghai, 200031, China. 14
4 Institutes of Biomedical Science, Fudan University, Shanghai, 200032, China. 15
5 NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, 16
China. 17
6 Department of Otolaryngology, University of Miami School of Medicine, Miami, FL, 18
33136, USA. 19
7 Wolfson Sensory, Pain and Regeneration Centre, King’s College London, London, UK. 20
8 These authors contributed equally: Wenliang Zhu, Wan Du 21
* Corresponding author. Email: zheng-yi_chen@meei.harvard.edu (Z.C.) 22
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Abstract 23
24
Mutations in microRNA-96 (MIR96) cause dominant delayed onset hearing loss 25
DFNA50 without treatment. Genome editing has shown efficacy in hearing recovery by 26
intervention in neonatal mice, yet editing in the adult inner ear is necessary for clinical 27
applications. Here, we developed an editing therapy for a C>A point mutation in the 28
seed region of the Mir96 gene, Mir9614C>A associated with hearing loss by screening 29
gRNAs for genome editors and optimizing Cas9 and sgRNA scaffold for efficient and 30
specific mutation editing in vitro. By AAV delivery in pre-symptomatic (3-week-old) and 31
symptomatic (6-week-old) adult Mir9614C>A mutant mice, hair cell on-target editing 32
significantly improved hearing long-term, with an efficacy inversely correlated with 33
injection age. We achieved transient Cas9 expression without the evidence of AAV 34
genomic integration to significantly reduce the safety concerns associated with editing. 35
We developed an AAV-sgmiR96-master system capable of targeting all known human 36
MIR96 mutations. As mouse and human MIR96 sequences share 100% homology, our 37
approach and sgRNA selection for efficient and specific hair cell editing for long-term 38
hearing recovery lays the foundation for future treatment of DFNA50 caused by MIR96 39
mutations. 40
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
INTRODUCTION 41
42
Hearing loss is a multi-factorial condition affecting a significant portion of the global 43
population (World Health Organization, http://www.who.int/en/). Single genetic 44
mutations causing hearing loss account for more than 50% of all congenital 45
sensorineural hearing loss (SNHL)1, yet no pharmaceutical drug or biological treatment 46
is available to slow down or reverse genetic deafness2-4. Among these deafness genes, 47
microRNAs (miRNAs), which are short (~20–24nt) endogenous non-coding RNAs that 48
play a crucial role in the regulation of the expression of protein-coding genes, are 49
considered important factors in the development of the inner ear and are required for 50
normal hearing5,6. Specifically, mutations in MIR96 (microRNA-96; miR-96) have been 51
identified as causative factors for non-syndromic progressive hearing loss DFNA50 in 52
humans and mice7-11. This mutation in the seed region of the human MIR96 is the first 53
example of point mutations in the mature sequence of a miRNA with an etiopathogenic 54
role in a human Mendelian disease6,11,12. 55
MIR96 is specifically expressed in the sensory cells of the inner ear13 and plays a critical 56
role in cochlear development and the maintenance of hearing by reducing expression of 57
the mRNAs of multiple target genes8,14-16. Point mutations in the seed region of MIR96 58
lead to late onset progressive hearing loss with dominant inheritance patterns in human 59
families10,11. Mice carrying an ENU-induced mutation, diminuendo (Dmdo), exhibit the 60
phenotype of progressive hearing loss17. Homozygous Dmdo mice (Mir96Dmdo/Dmdo) and 61
homozygous null mice (knockout of both Mir96 and Mir183, Mir96dko/dko) are completely 62
deaf, displaying abnormal hair cell stereocilia bundles and early-onset reduced hair cell 63
numbers, demonstrating the requirement for Mir96 in normal hair cell developmental8,18. 64
In contrast, heterozygous Mir96 null mice (Mir96/dko-) maintain normal hearing, while 65
heterozygous Dmdo mice (Mir96Dmdo/+ ) develop adult-onset non-syndromic progressive 66
hearing loss, suggesting that the hearing loss phenotype arises from the gain of novel 67
target genes rather than loss of function18. In humans, heterozygous MIR96 mutations 68
also result in late onset non-syndromic progressive hearing loss, suggesting a similar 69
gain of function due to the mutation. Delayed onset and progressive hearing loss offers 70
a window of opportunity for intervention for hearing rescue. 71
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
In addition to its dominant inheritance pattern, overexpression of Mir96 has been found 72
to cause cochlear defects leading to hearing loss, rendering traditional gene therapy via 73
ectopic Mir96 expression unsuitable9. However, genome editing technologies that target 74
the dominant MIR96 mutations hold great promise to treat DFNA50. In vivo Cas9 75
nuclease delivery has demonstrated effectiveness in disrupting dominant alleles in 76
multiple hearing loss disease models, including Lipofectamine RNP delivery19,20 or 77
adeno-associated virus (AAV)-mediated delivery21-26. Inner ear base editing has also 78
been employed to repair a Tmc1 dominant mutation in mice27. In all the editing studies 79
of genetic hearing loss, the interventions were done in neonatal mice with an immature 80
cochlea28. In contrast, newborn human inner ears are fully developed29. The mouse 81
cochlea undergoes significant developmental changes from the neonatal to adult 82
stages, including changes in size, structure, and function28. This distinction highlights 83
the need to evaluate the efficacy, safety, and time window of genome editing treatment 84
in the mature cochlea to establish the feasibility of intervention for potential clinical 85
applications. 86
In this study, we performed editing by NHEJ to target a dominant Mir96 mutation 87
(Mir96tm3.1Wtsi, referred to as Mir9614C>A in this report; equivalent of human rs587776523, 88
g.7:129414596G>T in GRCh38) by AAV-mediated adult delivery in mice with hearing 89
loss. We showed that AAV-mediated adult delivery of the editing complex was well-90
tolerated without impairing normal hearing. We screened sgRNAs for different editors 91
and identified one for KKH-saCas9 for further modification, resulting in efficient and 92
specific editing in vitro and in vivo. We tested interventions before and after the onset of 93
hearing loss and established a time window when the treatment results in sustained and 94
significant hearing preservation. To improve the safety profile of the editing strategy, we 95
established the dosing without detectable AAV integrations and showed the lack of 96
KKH-saCas9 expression 12 weeks post injection. As human and mouse Mir96 share 97
100% sequences in the seed region, we developed a dual AAV system capable of 98
targeting multiple human MIR96 mutations. We successfully demonstrated the 99
efficiency and specificity of this dual AAV system in MIR96 mutant human cell lines, 100
providing evidence for its potential generalization and broad utility. In conclusion, our 101
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
study establishes the feasibility of in vivo genome editing as a viable approach for 102
treating hearing loss caused by genetic mutations in adult animals. 103
104
RESULTS 105
106
A human microRNA-96 mutation Mir9614C>A causes dominant adult-onset 107
progressive hearing loss in mice. MiR-96 is highly expressed in mammalian sensory 108
cells, including the inner ear, retina, and dorsal root ganglia, and functions as a master 109
regulator controlling expression of many genes8,17. Mir96 is super conserved across 110
vertebrates, from zebrafish to humans, with the nucleotides within the Mir96 seed region 111
showing 100% identity in DNA sequence (Fig.1a). 112
Two mutations in the seed region, +13G >A (rs587776522) and +14C >A 113
(rs587776523), have been identified as the cause of autosomal dominant non-114
syndromic progressive hearing loss DFNA5011. Our mouse model carries the Mir9614C>A 115
mutation, which substitutes the fourteenth nucleotide of cytosine in the conserved seed 116
region with adenine30 (Fig.1a, b). To evaluate how hearing is affected by the mutation, 117
we performed auditory brain stem response (ABR) and distortion product otoacoustic 118
emission (DPOAE) tests on Mir9614C>A mice (Fig. 1c-h). Mir9614C>A/14C>A mice exhibited 119
complete hearing loss across all frequencies and age groups by ABR and DPOAE at 4 120
weeks of age (Fig. 1c, f). Mir9614C>A/+ mice at 4 weeks of age, we observed a 30 dB 121
elevation in ABR and a 19 dB elevation in DPOAE at the frequency of 32 kHz but no 122
significant changes in ABR/DPOAE thresholds at other frequencies compared to wild-123
type (WT) ears (Fig. 1c, f), indicating the onset of hearing loss starting at 4 weeks at 32 124
kHz. By 8 weeks, the ABR thresholds were significantly elevated from 16 to 32 kHz, 125
with an average increase of 20 dB in Mir9614C>A/+ ears compared to WT ears. Similarly, 126
DPOAE thresholds were significantly elevated by an average of 16 dB from 11.3 to 22.6 127
kHz frequencies Mir9614C>A/+ ears compared to WT ears (Fig. 1g). At 12 weeks, the 128
hearing loss became more severe, as ABR thresholds were significantly elevated 129
across all frequencies when compared to WT ears, with an average elevation of 15 dB. 130
Additionally, DPOAE thresholds were significantly elevated 15 dB on average compared 131
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
to WT ears, particularly at 11.3 and 16 kHz (Fig. 1h). These results showed that 132
Mir9614C>A/+ mice exhibit adult-onset hearing loss starting at 4 weeks at the high 133
frequency of 32kHz, which becomes progressive with further elevation of ABR/DPOAE 134
thresholds at 8 and 12 weeks across other frequencies. 135
136
Screening of genome editing systems in mouse and human cells. To specifically 137
disrupt the Mir9614C>A allele, sgRNAs were designed to target the allele for each of five 138
types of CRISPR nucleases: spCas931, scCas9++32, KKH-saCas933, sauriCas934, LZ3 139
Cas935 (Fig. 2a). All Cas9/sgRNA combinations were in the same pMAX-Cas9/sgRNA 140
backbone with three nuclear localization signals (NLSs) to ensure consistency in 141
plasmid structure (Fig. 2b and Extended Data Fig. 1a). Using primary Mir9614C>A/+ 142
fibroblasts, spCas9/sgRNA-1 exhibited the highest editing efficiency (14.3± 2.3%) (Fig. 143
2c, d). LZ3 Cas9/sgRNA-1 and KKH-saCas9/sgRNA-4 also showed comparable editing 144
efficiency (12.7± 2.0% and 10.1± 2.1%), while scCas9++/sgRNA-1, scCas9++/sgRNA-2 145
and sauriCas9/sgRNA-4 displayed modest indel frequency (Fig. 2c, d). spCas9/sgRNA-146
1 and KKH-saCas9/sgRNA-4 exhibited high specificity in targeting the Mir9614C>A allele, 147
shown by next generation sequencing (NGS) analysis that all the indels occurred 148
exclusively at the mutant allele and were undetectable in Mir96+/+ cells (less than 149
0.03%) (Fig. 2d-f and Extended Data Fig. 1b). We compared the editing efficiency of 150
Cas9/sgRNA ribonucleoprotein (RNP) and plasmids in primary fibroblasts of Mir9614C>A/+ 151
mice. RNP nucleofection yielded the highest editing efficiency, while lipofectamine-152
mediated RNP delivery was less efficient (Extended Data Fig. 1c). The AAV systems 153
with spCas9/sgRNA1 dual AAV plasmids (7.3± 1.5%) and KKH-saCas9/sgRNA4 (10.4± 154
1.8%) single AAV plasmid exhibited editing frequencies comparable to the pMAX-Cas9 155
plasmid transfection (14.3± 2.3%) (Extended Data Fig. 1c). 156
Due to 100% homology in the Mir96 seed region across species, the sgRNAs design 157
targeting the Mir9614C>A allele in mice can be used in the human MIR96 with the same 158
mutation. To validate the editing systems in human cells, we generated human and 159
mouse cell lines carrying the Mir9614C>A mutation using the PiggyBac transposon 160
system (Fig. 2g). In cells transfected with spCas9/sgRNA-1 and KKH-saCas9/sgRNA-4, 161
NGS analysis showed a high level of indel formation in both human and mouse Mir96 162
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
+14C>A cell lines, where negligible indels were observed in the Mir96 wild-type cell 163
lines (Fig. 2h). The indel profile showed various types of indels for spCas9/sgRNA-1 164
and KKH-saCas9/sgRNA-4, with single nucleotide deletions (-1 and +1) being the most 165
common type for spCas9/sgRNA-1, and a six-nucleotide deletion being the most 166
common for KKH-saCas9/sgRNA-4 (Fig. 2i, j). 167
In addition to disrupting the mutated allele with CRISPR nucleases, we also attempted 168
to correct the Mir9614C>A mutation using prime editing36-38(Extended Data Fig. 2a). We 169
designed a pegRNA located upstream of the mutate nucleotide, containing pegRNA-170
extensions with 13 bp primer binding sites (PBSs) and 16 bp RT-templates harboring 171
the corrected sequence (Extended Data Fig. 2b). We transfected HEI-OC1 cells 172
carrying the Mir96 +14C>A mutation with plasmids encoding primer editors and 173
pegRNA. NGS analysis showed that prime editing successfully corrected the mutation 174
in Mir9614C>A cells with an efficiency of 2.8±1.4% (Extended Data Fig. 2c, d). However, 175
due to the modest efficiency of prime editing at this locus and the lack of an efficient in 176
vivo delivery system for the inner ear, CRISPR nuclease-mediated knock-out of the 177
mutated allele is a more suitable choice for treating hearing loss caused by the Mir96 178
mutation. 179
180
Optimization of CRISPR/sgRNA delivery system for mature cochlea in adult mice. 181
We chose AAV serotype 2 (AAV2), which has demonstrated high efficiency and 182
specificity in transducing both mature inner hair cells (IHCs) and outer hair cells (OHCs) 183
compared to other AAV serotypes39,40, making AAV2 an excellent candidate vehicle for 184
delivering Cas9/sgRNA to hair cells. To further assess the delivery efficiency of AAV2, 185
we initially delivered AAV2 carrying a GFP cassette into the adult cochlea through round 186
window membrane and canal fenestration (RWM+CF) injection in 18-week-old mice 187
(Extended Data Fig. 3a). Four weeks after injection, robust GFP expression was 188
observed in nearly all IHCs throughout the cochlear turns and robust OHC transduction 189
with an apex-to-base gradient (Extended Data Fig. 3a-d). Importantly, the majority of 190
GFP-positive cells were hair cells, with only limited GFP+ cells observed in other cell 191
types, including supporting cells, indicating that AAV2 specifically and efficiently targets 192
auditory hair cells in adult mice. 193
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
To maximize editing efficiency in vivo to improve hearing rescue, we modified AAV 194
Cas9/sgRNA construction by incorporating a bipartite nuclear localization signal 195
(bpNLS), an sv40 NLS at the N-termini, and another bpNLS at the C-termini (Fig. 3a) to 196
enhance nuclear import and their access to genomic DNA41,42. The KKH-saCas9 197
sgRNAs were optimized by using an 84 nt sgRNA (crRNA-tracrRNA duplex extension) 198
which exhibits higher activity than the canonical full length sgRNA33. Additionally, we 199
introduced a 3rd A-U flip in the stem loop to remove a putative RNA Pol III terminator 200
sequence (4 consecutive U’s), further enhancing expression levels under the U6 201
promoter43 (Fig. 3b, c). The optimized construct was tested in the primary fibroblasts 202
derived from Ai14 mice, which contain “CAG-STOP-tdTomato” cassette44. In the cells 203
transfected with the optimized KKH-saCas9/sgRNA constructs, an increase in editing 204
efficiency of approximately 3.05-fold and 1.98-fold was detected for KKH-saCas9/sgtdT-205
1 and KKH-saCas9/sgtdT-2 compared to the original designs, respectively, shown by 206
the tdT+ cells (Fig. 3d, e). The editing efficiency was also evaluated in HEK-GFP cells 207
using KKH-Cas9/sgGFP targeting GFP, and the ratio of GFP-negative cells showed 208
significantly higher efficiency when using the optimized AAV constructions compared to 209
the original design (Extended Data Fig. 3e, f). The optimized AAV plasmid design and 210
gRNA modifications were used for the production of AAV-KKH-saCas9/sgRNA-4 for the 211
in vivo editing study. 212
213
In vivo genome editing of Mir9614C>A allele in mature cochlea. KKH-saCas9/sgRNA-214
4 was packaged into AAV2 (Fig. 4a) and delivered to adult Mir9614C>A/+ cochlea (6 215
weeks old) via round window membrane and canal fenestration (RWM+CF) injection at 216
a dose of 6×109 vg (Fig. 4a). Cochleae were collected 4- and 8-weeks post-injection for 217
NGS (Fig. 4b). In vivo NGS of cochlear tissue detected the presence of indels at the 218
Mir96 mutant locus in the injected ear at 4 and 8 weeks, while no indels were detected 219
in the contralateral uninjected ears (Fig. 4c, d and Extended Data Fig. 4a-c). 220
Importantly, AAV2-KHH-SaCas9-sgRNA-4 specifically edited the Mir9614C>A allele, as 221
no indels were observed in AAV2-KKH-saCas9/sgRNA-4 treated wild-type (WT) mice or 222
AAV2-KKH-saCas9/sgCtrl treated Mir9614C>A/+ mice (Fig. 4d and Extended Data Fig. 4a-223
c). After 8 weeks of injection, the indel rate was determined to be 0.76 ± 0.13%, slightly 224
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
higher than the rate observed 1 month after injection (0.63 ± 0.14%) (Fig. 4d). However, 225
the indel frequency from organ of Corti samples does not accurately reflect the editing 226
efficiency in hair cells, which constitutes less than 5% of cochlear cells. 227
To obtain a precise measurement of editing efficiency in hair cells, we performed 228
injection and assessed isolated hair cells after labeling hair cells with the FM1-43 229
uptake assay for NGS assay. FM1-43 and its fixable analog FM1-43FX can specifically 230
label hair cells by passing through open mechanotransduction channels45,46 (Extended 231
Data Fig. 4d). The labeled cells were hand-picked under an inverted fluorescent 232
microscope, lysed, with DNA extracted for PCR and NGS to study indels (Fig. 4e). 233
Indel-containing Mir96 sequencing reads from injected Mir9614C>A/+ mice allowed us to 234
directly assess the allele specificity of Mir9614C>A allele in vivo. Robust indel formation 235
was observed in isolated hair cells from injected animals 8 weeks after treatment, with 236
an indel frequency of 16.3-25.73% in the Mir9614C>A allele and a wider range of indel 237
types including -2bp, -4bp, +1bp, -1bp, compared to the indels observed in cochlear 238
samples (Fig. 4f, g), indicating efficient targeted genome editing in hair cells. From three 239
independent experiments, the ratio of Mir96 wild-type allele reads to Mir9614C>A allele 240
reads was analyzed based on NGS results after editing. In the injected ear, the ratio 241
between the Mir9614C>A allele that combined unedited with indel-containing reads and 242
that of Mir96 wild-type reads was 52.3%:47.7% (Fig. 4h), similar to that observed in 243
uninjected ears (Extended Data Fig. 4e), indicating no noticeable chromatin lesion, 244
insertion or large deletions caused by genome editing. 245
246
In vivo genome editing improved short- and long-term hearing preservation. We 247
injected AAV2-KKH-saCas9-sgRNA-4 into the cochlea of 6-week-old (P40-P45) 248
Mir9614C>A/+ mice through the round window membrane (RWM) with canal fenestration, 249
with the contralateral uninjected ears serving as controls. 10 weeks after the injection, at 250
16 weeks of age, the injected ears showed overall lower auditory brainstem response 251
(ABR) thresholds compared to the uninjected control ears, at frequencies from 5.6 to 16 252
kHz (Fig. 5b). The ABR threshold reduction ranged from 12 dB at 5.6 kHz to 18 dB at 16 253
kHz for frequencies below 22.6 kHz, with an average reduction of 13 dB. Additionally, 254
lower distortion product otoacoustic emission (DPOAE) thresholds were observed in the 255
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
injected ears compared to the control ears at frequencies of 11.3 and 16 kHz, with 256
reductions of 14 dB to 21 dB, respectively (Fig. 5c). 257
To assess long-term auditory function following hair cell-targeted genome editing, 258
hearing tests were conducted over an extended period. 14 weeks after injection (i.e., 20 259
weeks of age), significantly lower ABR thresholds were detected in injected ears 260
compared to the uninjected control ears at frequencies of 5.6, 8, 11.3, and 16 kHz, with 261
an average reduction of 21 dB in ABR thresholds. At the frequencies of 8 and 11.3 kHz, 262
the reduction is 21 and 26 dB, respectively (Fig. 5d). Significant reductions in DPOAE 263
thresholds were observed in the injected ears compared to the uninjected control ears 264
at the frequencies of 11.3 and 16 kHz, with reductions at 15 dB and 19 dB, respectively 265
(Fig. 5e). 20 weeks post injection (26 weeks age), more dramatic lower ABR thresholds 266
were detected in the injected ears compared to the uninjected control ears from 5.6 to 267
16 kHz, with an average reduction of 28 dB in ABT thresholds (Fig. 5f). For the 268
frequencies of 5.6 and 8 kHz, the ABR thresholds were reduced by 33 dB and 34 dB, 269
respectively. Similarly, dramatic lower DPOAE thresholds were detected in injected ears 270
compared with uninjected control ears at frequencies of 11.3 and 16 kHz, with 271
reductions of 27 dB and 7 dB at respectively (Fig. 5g). Representative ABR waveforms 272
recorded from an AAV2-KKH-saCas9-sgRNA-4 injected ear (Fig. 5h-j) and the 273
contralateral uninjected ear (Fig. 5i-k) 14 weeks post injection using 11.3 kHz (Fig. 5h-i) 274
and 16 kHz (Fir. 5j-k) tone bursts at incrementally increasing sound pressure levels 275
(SPLs) from 20 to 100 dB (Fig. 5h-k) further illustrated the improved hearing 276
preservation. Collectively, these results demonstrate that AAV2-mediated genome 277
editing effectively abolished the expression of the mutant Mir96, leading to improved 278
short- and long-term hearing preservation. 279
Delayed onset progressive hearing loss caused by MIR96 mutations offers a window of 280
opportunity for intervention. To determine the optimal time window for genome editing 281
treatment in Mir9614C>A/+ mice, we further performed AAV2-KKH-saCas9-sgRNA-4 282
injections in 3-week-old mice in which initial hearing loss was detected only at the high 283
frequency and in 16-week-old mice in which severe hearing loss was detected in all 284
frequencies (Fig. 1c, f; Extended Data Fig. 5a). For 3-week-old injection, significantly 285
lower ABR thresholds were detected 13 weeks post injection in AAV2-KKH-saCas9-286
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
sgRNA-4 treated ears compared to the untreated control ears, with an average ABR 287
threshold reduction of 19 dB from 5.6 to 16 kHz (Extended Data Fig. 5b). A lower ABR 288
threshold of 8 dB was seen at 22.6 kHz although the reduction is not statistically 289
significant. The average ABR threshold reduction in 3-week-old Mir9614C>A/+ mouse 290
injection was greater than that in 6-week-old Mir9614C>A/+ mouse injection, 19 dB vs. 13 291
dB. Significantly lower DPOAE thresholds were also detected in the AAV2-KKH-292
saCas9-sgRNA-4 treated ears compared to the contralateral control ears at frequencies 293
of 11.3 and 16 kHz, with a reduction of 31 dB at 11.3 kHz and 24 dB at 16 kHz. Lower 294
DPOAE threshold of 9 dB was detected at 22.6 kHz; however, it is not statistically 295
different (Extended Data Fig. 5c). Again, the DPOAE threshold reduction in Mir9614C>A/+
296
mice injected at 3-week-old was greater than that in Mir9614C>A/+ mice injected at 6-297
week-old, 27 dB vs. 18 dB. 298
In the 16-week-old mice injection group, no difference in ABR and DPOAE thresholds 299
was detected between AAV2-KKH-saCas9-sgRNA-4 treated and contralateral control 300
ears at 26 weeks age (Extended Data Fig. 5d-e). The data demonstrate that at the later 301
stage with severe hearing loss deterioration, genome editing therapy is no longer an 302
effective treatment. Combined with the data from 3-, 6- and 16-week injections, the 303
results highlight the importance of early intervention for more effective outcomes before 304
substantial hearing loss has initiated. 305
306
In vivo genome editing preserves hair cell survival. To test the effect of editing on 307
HCs in vivo, we injected the AAV2-KKH-saCas9-sgRNA-4 into the inner ear of 6-week-308
old Mir9614C>A/+ mice. Cochleae were harvested 10 weeks after the injection for hair cell 309
labeling using an anti-MYO7A antibody and confocal imaging (Fig. 6a). In the uninjected 310
Mir9614C>A/+ inner ears, outer hair cell (OHC) loss across cochlear turns was observed 311
with the most severe loss in the basal turn (Fig. 6c, e, f). A slight loss of the inner hair 312
cells (IHC) was detected in the basal turn only (Fig. 6c, e, f). In the injected Mir9614C>A/+ 313
ears, improved OHC survival was seen across all frequency regions compared to the 314
control group (Fig. 6b, d, f); whereas a slight reduction in the IHC number was seen in 315
the base similar to uninjected ears (Fig. 6g). To further examine hair cell structure, 316
scanning electron microscopy (SEM) was performed to visualize the details of OHC and 317
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
IHC stereocilia. Cochleae from uninjected and AAV2-KKH-saCas9-sgRNA-4 injected 318
Mir9614C>A/+ mice were harvested at 14 weeks after injection and imaged by SEM (Fig. 319
6a). Hair cells from the uninjected Mir9614C>A/+ mice cochleae showed signs of 320
degeneration, including missing stereocilia in the outer hair cells and disorganized 321
stereocilia in the inner hair cells (Fig. 6i, k). In contrast, the hair cells of injected 322
Mir9614C>A/+ mouse cochlea had organized and well-preserved stereocilia (Fig. 6h, j). 323
These findings demonstrate that in vivo genome editing by AAV2-KKH-saCas9-sgRNA-324
4 rescues hair cells in Mir9614C>A/+ mice by promoting their survival and maintaining the 325
structure of stereocilia. 326
Safety assessment of AAV-mediated genome editing treatment in cochleae of 327
adult mice. Development of editing therapy for hearing loss requires safety 328
assessment. To address potential safety concerns about AAV-delivered CRISPR 329
genome editing in vivo, the chronic expression of saCas9 and the risk of AAV vector 330
integration were evaluated. RT-PCR analysis revealed that KKH-saCas9 RNA levels 331
peaked at 4 weeks, decreased at 8 weeks, and became undetectable after 12 weeks in 332
the injected ear (Fig. 7a). Only trace levels of KKH-saCas9 mRNA were detected in the 333
contralateral uninjected ear at 4 weeks post-injection (Fig. 7a). By Western blotting, we 334
confirmed the presence of KKH-saCas9 protein at 4 weeks post-injection and its 335
absence after 12 weeks (Fig. 7b). These findings collectively show that the expression 336
of KKH-saCas9 expression was effectively shut off within three months after injection, 337
likely due to the silencing of the CMV promoter, which is known to undergo silencing 338
over time in vivo47-49. 339
Another concern is the risk of AAV vector integration into CRISPR-induced DNA 340
breaks50,51. To assess AAV vector integration, specific primers were designed for PCR 341
and NGS analysis to detect the integration of Mir96 and AAV ITR (Fig. 7c). In cultured 342
HEK-293T-Mir9614C>A cells treated with different AAV dosages, AAV vector integration 343
at the double-strand break locus became detectable at a dosage of 103 vg/cell (Fig. 7d 344
and Extended Data Fig. 6a, b). The indel frequency and the integration ratio were 345
correlated with the increasing AAV dosage. By varying AAV dosages, we identified an 346
optimal dosage range from 102 vg/cell to 104 vg/cell, where the indel frequency was 347
near its peak while maintaining low levels of AAV vector integration in HEK cells (Fig. 7d 348
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
and Extended Data Fig. 6a, b). These results suggest that by fine-tuning the AAV 349
dosage, the potential risk of AAV vector integration can be greatly reduced while 350
maintaining efficient genome editing. 351
We further performed in vivo injection and integration assay. We did not detect any 352
integration in the whole cochlear samples (Fig. 7e), but detected minimal integration in 353
isolated hair cells (Fig. 7f). NGS revealed that out of a total of 82,453 reads, the Mir96 354
wild-type allele had 42,991 reads, while the remaining reads were associated with 355
Mir9614C>A reads. Among the Mir9614C>A reads, indel-containing reads accounted for 356
25.7% (Fig. 7g). Additionally, there were 102 integration reads, making up 0.26% of the 357
total reads (Fig. 7g). Measurement of miR96 levels using qPCR showed a decrease in 358
miR96 levels in the injected ear (89.2± 3.0%) compared to the uninjected ear (Fig. 7h), 359
likely due to RNA degradation after genome editing. No significant change was 360
observed in qRNA analysis using miR96-specific and ITR-R primers (Fig. 7i), which 361
were used to measure the miR96-ITR mRNA, indicating the absence of miR96-ITR 362
chimera transcripts after genome editing. These results demonstrate that AAV2-363
mediated genome editing in the mature cochlea exhibits minimal or negligible vector 364
integration. To further explore the possibility of eliminating AAV vector integration, a 365
reduced dose of AAV2-KKH-saCas9-sgRNA4 (1×109 vg per cochlea) was 366
administered. After a 10-week period following the injection, no integration of ITR was 367
detected in isolated hair cells (Extended Data Fig. 7a), as evidenced by NGS data with 368
an average indel frequency of 12.05% (n=3) and ITR integration reads ratio below the 369
background error (Extended Data Fig. 7b, c). These findings demonstrate that AAV 370
vector integration rate is correlated with the administrated AAV dosage, and the dosage 371
we used achieve the therapeutic threshold for genome editing therapy while minimize 372
the AAV vector integration. 373
To assess off-target editing, CIRCLEseq was performed to identify potential off-target 374
sites52. Only two off-target sites were identified from the mouse genomic DNA (Fig. 7j). 375
Computational predictions were also used to identify potential off-target loci53,54. We 376
analyzed the top ten off-target hits in mouse genome according to the cutting frequency 377
determination (CFD) score53 after KKH-saCas9/sgRNA-4 editing (Extended Data Fig. 378
6c, d), which includes the two off-target sites identified by CIRCLEseq analysis. NGS 379
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
analysis identified one off-target site with low level of indels (1.97%), while the on-target 380
editing efficiency is up to 84.6% (Fig. 7k). The off-target editing was in the intergenic 381
region (Gm19782-Fam135b), which means unlikely to disrupt any gene function. Using 382
the isolated hair cells from injected mice, we did not detect any off-target indels (Fig. 7l). 383
Together, these results show that the delivery of AAV2-KKH-saCas9-sgRNA-4 into 384
Mir9614C>A/+ cells results in minimal off-target modification, and the hearing-related 385
phenotypes are the result from on-target editing. 386
387
A dual AAV2 system enables targeting multiple human MIR96 mutations. As the 388
Mir96 seed sequence is 100% conserved across mammalian species, the gRNA 389
designs that target mouse mutations can be directly used to target orthologous human 390
mutations. In addition to the +14C>A mutation, there are two known dominant 391
mutations in the MIR96 seed region, MIR96 13G>A and MIR96 15A>T, in humans11,17. 392
We designed gRNAs for each of the two mutations (Fig. 8a). Cas9 and multiple sgRNA 393
cassettes are too big to fit in single AAV, we then created a dual AAV set consisting of 394
AAV-U1A-spCas9-polyA with three nuclear localization signals (NLSs) and sgRNA 395
cassettes to target all three mutations (14C>A, 13G>A and 15A>T), AAV-sgmiR96-396
master (Fig. 8b, c). To validate the dual AAV set, human cell lines containing the three 397
MIR96 mutations were generated using the PiggyBac system, and each cell line was 398
transfected with AAV-U1A-spCas9-polyA/AAV-sgmiR96-master. NGS revealed robust 399
indel formation in all three MIR96 mutation lines, with a similar editing efficiency (Fig. 400
8d-g and Extended Data Fig. 8a-f). Less than 1% of the reads had indels detected at 401
the MIR96 locus (Fig. 8e). The data support specific disruption of each mutant allele by 402
its corresponding gRNA. We also analyzed top 10 potential off-target sites in human 403
genome and NGS showed no indels after Cas9/sgRNA transfection (Fig. 8h, j and 404
Extended Data Fig. 8g). These findings suggest that the combination of AAV-U1A-405
spCas9-polyA/AAV-sgmiR96-master can effectively target each of the three known 406
human MIR96 mutations and holds promise for treating dominant hearing loss caused 407
by MIR96-related mutations. The design of the AAV-sgmiR96-master system expands 408
the targeting scope and simplifies the efficacy and safety evaluation of the AAV delivery 409
system. 410
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
411
DISCUSSION 412
413
With the goal of developing a clinical treatment for human genetic hearing loss of 414
DFNA50, we conducted genome editing in adult Mir9614C>A/+ cochleae and successfully 415
improved auditory function in the long term. miRNAs are involved in multiple 416
physiological and pathological inner ear processes55, and have been linked to different 417
types of hearing loss, including deafness related to hair cell development 8,56, age-418
related hearing loss57, noise-induced hearing loss58, and inner ear inflammation59. In 419
humans, heterozygous MIR96 mutations result in late onset non-syndromic progressive 420
hearing loss, indicating the potential loss of sensory hair cell identity and subsequent 421
dysfunction in the mature cochlea, thereby offering an opportunity for genetic 422
intervention in adult patients. 423
Specific and efficient sgRNAs and genome editors, spCas9-sgRNA-1 and KKH-saCas9-424
sgRNA-4 were identified and optimized for targeting the Mir9614C>A mutation. AAV2 was 425
chosen as the delivery vehicle for its efficient transduction in inner and outer hair cells. 426
Genetic disruption of the Mir9614C>A allele leads to improved survival of hair cells and the 427
preservation of auditory function in Mir9614C>A/+ mice. We conducted injections of AAV2 428
into adult mice across various age groups. The therapy was effective in improving long-429
term hearing preservation in both pre-symptomatic and symptomatic stages of hearing 430
loss in Mir9614C>A/+ mice. Notably, we observed transient expression of KKH-saCas9 in 431
the inner ear, as the CMV promoter became silenced over time in vivo. This temporary 432
expression provides a controlled and regulated editing process, which enhances the 433
safety and precision of the therapy. Additionally, we demonstrated that fine-tuning the 434
dosage of AAV helps minimize the risk of AAV-mediated integration and improves the 435
safety profile of the therapy. These findings greatly enhance the safety profile of in vivo 436
genome editing therapy. The study also introduces a dual-AAV system capable of 437
targeting all known human MIR96 seed region mutations, making it a promising 438
candidate for treating various MIR96 dominant mutation-caused genetic hearing loss 439
conditions. Our study highlights the editing efficiency, safety, long-term therapeutic 440
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
efficacy of genome editing in treating hearing loss due to Mir96 mutations in adult 441
animal models and provides a promising path for clinical applications. 442
The selection of the most suitable editor and delivery vehicle is crucial in developing a 443
genome editing therapy for specific genetic disorders. In this study, we chose Cas9 444
nucleases to disrupt the mutated allele of Mir9614C>A/+ due to its gain-of-function 445
inheritance pattern18 meaning that the resulting indels would disable the expression of 446
the disease-causing mutant Mir96. While prime editing has the potential to correct the 447
Mir96 mutation, our in vitro data showed modest efficiency at the Mir96 locus, requiring 448
further optimization. AAV2 was selected as the preferred delivery vehicle for CRISPR 449
due to due to its specific targeting of inner and outer hair cells in the cochlea of adult 450
animals39. Besides, AAV2 serotype remains the most used throughout the study period 451
and has the most safety and efficacy evidence, with the most completed clinical trials60. 452
One of the major concerns associated with AAV-mediated CRISPR genome editing in 453
vivo is the potential for off-target editing caused by prolonged expression of 454
Cas9/sgRNA61. High-fidelity versions of CRISPR systems35,62,63 can be applied to 455
improve specificity. For example, we have demonstrated LZ3 Cas9 can target the 456
Mir9614C>A allele robustly and specifically. In addition, minimizing genome editing in 457
unintended tissues and cell types is also an important way to enhance the safety of 458
CRISPR-based therapeutics. AAV2 can specifically deliver Cas9/sgRNA into hair cells 459
through local injection, reducing the likelihood of editing unwanted cells or tissues. 460
Besides, the use of the CMV promoter for KKH-saCas9 expression helps minimize off-461
target editing as it tends to become silenced over time in vivo47-49,64-66. We 462
demonstrated that CMV-driven KKH-Cas9 was only transiently expressed and silenced 463
in 3 months in the cochlea. Another concern is the risk of vector integration at DNA 464
double-stranded breaks (DSBs)50,51. The integration ratio of AAV varies across different 465
genomic loci, tissues, and cell types50. By focusing on hair cell-limited genome editing, 466
we were able to reduce the occurrence of vector integration, as demonstrated in our 467
results. Furthermore, we found that a higher dose of AAV increased the chance of 468
vector integration, underscoring the importance of carefully fine-tuning the dosage. To 469
advance clinical studies, it will be necessary to dissect the AAV injection dosage to 470
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
identify the threshold for achieving robust genome editing while minimizing AAV vector 471
integration. 472
MicroRNA-96 serves as a key regulator and plays a crucial role in hair cell development 473
and hearing maintenance. Our study demonstrated in vivo genome editing in adult 474
Mir9614C>A/+ mice, in both pre-symptomatic and symptomatic stages of hearing loss, can 475
improve auditory function and prevent long-term deterioration. Moreover, our study 476
highlights the importance of genetic intervention in adult mice of different age groups to 477
determine the optimal treatment window, which has implications for future clinical 478
studies. Moving forward, it will be necessary to conduct further investigations in large 479
animals such as non-human primates to evaluate the efficacy of targeted genome 480
editing, determine the ideal AAV administration dosage, and identify the most effective 481
intervention time window for maximum therapeutic benefit. Our work holds significant 482
potential in advancing the clinical application of genome editing therapy for treating 483
genetic inherited hearing loss. 484
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Methods 485
486
Animals and surgery. The Mir9614C>A allele (official symbol Mir96tm3.1Wtsi, referred to 487
here as Mir9614C>A) was generated at the Wellcome Sanger Institute as part of the 488
Mouse Genetics Project, by targeting of the BEPD0003_D07 ES cell of C57BL/6N origin 489
and was maintained on the same genetic background. The selection cassette was 490
removed by FLP recombinase, leaving a single downstream FRT site. All procedures for 491
generating the mutant mouse were carried out in accordance with UK Home Office 492
regulations and the UK Animals (Scientific Procedures) Act of 1986 (ASPA) under UK 493
Home Office licences, and the study was approved by the Wellcome Sanger Institute 494
Ethical Review Committee. After shipping, all animals were bred and housed in Mass 495
Eye and Ear Infirmary. All studies involving animals were approved by the HMS 496
Standing Committee on Animals and the Mass Eye and Ear Infirmary Animal Care and 497
Use Committee. All mice were housed in a room maintained on a 12h light and dark 498
cycle with ad libitum access to standard rodent diet. 499
Mir9614C>A/+ and wild type (WT) mice of either sex were anesthetized using 500
intraperitoneal (i.p.) injection of ketamine (100mg/kg) and xylazine (10mg/kg). The post-501
auricular incision was exposed by shaving and disinfected using 10% povidone iodine. 502
The AAV2-CMV-KKH-saCas9-sgRNA4 of two titers was injected into the inner ears of 503
Mir9614C>A/+ mice. The AAV2-GFP was injected into the inner ears of WT mice. The total 504
volume for each injection was 1~1.2 μl virus per cochlea. 505
Plasmid construction. pMax-spCas9 is obtained from previous article67, U6-sgRNA 506
sequence was also obtained from PX458 (Addgene 48138)68, other Cas9 nuclease, 507
including scCas9++ (Addgene 155011)32, KKH-saCas9 (Addgene 70707)33, sauriCas9 508
(Addgene 135964)34, LZ3 Cas9 (Addgene 140561)35 cDNA are acquired from addgene. 509
Vectors for in vitro screening were constructed via Gibson assembly (NEB, E2611S). 510
KKH-saCAs9 AAV vectors are based on PX601 (Addgene 61591)69. AAV-U1a-opti-511
spCas9-PA (Supplementary Fig. 2) is based on Addgene 12150770. AAV-sgmir96-512
Master (Supplementary Fig. 3) is based on pAAV-U6-sgRNA-CMV-GFP (Addgene 513
85451)71. Plasmids encoding recombinant AAV (rAAV) genomes were cloned by Gibson 514
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
assembly. All plasmids were purified using Plasmid Plus Miniprep or Maxiprep kits 515
(Qiagen). 516
AAV production. AAV vectors were produced by Mass eye and ear infirmary vector 517
core (Boston, MA, USA). AAV plasmids containing “CMV-KKH-SaCas9-PA” cassette, 518
“U6-sgRNA-4” or “U6-sgCtrl” cassette was sequenced before packaging (MGH DNA 519
Core, complete plasmid sequencing) into AAV2/2 (Supplementary Fig. 1). Vector titer 520
was 5.76 × 1012 vg/ml for KKH-saCas9/sgRNA-4 and 4.55× 1012 vg/ml for KKH-521
saCas9/sgCtrl as determined by qPCR specific for the inverted terminal repeat of the 522
virus. 523
Isolation and culture of Primary Fibroblasts from Mice. Mir9614C>A/+, Ai14 (CAG-524
STOP-tdTomato) and wild type mice were euthanized and cleaned with 70% ethanol. 525
The dorsal skin (~1 cm diameter) of the mice was collected and rinsed with DPBS, 526
subcutaneous fat was removed by forceps. Subsequently, the samples were cut into 527
small fragments and incubated with Dispase II (Sigma-Aldrich, USA) for overnight at 528
4 °C. The dermal layers of the skin were separated using forceps and further subjected 529
to incubation with type I collagenase (1 mg/ml Gibco, USA) for 2 hours at 37 degrees. 530
The resulting cell suspension was strained using a 40-micron strainer and centrifuged at 531
950 rpm to obtain the cell pellet. The cell pellet was seeded in T-75 flask containing 532
DMEM high glucose media (Gibco, USA) containing 10 % FBS (Gibco, USA). 533
Fibroblasts were cultured for about 2–3 days to reach ~90% confluence, then passaged 534
in T75 flasks with TrypLE Express and cultured in DMEM: F12 medium (ThermoFisher) 535
with 10% fetal bovine serum (FBS) supplemented with GlutaMax (ThermoFisher). 536
Construction of Mir9614C>A cell line using PiggyBac. Mouse Mir9614C>A (~0.6 kb) 537
harboring the +14C>A mutation was amplified by PCR from Mir9614C>A/14C>A mouse 538
genomic DNA. The PCR products were cloned into the PiggyBac donor backbone (PB-539
CAG-mNeonGreen-P2A-BSD-polyA) using Gibson Assembly. The constructed donor 540
plasmid was co-transfected with PiggyBac transposon vector (PB210PA, System 541
Biosciences) into HEI-OC1 cells. Cells were cultured and selected in the medium 542
containing 10 µg/mL Blasticidin for 2 weeks. For human MIR96 +14C>A fragment, 543
mutation was introduced by PCR and cloned into the same PiggyBac donor backbone 544
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
using Gibson Assembly. Cells were transfected by PiggyBac plasmids and selected by 545
Blasticidin for 2 weeks. Successful insertion was confirmed by PCR and sequencing 546
analysis. Clones from the positive Mir9614C>A selection were expanded for subsequent 547
studies. 548
Genome editing in vitro. We performed nucleofection using LONZA 4D-Nucleofector. 549
Cells were digested by Trypsin-EDTA (0.05%) (Thermo Fisher) and future dispersed 550
into single cells. 100,000 cells were resuspended in 20μl P3 reagent of the P3 Primary 551
Cell 4D-Nucleofector® X Kit S (Lonza V4XP-3032). 1 μg total plasmid was used for a 552
single nucleofection event and nucleofected by program EH-100. Cells were not sorted. 553
5 days after the nucleofection, cells were lysed by QuickExtract™ DNA Extraction 554
Solution (Lucigen) to extract genomic DNA. Genomic PCR was carried out using 555
NEBNext® Ultra™ II Q5® Master Mix (NEB, M0544S) to amply Mir96 loci, 400~800 ng 556
of purified PCR product were used for next generation sequencing (NGS), samples 557
were sequenced and analyzed to detect CRISPR variants from NGS reads using 558
CRISPResso2 (http://crispresso.pinellolab.org/submission) as instructed72. The sgRNA 559
protospacer sequences can be found in Supplementary Table 3. 560
Hair cells isolation and NGS analysis. Cochleae were harvested with the sensory 561
epithelia dissociated using needles under the microscope (Axiovert 200M, Carl Zeiss). 562
Inner ear tissue was immersed in 1 μM FM 1-43FX (ThermoFisher, F35355) dissolved 563
in DPBS (ThermoFisher) for 15s at room temperature in the dark, then washed by 564
DPBS. The sensory epithelia were treated with 100μl 0.05% trypsin-EDTA 565
(ThermoFisher, 25300054) for 10-20 min. During incubation, the tissue was carefully 566
dispersed into small cell clusters or single cells using a 200μl Eppendorf pipette tip. 567
Cells were then transferred into 6-well plate and placed under a fluorescent microscope 568
(ZEISS) equipped with a camera. Cells with FM 1-43FX dye were collected by a 10μl 569
Eppendorf pipette tip. Above 300 cells were collected from single cochlea. Hair cells 570
were transferred into 200μl PCR tube, and centrifuged for 5 min, 200 rcf. Supernatant 571
were carefully discarded and the isolated hair cells were lysed by 5μl QuickExtract™ 572
DNA Extraction Solution (Lucigen) and incubated in 65°C for 6 min then 98°C 3 min. All 573
5μl of the cell lysis were used for each Genomic PCR amplification using NEBNext® 574
Ultra™ II Q5® Master Mix (NEB, M0544S). PCR program is 1 cycle: 98°C 5min; 42 575
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
cycles: 98°C 15s, 60°C 20s, 72°C 10s; 1 cycle: 72°C 4min; 4°C. PCR products 576
visualization, purification and NGS analysis are the same with that described above. 577
Hearing Function Testing. Mir9614C>A/+ mice of either sex were anesthetized using 578
intraperitoneal (i.p.) injection of ketamine (100mg/kg) and xylazine (10mg/kg). For ABR 579
measurements, subcutaneous needle electrodes were inserted at the vertex, ventral 580
edge of the pinna (active electrode), and a ground reference near the tail. The mice 581
were placed in a sound-proof chamber and exposed to 5-ms tone pips delivered at a 582
rate of 35/s. The response was amplified 10,000-fold, filtered with a band-pass of 100 583
Hz to 3 kHz, digitized, and averaged using 1,024 responses at each sound pressure 584
level (SPL). The sound level was elevated in 5 dB steps from 20dB up to 90dB SPL, 585
with stimuli ranging from 5.66-45.24 kHz frequencies (in half-octave steps). The 586
“threshold” and wave 1 amplitude were identified as described previously19,20. During 587
the same recording session, DPOAEs were measured under the same conditions as for 588
ABRs. Briefly, two primary tones (f2/f1=1.2) were set with f2 varied between 5.66 and 589
45.24 kHz in half-octave steps. Primaries were swept from 20dB SPL to 80dB SPL (for 590
f2) in 5-dB steps. Thresholds required to produce a DPOAE at 5dB SPL were computed 591
by interpolation as f2 level. 592
Immunofluorescence staining. Cochleae, both injected and non-injected, were 593
harvested following CO2 inhalation as a means of animal euthanasia. Temporal bones 594
were fixed in 4% paraformaldehyde at 4°C overnight and subsequently decalcified in 595
120 mM EDTA for a week. Then the organ of Corti was dissected for whole-mount 596
immunofluorescence. The dissected tissues were blocked with a blocking solution (PBS 597
with 8% donkey serum and 0.3-1% Triton X-100) for 1 hour at room temperature. 598
Subsequently, the specimens were subjected to overnight incubation with the primary 599
antibodies: anti-MYO7A (#25-6790, Proteus BioSciences), anti-GFP (ab13970, Abcam). 600
After three rinses with PBS, the tissues were incubated with the corresponding 601
secondary antibodies for 1 hour. Finally, all specimens were mounted with 602
VECTASHIELD antifade mounting medium containing DAPI (VECTOR 603
LABORATORIES, #H-1200). Images were taken with a Leica SP8 confocal laser 604
scanning microscope (Leica Microsystems, Germany). For hair cell counting, MYO7A-605
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
positive hair cells per 100μm length were calculated in the apex, middle, and middle-606
base turns of cochleae. We counted from three independent cochleae. 607
Scanning Electron Microscopy. Following cochlea dissection, the harvested tissues 608
were placed in 2.5% glutaraldehyde solution in 0.1 M cacodylate buffer (EMS) 609
supplemented with 2 mM CaCl2. The immersion was performed for 1.5-2 hours at room 610
temperature on a tissue rotator. Subsequently, the samples were rinsed three times with 611
distilled water. The samples underwent three 10-minute rinses with 0.1 M sodium 612
cacodylate buffer. Next, they were treated with 1% osmium tetroxide for a duration of 613
one hour, followed by another three 10-minute rinses with distilled water. The samples 614
were then subjected to treatment with saturated thiocarbohydrazide in distilled water for 615
30 minutes. This cycle of treatments (one-two-one-two-one) was repeated. 616
For dehydration, the samples were transferred to 20 ml scintillation vials containing 2 ml 617
of distilled water. The vials were supplemented with 50 µL of 100% ethanol, with the 618
volume doubled every 10 minutes until the vial was full. At that point, the samples were 619
transferred to 100% ethanol. Subsequently, the samples were dried using liquid CO2 in 620
a Tousimis Autosamdri 815 to reach the critical point. Finally, the samples were 621
mounted onto aluminum specimen stubs using carbon tape, spatter-coated with a 4.5 622
nm layer of platinum using a Leica EM ACE600 and analyzed using a Hitachi S-4700 623
scanning electron microscope (SEM). 624
RNA isolation and qRT-PCR. Total RNA was extracted from inner ear tissue using the 625
ReliaPrep RNA Tissue Miniprep System (Promega, z6111). Then first-strand cDNA was 626
produced using ProtoScript® II First Strand cDNA Synthesis Kit (NEB, E6560s) with 627
random primers, following the manufacturer’s instructions. STEM-LOOP qRT-PCR were 628
used to measure Mir96 level, first-strand cDNA was produced using Mir96 specific 629
primers (rtmri96: 630
CTCAACTGGTGTCGTGGAGTCGGCAATTCAGTTGAGCAAAAATGTG). Real time 631
quantitative PCR was performed using Power SYBR Green PCR Master Mix (Applied 632
Biosystems, 4368708) on the ABI QuantStudio 3 Flex Real-Time PCR System (Applied 633
Biosystems). qmiR96-F: TCGGCAGGTTTGGAACTAGCAC; qmiR96-R: 634
CTCAACTGGTGTCGTGGA. 635
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Western blotting. After dissection of cochleae, inner ear tissue samples were lysed 636
using by RIPA Lysis and Extraction buffer (ThermoFisher, 89900) containing protease 637
inhibitor (ThermoFisher) for 30 min on ice. Protein was quantified and 80 μg of each 638
lysate were loaded per lane of a NuPAGE™ 4-12% Bis-Tris Protein Gel (Thermo Fisher 639
Scientific, NP0335PK2). Samples were separated on 200V for 35 min in the mini gel 640
tank (ThermoFisher). Protein were then transferred to Nitrocellulose Blotting 641
Membranes (PALL, P66485) at 200 mA for 2 hours. The membranes were blocked in 642
5% evaporated milk in Tris-based saline with Tween 20 (0.05% TBST) for 1 hour and 643
followed by incubating with primary antibodies overnight at 4 ºC (Anti-HA: Cell Signaling 644
Technology, 3724S; anti-GAPDH: Thermo, MA5-15738). After three rinses with TBST, 645
NC membranes were incubated with HRP conjugated secondary antibodies (Thermo 646
Fisher Scientific) for 2 hours at RT. Protein bands were visualized using SuperSignal 647
West Pico PLUS Chemiluminescent Substrate (ThermoFisher, 34577) and blot image 648
were captured by ChemiDoc Imaging system (BioRad). Uncropped images can be 649
found in Supplementary Fig. 4. 650
AAV vector integration assay. HEK 293T-Mir9614C>A cells were treated with AAV2-651
CMV-KKH-saCas9-sgRNA4 of different dosages from 1 to 107 genomic copies per cell. 652
Control cells were transduced with AAV2-CMV-KKH-saCas9-sgCtrl, 105 genomic copies 653
per cell. Cells were collected 7 days later and genomic DNA was isolated. Primers P1-654
F/ITR-R were used for detecting of AAV vector integration. For in vivo integration 655
detection, Primers P1-F/P2-R and primers P1-F/ITR-R were used to amply genomic 656
fragment from isolated hair cells, then PCR products were merged together for NGS 657
analysis. Primers premir96-F/ITR-R were used in the qRT-PCR study to detect mir96-658
ITR transcription. All primers used for AAV vector integration assay were listed in 659
Supplementary Table. 1. 660
Off-target analysis. To identify off-target sites, the CIRCLE-seq was performed 661
essentially as previously described with the minor modifications. Briefly, 50 μg genomic 662
DNA were purified from HEI-OC1 cells. Genomic DNA was sheared (NEB Ultra II FS kit, 663
E7805) and circularized. In vitro cleavage reaction of circularized genomic DNA is 664
performed with KKH-saCas9/sgRNA-4. Then the sequencing libraries were prepared 665
and sequenced on an Illumina Miseq as previously described52. Off-target sites were 666
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
identified with the standard pipeline. The negative control sample were treated with the 667
Cas9 alone to assess background. 668
Besides, we listed top10 potential off-target sites according to the cutting frequency 669
determination (CFD) score53. We designed PCR primers on the two flanks of each 670
sgRNA target sequences for amplifying 250-280 bp DNA fragments. Then amplified 671
fragments were purified for NGS to identify whether there is any off-target mutation. All 672
primers used for off-target analysis were listed in Supplementary Table. 2. 673
Statistical analysis. The number of biological and technical replicates, and parameters 674
are indicated in corresponding Figure legends. Data acquisition were performed by 675
investigators blinded to experimental groups. Statistical analysis were performed using 676
GraphPad Prism 8. The student’s t test was used to calculate statistical significance 677
between two groups. The p values < 0.05 were considered as significantly different. **** 678
p < 0.0001, *** p < 0.001, ** p < 0.01, * p < 0.05. 679
Reporting summary. Further information on research design is available in the Nature 680
Research Reporting Summary linked to this article. 681
Data availability 682
The data supporting the results in this study are available within the paper and its 683
Supplementary Information. AAV genome sequences are provided in the Supplemental 684
Data. All data are available within this study are available from the corresponding author 685
upon reasonable request. The mutant mice are available from the European Mutant 686
Mouse Archive (EMMA). 687
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
References 688
1 Angeli, S., Lin, X. & Liu, X. Z. Genetics of hearing and deafness. Anat Rec (Hoboken) 295, 689
1812-1829, doi:10.1002/ar.22579 (2012). 690
2 Géléoc, G. S. & Holt, J. R. Sound strategies for hearing restoration. Science 344, 1241062, 691
doi:10.1126/science.1241062 (2014). 692
3 Müller, U. & Barr-Gillespie, P. G. New treatment options for hearing loss. Nat Rev Drug 693
Discov 14, 346-365, doi:10.1038/nrd4533 (2015). 694
4 Omichi, R., Shibata, S. B., Morton, C. C. & Smith, R. J. H. Gene therapy for hearing loss. 695
Hum Mol Genet 28, R65-r79, doi:10.1093/hmg/ddz129 (2019). 696
5 Mahmoudian-Sani, M. R. et al. MicroRNAs: effective elements in ear-related diseases 697
and hearing loss. Eur Arch Otorhinolaryngol 274, 2373-2380, doi:10.1007/s00405-017-698
4470-6 (2017). 699
6 Mendell, J. T. & Olson, E. N. MicroRNAs in stress signaling and human disease. Cell 148, 700
1172-1187, doi:10.1016/j.cell.2012.02.005 (2012). 701
7 Xiang, L. et al. miR-183/96 plays a pivotal regulatory role in mouse photoreceptor 702
maturation and maintenance. Proc Natl Acad Sci U S A 114, 6376-6381, 703
doi:10.1073/pnas.1618757114 (2017). 704
8 Kuhn, S. et al. miR-96 regulates the progression of differentiation in mammalian 705
cochlear inner and outer hair cells. Proc Natl Acad Sci U S A 108, 2355-2360, 706
doi:10.1073/pnas.1016646108 (2011). 707
9 Weston, M. D. et al. A mouse model of miR-96, miR-182 and miR-183 misexpression 708
implicates miRNAs in cochlear cell fate and homeostasis. Sci Rep 8, 3569, 709
doi:10.1038/s41598-018-21811-1 (2018). 710
10 Soldà, G. et al. A novel mutation within the MIR96 gene causes non-syndromic inherited 711
hearing loss in an Italian family by altering pre-miRNA processing. Hum Mol Genet 21, 712
577-585, doi:10.1093/hmg/ddr493 (2012). 713
11 Mencía, A. et al. Mutations in the seed region of human miR-96 are responsible for 714
nonsyndromic progressive hearing loss. Nat Genet 41, 609-613, doi:10.1038/ng.355 715
(2009). 716
12 Meola, N., Gennarino, V. A. & Banfi, S. microRNAs and genetic diseases. PathoGenetics 717
2, 7, doi:10.1186/1755-8417-2-7 (2009). 718
13 Weston, M. D., Pierce, M. L., Rocha-Sanchez, S., Beisel, K. W. & Soukup, G. A. MicroRNA 719
gene expression in the mouse inner ear. Brain Res 1111, 95-104, 720
doi:10.1016/j.brainres.2006.07.006 (2006). 721
14 Schlüter, T. et al. miR-96 is required for normal development of the auditory hindbrain. 722
Hum Mol Genet 27, 860-874, doi:10.1093/hmg/ddy007 (2018). 723
15 Xu, S., Witmer, P. D., Lumayag, S., Kovacs, B. & Valle, D. MicroRNA (miRNA) 724
transcriptome of mouse retina and identification of a sensory organ-specific miRNA 725
cluster. J Biol Chem 282, 25053-25066, doi:10.1074/jbc.M700501200 (2007). 726
16 Wienholds, E. et al. MicroRNA expression in zebrafish embryonic development. Science 727
309, 310-311, doi:10.1126/science.1114519 (2005). 728
17 Lewis, M. A. et al. An ENU-induced mutation of miR-96 associated with progressive 729
hearing loss in mice. Nat Genet 41, 614-618, doi:10.1038/ng.369 (2009). 730
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
18 Lewis, M. A., Di Domenico, F., Ingham, N. J., Prosser, H. M. & Steel, K. P. Hearing 731
impairment due to Mir183/96/182 mutations suggests both loss and gain of function 732
effects. Dis Model Mech 14, doi:10.1242/dmm.047225 (2020). 733
19 Gao, X. et al. Treatment of autosomal dominant hearing loss by in vivo delivery of 734
genome editing agents. Nature 553, 217-221, doi:10.1038/nature25164 (2018). 735
20 Tao, Y. et al. Treatment of monogenic and digenic dominant genetic hearing loss by 736
CRISPR-Cas9 ribonucleoprotein delivery in vivo. Nat Commun 14, 4928, 737
doi:10.1038/s41467-023-40476-7 (2023). 738
21 György, B. et al. Allele-specific gene editing prevents deafness in a model of dominant 739
progressive hearing loss. Nat Med 25, 1123-1130, doi:10.1038/s41591-019-0500-9 740
(2019). 741
22 Wu, J. et al. Single and Dual Vector Gene Therapy with AAV9-PHP.B Rescues Hearing in 742
Tmc1 Mutant Mice. Mol Ther 29, 973-988, doi:10.1016/j.ymthe.2020.11.016 (2021). 743
23 Gu, X. et al. Prevention of acquired sensorineural hearing loss in mice by in vivo Htra2 744
gene editing. Genome Biol 22, 86, doi:10.1186/s13059-021-02311-4 (2021). 745
24 Xue, Y. et al. Gene editing in a Myo6 semi-dominant mouse model rescues auditory 746
function. Mol Ther 30, 105-118, doi:10.1016/j.ymthe.2021.06.015 (2022). 747
25 Noh, B. et al. In vivo outer hair cell gene editing ameliorates progressive hearing loss in 748
dominant-negative Kcnq4 murine model. Theranostics 12, 2465-2482, 749
doi:10.7150/thno.67781 (2022). 750
26 Cui, C. et al. Precise detection of CRISPR-Cas9 editing in hair cells in the treatment of 751
autosomal dominant hearing loss. Mol Ther Nucleic Acids 29, 400-412, 752
doi:10.1016/j.omtn.2022.07.016 (2022). 753
27 Yeh, W. H. et al. In vivo base editing restores sensory transduction and transiently 754
improves auditory function in a mouse model of recessive deafness. Sci Transl Med 12, 755
doi:10.1126/scitranslmed.aay9101 (2020). 756
28 Jiang, L., Wang, D., He, Y. & Shu, Y. Advances in gene therapy hold promise for treating 757
hereditary hearing loss. Mol Ther 31, 934-950, doi:10.1016/j.ymthe.2023.02.001 (2023). 758
29 Yoshimura, H., Shibata, S. B., Ranum, P. T., Moteki, H. & Smith, R. J. H. Targeted Allele 759
Suppression Prevents Progressive Hearing Loss in the Mature Murine Model of Human 760
TMC1 Deafness. Mol Ther 27, 681-690, doi:10.1016/j.ymthe.2018.12.014 (2019). 761
30 Morag A. Lewis, M. L., Miguel Angel Moreno-Pelayo and Karen P. Steel. in Association 762
for Research in Otolaryngology midwinter meeting abstracts (2020). 763
31 Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive 764
bacterial immunity. Science 337, 816-821, doi:10.1126/science.1225829 (2012). 765
32 Chatterjee, P. et al. An engineered ScCas9 with broad PAM range and high specificity 766
and activity. Nat Biotechnol 38, 1154-1158, doi:10.1038/s41587-020-0517-0 (2020). 767
33 Kleinstiver, B. P. et al. Broadening the targeting range of Staphylococcus aureus CRISPR-768
Cas9 by modifying PAM recognition. Nat. Biotechnol. 33, 1293-1298, 769
doi:10.1038/nbt.3404 (2015). 770
34 Hu, Z. et al. A compact Cas9 ortholog from Staphylococcus Auricularis (SauriCas9) 771
expands the DNA targeting scope. PLoS Biol 18, e3000686, 772
doi:10.1371/journal.pbio.3000686 (2020). 773
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
35 Schmid-Burgk, J. L. et al. Highly Parallel Profiling of Cas9 Variant Specificity. Mol Cell 78, 774
794-800.e798, doi:10.1016/j.molcel.2020.02.023 (2020). 775
36 Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks 776
or donor DNA. Nature 576, 149-157, doi:10.1038/s41586-019-1711-4 (2019). 777
37 Chen, P. J. & Liu, D. R. Prime editing for precise and highly versatile genome 778
manipulation. Nat Rev Genet 24, 161-177, doi:10.1038/s41576-022-00541-1 (2023). 779
38 Nelson, J. W. et al. Engineered pegRNAs improve prime editing efficiency. Nat 780
Biotechnol 40, 402-410, doi:10.1038/s41587-021-01039-7 (2022). 781
39 Omichi, R., Yoshimura, H., Shibata, S. B., Vandenberghe, L. H. & Smith, R. J. H. Hair Cell 782
Transduction Efficiency of Single- and Dual-AAV Serotypes in Adult Murine Cochleae. 783
Mol Ther Methods Clin Dev 17, 1167-1177, doi:10.1016/j.omtm.2020.05.007 (2020). 784
40 Du, W. et al. Rescue of auditory function by a single administration of AAV-TMPRSS3 785
gene therapy in aged mice of human recessive deafness DFNB8. Mol Ther, 786
doi:10.1016/j.ymthe.2023.05.005 (2023). 787
41 Wu, Y. et al. Highly efficient therapeutic gene editing of human hematopoietic stem 788
cells. Nat Med 25, 776-783, doi:10.1038/s41591-019-0401-y (2019). 789
42 Koblan, L. W. et al. Improving cytidine and adenine base editors by expression 790
optimization and ancestral reconstruction. Nat Biotechnol 36, 843-846, 791
doi:10.1038/nbt.4172 (2018). 792
43 Chen, B. et al. Expanding the CRISPR imaging toolset with Staphylococcus aureus Cas9 793
for simultaneous imaging of multiple genomic loci. Nucleic Acids Res 44, e75, 794
doi:10.1093/nar/gkv1533 (2016). 795
44 Staahl, B. T. et al. Efficient genome editing in the mouse brain by local delivery of 796
engineered Cas9 ribonucleoprotein complexes. Nat Biotechnol 35, 431-434, 797
doi:10.1038/nbt.3806 (2017). 798
45 Meyers, J. R. et al. Lighting up the senses: FM1-43 loading of sensory cells through 799
nonselective ion channels. J Neurosci 23, 4054-4065, doi:10.1523/jneurosci.23-10-800
04054.2003 (2003). 801
46 Shu, Y. et al. Renewed proliferation in adult mouse cochlea and regeneration of hair 802
cells. Nat Commun 10, 5530, doi:10.1038/s41467-019-13157-7 (2019). 803
47 Brooks, A. R. et al. Transcriptional silencing is associated with extensive methylation of 804
the CMV promoter following adenoviral gene delivery to muscle. J Gene Med 6, 395-805
404, doi:10.1002/jgm.516 (2004). 806
48 Levy, J. M. et al. Cytosine and adenine base editing of the brain, liver, retina, heart and 807
skeletal muscle of mice via adeno-associated viruses. Nat Biomed Eng 4, 97-110, 808
doi:10.1038/s41551-019-0501-5 (2020). 809
49 Gray, S. J. et al. Optimizing promoters for recombinant adeno-associated virus-mediated 810
gene expression in the peripheral and central nervous system using self-complementary 811
vectors. Hum Gene Ther 22, 1143-1153, doi:10.1089/hum.2010.245 (2011). 812
50 Hanlon, K. S. et al. High levels of AAV vector integration into CRISPR-induced DNA 813
breaks. Nat Commun 10, 4439, doi:10.1038/s41467-019-12449-2 (2019). 814
51 Nelson, C. E. et al. Long-term evaluation of AAV-CRISPR genome editing for Duchenne 815
muscular dystrophy. Nat Med 25, 427-432, doi:10.1038/s41591-019-0344-3 (2019). 816
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
52 Tsai, S. Q. et al. CIRCLE-seq: a highly sensitive in vitro screen for genome-wide CRISPR-817
Cas9 nuclease off-targets. Nat Methods 14, 607-614, doi:10.1038/nmeth.4278 (2017). 818
53 Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target 819
effects of CRISPR-Cas9. Nat Biotechnol 34, 184-191, doi:10.1038/nbt.3437 (2016). 820
54 Tycko, J. et al. Pairwise library screen systematically interrogates Staphylococcus aureus 821
Cas9 specificity in human cells. Nat Commun 9, 2962, doi:10.1038/s41467-018-05391-2 822
(2018). 823
55 Rudnicki, A. & Avraham, K. B. microRNAs: the art of silencing in the ear. EMBO Mol Med 824
4, 849-859, doi:10.1002/emmm.201100922 (2012). 825
56 Li, H., Kloosterman, W. & Fekete, D. M. MicroRNA-183 family members regulate 826
sensorineural fates in the inner ear. J Neurosci 30, 3254-3263, 827
doi:10.1523/jneurosci.4948-09.2010 (2010). 828
57 Xiong, H. et al. Modulation of miR-34a/SIRT1 signaling protects cochlear hair cells 829
against oxidative stress and delays age-related hearing loss through coordinated 830
regulation of mitophagy and mitochondrial biogenesis. Neurobiol Aging 79, 30-42, 831
doi:10.1016/j.neurobiolaging.2019.03.013 (2019). 832
58 Miguel, V. et al. The Role of MicroRNAs in Environmental Risk Factors, Noise-Induced 833
Hearing Loss, and Mental Stress. Antioxidants & Redox Signaling 28, 773-796, 834
doi:10.1089/ars.2017.7175 (2017). 835
59 Tan, P. X. et al. MicroRNA-207 enhances radiation-induced apoptosis by directly 836
targeting Akt3 in cochlea hair cells. Cell Death Dis 5, e1433, doi:10.1038/cddis.2014.407 837
(2014). 838
60 Kuzmin, D. A. et al. The clinical landscape for AAV gene therapies. Nat Rev Drug Discov 839
20, 173-174, doi:10.1038/d41573-021-00017-7 (2021). 840
61 Wang, D., Zhang, F. & Gao, G. CRISPR-Based Therapeutic Genome Editing: Strategies and 841
In Vivo Delivery by AAV Vectors. Cell 181, 136-150, doi:10.1016/j.cell.2020.03.023 842
(2020). 843
62 Yuen, C. T. L. et al. High-fidelity KKH variant of Staphylococcus aureus Cas9 nucleases 844
with improved base mismatch discrimination. Nucleic Acids Res 50, 1650-1660, 845
doi:10.1093/nar/gkab1291 (2022). 846
63 Akcakaya, P. et al. In vivo CRISPR editing with no detectable genome-wide off-target 847
mutations. Nature 561, 416-419, doi:10.1038/s41586-018-0500-9 (2018). 848
64 Davis, K. M., Pattanayak, V., Thompson, D. B., Zuris, J. A. & Liu, D. R. Small molecule-849
triggered Cas9 protein with improved genome-editing specificity. Nat Chem Biol 11, 316-850
318, doi:10.1038/nchembio.1793 (2015). 851
65 McCown, T. J., Xiao, X., Li, J., Breese, G. R. & Samulski, R. J. Differential and persistent 852
expression patterns of CNS gene transfer by an adeno-associated virus (AAV) vector. 853
Brain Res 713, 99-107, doi:10.1016/0006-8993(95)01488-8 (1996). 854
66 Paterna, J. C., Moccetti, T., Mura, A., Feldon, J. & Büeler, H. Influence of promoter and 855
WHV post-transcriptional regulatory element on AAV-mediated transgene expression in 856
the rat brain. Gene Ther 7, 1304-1311, doi:10.1038/sj.gt.3301221 (2000). 857
67 Zhu, W. et al. Precisely controlling endogenous protein dosage in hPSCs and derivatives 858
to model FOXG1 syndrome. Nat Commun 10, 928, doi:10.1038/s41467-019-08841-7 859
(2019). 860
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
68 Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 861
2281-2308, doi:10.1038/nprot.2013.143 (2013). 862
69 Ran, F. A. et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature 520, 863
186-191, doi:10.1038/nature14299 (2015). 864
70 Wang, D. et al. Cas9-mediated allelic exchange repairs compound heterozygous 865
recessive mutations in mice. Nat Biotechnol 36, 839-842, doi:10.1038/nbt.4219 (2018). 866
71 Duan, Y. et al. The Clustered, Regularly Interspaced, Short Palindromic Repeats-867
associated Endonuclease 9 (CRISPR/Cas9)-created MDM2 T309G Mutation Enhances 868
Vitreous-induced Expression of MDM2 and Proliferation and Survival of Cells. J Biol 869
Chem 291, 16339-16347, doi:10.1074/jbc.M116.729467 (2016). 870
72 Clement, K. et al. CRISPResso2 provides accurate and rapid genome editing sequence 871
analysis. Nat Biotechnol 37, 224-226, doi:10.1038/s41587-019-0032-3 (2019). 872
873
874
875
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Acknowledgments 876
877
Funding: This work was supported by U.S. NIH R01 DC016875, R01 DC019404, 878
UG3TR002636, U24HG010423, UH3TR002636, Curing Kids Massachusetts Eye & Ear 879
and Ines-Fredrick Yeatts Fund (to Z.-Y.C.), NIH R01 DC019404 (to X.L. and Z.-Y.C.), 880
R01DC012115, R01DC005575, and DOD RH220053 (to X.L.). This work was funded in 881
part by the Wellcome Trust (098051, 100669, 089622; KPS). For the purpose of Open 882
Access, the author has applied a CC BY public copyright license to any Author Accepted 883
Manuscript (AAM) version arising from this submission. 884
We thank the Wellcome Sanger Institute Mouse Genetics Project for generating and 885
providing the Mir96 14C>A mutant mouse. 886
We thank the Harvard Medical School Electron Microscopy Facility for their help on the 887
Scanning Electron Microscopy Imaging. 888
889
890
Author contributions: 891
Conceptualization: ZYC, WZ, WD 892
Methodology: WZ, WD 893
Experiment: WZ, WD, APR, AA, SS, YK, WW 894
Data analysis: all authors 895
Supervision: ZYC 896
Manuscript writing: WZ, WD, ZYC 897
Manuscript review and editing: WZ, WD, APR, AA, SS, YK, WW, YS, XL, MAL, KPS, 898
ZYC 899
900
Competing interests: 901
Z-Y.C is a cofounder of Salubritas Therapeutics Inc. ZY. C, W. Z. and W. D. have filed 902
patent applications based on this work, PCT/US2023/023052. X.L. is a SAB member of 903
Rescue Hearing Inc, and a SAB member of Salubritas Therapeutics. The other authors 904
declare no competing interests. 905
906
907
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
908
909
910
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
911
Main Figures 912
913
Fig. 1. Mir9614C>A mutant mice present adult onset progressive hearing loss. a, 914
Mir96 sequence in human, mouse, macaque and zebrafish and 14C>A mutation. The 915
blue text region shows 100% conservation. The mutation nucleotide in Mir9614C>A is 916
displayed in red. b, Representative Sanger sequencing results showed the Mir96 917
mutation locus in WT mice, Mir9614C>A/+, Mir9614C>A/14C>A. The red arrow indicates the 918
mutated nucleotide. c-e, ABR thresholds in Mir9614C>A/+ ears (red), compared to wild-919
type ears (blue) and Mir9614C>A/14C>A ears (black) at 4 weeks (c), 8 weeks (d), and 12 920
weeks (e), respectively. f-h, DPOAE thresholds in Mir9614C>A/+ ears (red), compared to 921
wild-type ears (blue) and Mir9614C>A/14C>A ears (black) at 4 weeks (f), 8 weeks (g), and 922
12 weeks (h), respectively. i-n, ABR thresholds in Mir9614C>A/+ ears (red), compared to 923
wild-type ears (blue) and Mir9614C>A/14C>A ears (black) at 5.6 kHz (i), 8 kHz (j), 11.3 kHz 924
(k), 16 kHz (l), 22.6 kHz (m) and 32 kHz (n), respectively. Significant hearing loss, 925
shown by the elevated ABR and DPOAE thresholds, was seen at 8 weeks, which 926
became more severe at 12 weeks. N=8. Values and error bars reflect mean ± SD. 927
Statistical analyses were performed by two-way ANOVA with Bonferroni correction for 928
multiple comparisons: P value style, <0.05 (*), <0.01 (**), <0.001(***), <0.0001(****). 929
930
Fig. 2. Mir96 14C>A allele specific genome editing using different CRISPR 931
systems in mouse and human cells. a, Sequence of the Mir96 +14 C>A mutation loci 932
and the sgRNAs designs. The mutated nucleotide in the Mir9614C>A allele is displayed in 933
red. The protospacer adjacent motifs (PAMs) nucleotides are displayed in blue. b, 934
Schematic overview of plasmid constructions for different CRISPR systems used for in 935
vitro screening. c, Experiments design of preparation and genome editing of primary 936
fibroblasts from Mir9614C>A/+ mice. d, Bar chart of the indel frequency in Mir9614C>A/+ and 937
wild-type primary fibroblast after genome editing using different Cas9/sgRNA 938
combinations. n=3. Each dot represents a unique sequencing reaction. Values and error 939
bars reflect mean ± SD. e, f, Representative NGS results from KKH-saCas9/sgRNA-4 940
edited Mir9614C>A/+ and wild-type primary fibroblasts. The red arrow indicates the double-941
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
stranded DNA cutting site. g, Schematic overview of establishment of human MIR96 942
+14 C>A HEK-293T cell line and mouse Mir9614C>A NIH-3T3 cell line using PiggyBac 943
system and genome editing procedure. h, Bar chart showing the editing efficiency of 944
Mir96 mutation locus and wild-type locus using spCas9/sgRNA-1 and KKH-945
saCas9/sgRNA-4 in human MIR96 +14 C>A HEK-293T cells and mice Mir96 +14 C>A 946
NIH-3T3 cells. n=3. Each dot represents a unique sequencing reaction. Values and 947
error bars reflect mean ± SD. i, j, Indel profiles from spCas9/sgRNA-1 and KKH-948
saCas9/sgRNA-4 edited Mir96 +14C>A HEK-293T cells. Minus numbers represent 949
deletions, plus numbers represent insertions. 950
951
Fig. 3. Optimization of CRISPR constructs for inner ear delivery. a, The design of 952
the optimized AAV structure of KKH-saCas9/sgRNA vector with multiple NLS sites. b, c, 953
Sequence of unmodified and optimized KKH-saCas9 sgRNA, with sequence changes in 954
bold. d, Schematic view and representative fluorescence images of tdTomato reporter 955
from primary fibroblasts after editing by unmodified and optimized KKH-saCas9/sgRNA 956
systems. tdTomato+ cells are edited. e, Bar chart showing the editing efficiency by the 957
quantification of tdTomato+ cells after editing. Values and error bars reflect mean ± SD. 958
Each dot represents one independent experiment. 959
960
Fig. 4. AAV2-CRISPR mediated targeted genome editing at Mir9614C>A locus in hair 961
cells of adult Mir9614C>A/+ mice. a, Schematic view of the construction and packaging of 962
AAV2-CMV-KKH-saCas9-sgRNA4 for genome editing in adult mice. b, Experimental 963
overview for in vivo studies. AAV2 was injected in the inner ear of 6-week-old mice, and 964
one group was used for sequencing 4 and 8 weeks later. c, Representative NGS results 965
of cochlea samples from AAV2-KKH-saCas9-sgRNA-4 injected and the contralateral 966
uninjected ears of Mir9614C>A/+ mice. d, Quantification of indel frequency of the NGS 967
results from AAV2-KKH-saCas9-sgRNA-4 injected, AAV2-KKH-saCas9-sgCtrl injected 968
and uninjected ears from Mir9614C>A/+ mice, as well as AAV2-KKH-saCas9-sgRNA-4 969
injected ears from wild-type mice (n=9). Cochleae were collected 4 weeks and 8 weeks 970
after AAV injection. In uninjected mice, background indel frequencies ranged between 971
0% and 0.05%. Each dot represents a unique sequencing reaction from a combination 972
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
of 3 cochleae. Values and error bars reflect mean ± SD. e, Schematic overview of the 973
experimental protocol of hair cell isolation, cell lysis and NGS. f, Representative NGS 974
result of isolated hair cells from AAV2-KKH-saCas9-sgRNA-4 injected cochlea. g, 975
Quantification of Mir9614C>A allele-specific indel frequency from NGS of hair cell and 976
cochlea samples after AAV2-KKH-saCas9-sgRNA-4 injection (n=3 for each). Each dot 977
represents a unique sequencing reaction from 3 cochleae combination. Values and 978
error bars reflect mean ± SD. h, Percentage of Mir96 wild-type allele reads, 14C>A 979
reads, and indel-containing reads in the NGS results from AAV2-KKH-saCas9-sgRNA-4 980
injected hair cells from 3 independent experiments. 981
982
Fig. 5. Sustained hearing rescue by AAV2-KKH-saCas9-sgRNA-4 in adult 983
Mir9614C>A/+ mice. a. Schematic view of workflow of AAV2-KKH-saCas9-sgRNA-4 984
constructions for auditory function assay with the editing complex delivered into adult 985
Mir9614C>A/+ mouse cochleae. b-g. After AAV2-CMV-KKH-saCas9-sgRNA-4 injection 986
into 6-week-old Mir9614C>A/+ ears, significant reductions in ABR (b, d, f) and DPOAE (c, 987
e, g) thresholds were detected in injected (blue) vs. uninjected contralateral ears (red), 988
at 16 weeks age (b-c), 20 weeks age (d-e), and 26 weeks age (f-g). n=10. h-k. 989
Representative ABR waveforms recorded from an injected (left) and an uninjected ear 990
(right) of a mouse of 20 weeks of age at the frequencies of 11.3 kHz (h-i) and 16 kHz (j-991
k). The thresholds were determined by the detection of peak 1 (green color traces). 992
Values and error bars reflect mean ± SEM. Statistical analyses were performed by two-993
way ANOVA with Bonferroni correction for multiple comparisons: P value style, <0.05 994
(*), <0.01 (**), <0.001(***), <0.0001(****). 995
996
Fig. 6. AAV2-KKH-saCas9-sgRNA-4 editing on hair cell survival and maintains 997
stereocilia intergity in Mir9614C>A/+ mice. a, Schematic diagram of experimental 998
design, with confocal analysis and SEM study of the ears harvested 10 weeks and 14 999
weeks post injection, respectively. b-e, Representative confocal z-stack images of 1000
whole mount cochleae from uninjected (c, e) and AAV2-KKH-saCas9-sgRNA-4 injected 1001
(b, d) Mir9614C>A/+ mice. Hair cells were stained for MYO7A (green). Asterisks point to 1002
missing hair cells (i). Scale bar, 20μm. f, g, Quantification and comparison of the 1003
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
number of OHCs (f) and IHCs (g) across the cochlear turns from injected and uninjected 1004
Mir9614C>A/+ mice 10 weeks post injection. Error bar represents SD. h, i, Images of 1005
scanning electron microscopy (SEM) of injected (h) uninjected (i) Mir9614C>A/+ outer hair 1006
cell bundles at the apical turn. The asterisks indicate the missing stereocilia in an outer 1007
hair cell from an uninjected ear. j, k, Images of SEM of injected (j) and uninjected (k) 1008
Mir9614C>A/+ inner hair cell bundles at the apex turn. Scale bar, 2µm. 1009
1010
Fig. 7. Safety assessment of AAV delivery of genome complex in adult mice. a, 1011
qPCR analysis of KKH-saCas9 mRNA level in the injected cochlea and the contralateral 1012
uninjected cochlea (n=6). Each dot represents an independent result from 2 cochleae 1013
combined Values and error bars reflect mean ± SD. b, Western blotting of KKH-saCas9 1014
protein in injected cochlea and the contralateral uninjected cochlea, showing KKH-1015
saCas9 protein level. c, Schematic diagram of the primers and design of AAV vector 1016
integration assay. d, Quantification of indel frequency by NGS results and AAV vector 1017
ITR integration ratio from AAV2-KKH-saCas9-sgRNA-4 edited cells. HEK-293T-miR96-1018
14C>A cells were treated with different doses of AAV for 7 days before analysis. e, Gel 1019
image of the PCR from AAV2-KKH-saCas9-sgRNA-4 edited cochlea, showing the 1020
bands of the Mir96 locus, AAV vector DNA and miR96-ITR integration. f, Gel image of 1021
the PCR products, showing the bands of Mir96 locus, AAV vectors and miR96-ITR 1022
integration of isolated hair cells from injected ears. Asterisk indicates the putative 1023
integration fragment position. g, MiR96-ITR integration reads from NGS of isolated hair 1024
cells from injected ears, the reference is the AAV ITR sequence. h, qPCR analysis of 1025
Mir96 level in injected and uninjected cochlea (n=6). Each dot represents a independent 1026
result from 2 cochleae combined. Values and error bars reflect mean ± SD. i, qPCR 1027
analysis of miR96-ITR RNA level in injected and uninjected cochlea to test if there are 1028
miR96-ITR transcripts in injected cochlea (n=3). Each dot represents an independent 1029
experiment with 2 cochleae. Values and error bars reflect mean ± SD. j, CIRCLEseq 1030
analysis of KKH-saCas9-sgRNA-4 in Mir9614C>A/+ primary fibroblasts genomic DNA. k 1031
Quantification of indel frequency of potential off-target sites from the mouse genome. l 1032
Quantification of indel frequency of potential off-target sites from AAV2-KKH-saCas9-1033
sgRNA-4 injected cochlea. Values and error bars reflect mean ± SD. 1034
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
1035
Fig. 8. A dual AAV carrying multiple gRNAs targeting all known Mir96 seed region 1036
mutations in human cells specifically and efficiently. a, Sequence information of the 1037
human MIR96 locus and the sgRNAs design to target three known seed mutations. 1038
Red: mutatio site. Blue: the protospacer adjacent motifs (PAMs) nucleotides. b, 1039
Schematic view of dual AAV constructions. One contains “U1a-spCas9-polyA” cassette, 1040
the other contains three U6-sgRNA cassettes. c, Sequence of the optimized spCas9 1041
sgRNA, the bold lettersindicate the changes compared to the unmodified sequence. d, 1042
e, Representative NGS results from spCas9/sgmiR96-master edited HEK-miR96 (15A 1043
to T) cells and spCas9/sgmiR96-master edited HEK293T wild type cells. f, g, Indel 1044
profiles from spCas9/sgmiR96-master edited HEK-miR96 (15A to T) cells. Minus 1045
numbers represent deletions, plus numbers represent insertions. h, The indel frequency 1046
in the HEK-miR96 mutation cell lines and wild-type HEK293T cells after genome editing 1047
using spCas9/sgmiR96-master. Each dot represents an independent experiment. 1048
Values and error bars reflect mean ± SD. i, Off-target analysis in spCas9/sgmiR96-1049
master edited HEK-miR96 (15A to T) cells. Genomic DNA was pooled from three 1050
independent biological replicates for sequencing. Values and error bars reflect mean ± 1051
SD. 1052
1053
1054
1055
1056
1057
1058
1059
1060
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
1061
Extended Data 1062
1063
Extended Data Fig. 1 | Targeting Mir9614C>A mutation with different CRISPR 1064
nuclease systems. a, Schematic diagram of plasmid constructions for different 1065
CRISPR systems used for in vitro screening. b, Representative reads of the NGS from 1066
spCas9/sgRNA-1 edited Mir9614C>A/+and wild-type primary fibroblasts. Magenta Dotted 1067
Lines indicate the double stranded DNA cutting site. Green indicates the mutant 1068
nucleotide. c, The indel frequency in Mir9614C>A/+ and wild-type primary fibroblasts after 1069
editing by different delivery methods and editors. Each dot represents an independent 1070
experiment. Values and error bars reflect mean ± SD. 1071
1072
Extended Data Fig. 2 | The correction of Mir9614C>A mutation using prime editing in 1073
human cells. a, Schematic overview of the prime editing system. b, Sequence of the 1074
Mir96 +14 C>A mutation locus and the prime editing design. The mutation nucleotide in 1075
the Mir9614C>A allele is displayed in red. The protospacer adjacent motifs (PAMs) 1076
nucleotides of the pegRNA are displayed in blue. c, A to C correction frequency after 1077
prime editing. Each dot represents an independent experiment. Values and error bars 1078
reflect mean ± SD. d, Representative NGS result after prime editing, showing the 1079
corrected reads. Red arrow indicates the Mir96 +14 C>A mutation nucleotide. 1080
1081
Extended Data Fig. 3 | Genome editing efficiency of optimized AAV vectors. a, 1082
Schematic diagram of AAV2 constructions for GFP delivery used for in vivo delivery into 1083
adult mouse cochleae. b, Representative confocal images of AAV2 transduction in the 1084
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
apical region of adult cochlea. Hair cells were labeled with MYO7A (red) and AAV was 1085
labeled with GFP (green). Scale bar, 20μm. c,d, Transduction efficiency of IHCs (c) and 1086
OHC (d) by AAV2-GFP across different turns of the cochlea (Apex, Apex-Middle, and 1087
Middle-Base). Time of imaging: 22 weeks of age. Values and error bars reflect mean ± 1088
SD, n=3. Each dot represents an independent experiment. e, Representative 1089
fluorescence images of HEK-GFP cells after gemome editing of unmodified and 1090
optimized KKH-saCas9/sgRNA targeting GFP. GFP negative cells are the edited cells. 1091
f, Editing efficiency shown by the percentage of GFP negative cells after genome 1092
editing. Each dot represents an independent experiment. Values and error bars reflect 1093
mean ± SD. 1094
1095
Extended Data Fig. 4 | Genome editing at Mir9614C>A locus in adult Mir9614C>A/+
1096
mice. a-c, Representative NGS results of AAV2-KKH-saCas9-sgRNA-4 injected ears 1097
from Mir9614C>A/+ mice and Mir96+/+ mice, and AAV2-KKH-saCas9-sgCtrl injected ears 1098
from Mir9614C>A/+ mice. d, Representative images of FM1-43FX labeled hair cells after 1099
digestion of cochlear tissues. FM1-43FX labeled hair cells are shown in green. 1100
Arrowheads point to the GFP+ hair cells that were picked for NGS. e, Quantification of 1101
the percentage of indel-containing reads in the NGS results of AAV2-KKH-saCas9-1102
sgRNA-4 edited cochlea samples and isolated hair cells lysis. Values and error bars 1103
reflect mean ± SD. f, g, Representative NGS results of the isolated hair cells from 1104
AAV2-KKH-saCas9-sgRNA-4 injected and uninjected ears of Mir9614C>A/+ mice. 1105
1106
Extended Data Fig. 5 | Age-dependent treatment outcome by editing in Mir9614C>A/+
1107
mice. a, The experimental design of inner ear injections of AAV2-CMV-KKH-saCas9-1108
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
sgRNA-4 in 3-week-old and 6-week-old Mir9614C>A/+ mice followed by hearing test. b-e, 1109
Effects on auditory function restoration. ABR (b, d) and DPOAE (c, e) thresholds in 1110
Mir9614C>A/+ mice treated with AAV2-CMV-KKH-saCas9-sgRNA-4 ears (blue) and 1111
untreated ears (red) at 16 weeks age (b-c) and 26 weeks age (d-e). 1112
1113
Extended Data Fig. 6 | Safety assessment of AAV delivery of editing complex in 1114
adult mice. a, Gel image of the PCR showing the miR96-ITR integration fragment after 1115
different dosages of AAV treatment in HEK-miR96-14C>A cells. b, NGS results of 1116
miR96-ITR integration reads from AAV treated HEK-miR96-14C>A cells. c, The 1117
sequences of potential off-target genetic loci of KKH-saCas9/sgRNA-4 in mouse 1118
genome. None of these loci were associated with hearing function. d, The NGS result of 1119
off-target editing at the OT1 locus showed a low level of indel formation. 1120
1121
Extended Data Fig. 7 | Low titer AAV2 eliminated AAV vector integration. a, Gel 1122
image of the PCR showing the KKH-saCas9 expression and miR96-ITR integration 1123
fragment after 1×109 vg AAV2 treatment. b, c, Representative NGS results and pie 1124
chart analysis of 1×109 vg AAV2-KKH-saCas9-sgRNA-4 injected ears from Mir9614C>A/+ 1125
mice. 1126
1127
1128
Extended Data Fig. 8 | Efficient genome editing targeting multiple MIR96 seed 1129
region mutations in human cells. a, Representative NGS results from 1130
spCas9/sgmiR96-master edited HEK-miR96 (13G to A) cells. b, c, Indel profiles from 1131
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
spCas9/sgmiR96-master edited HEK-miR96 (13G to A) cells. Minus numbers represent 1132
deletions, plus numbers represent insertions. d, Representative NGS results from 1133
spCas9/sgmiR96-master edited HEK-miR96 (14C to A) cells. e, f, Indel profiles from 1134
spCas9/sgmiR96-master edited HEK-miR96 (14C to A) cells. Minus numbers represent 1135
deletions, plus numbers represent insertions. g, The sequences of potential off-target 1136
genetic loci of spCas9/sgmiR96-master in human genome. None of these loci were 1137
associated with hearing function. 1138
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Figure 1 1139
1140
1141
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Figure 2 1142
1143
1144
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Figure 3 1145
1146
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Figure 4 1147
1148
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Figure 5 1149
1150
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Figure 6 1151
1152
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Figure 7 1153
1154
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Figure 8 1155
1156
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Extended Data Figure 1 1157
1158
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Extended Data Figure 2 1159
1160
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Extended Data Figure 3 1161
1162
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Extended Data Figure 4 1163
1164
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Extended Data Figure 5 1165
1166
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Extended Data Figure 6 1167
1168
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Extended Data Figure 7 1169
1170
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Extended Data Figure 8 1171
1172
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Supplementary Information 1173
1174
1175
1176
Targeted genome editing restores auditory function in adult mice with 1177
progressive hearing loss caused by a human microRNA mutation 1178
1179
Wenliang Zhu1,2,8, Wan Du1,2,8, Arun Prabhu Rameshbabu1,2, Ariel Miura Armstrong1,2, 1180
Stewart Silver1,2, Yehree Kim1,2, Wei Wei1,2, Yilai Shu3,4,5, Xuezhong Liu6, Morag A 1181
Lewis7, Karen P. Steel7, Zheng-Yi Chen1, 2 * 1182
1183
1 Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech 1184
and Hearing Bioscience and Technology and Program in Neuroscience, Harvard 1185
Medical School, Boston, USA. 1186
2 Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, USA. 1187
3 ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key 1188
Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, 1189
Fudan University, Shanghai, 200031, China. 1190
4 Institutes of Biomedical Science, Fudan University, Shanghai, 200032, China. 1191
5 NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, 1192
China. 1193
6 Department of Otolaryngology, University of Miami School of Medicine, Miami, FL, 1194
33136, USA. 1195
7 Wolfson Sensory, Pain and Regeneration Centre, King’s College London, London, UK. 1196
8 These authors contributed equally: Wenliang Zhu, Wan Du 1197
* Corresponding author. Email: zheng-yi_chen@meei.harvard.edu (Z.C.) 1198
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
1199
AAV-CMV-KKH-saCas9-sgRNA-4: 1200
1201
CCTGCAGGCAGCTGCGCGCTCGCTCGCTCACTGAGGCCGCCCGGGCGTCGGGCGACCTTTGGTCGCCCGGCCTCAGTGAGC1202
GAGCGAGCGCGCAGAGAGGGAGTGGCCAACTCCATCACTAGGGGTTCCTGCGGCCTCTAGACTCGAGGCGTTGACATTGATT1203
ATTGACTAGTTATTAATAGTAATCAATTACGGGGTCATTAGTTCATAGCCCATATATGGAGTTCCGCGTTACATAACTTACGGTAA1204
ATGGCCCGCCTGGCTGACCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGTAACGCCAATAGGG1205
ACTTTCCATTGACGTCAATGGGTGGAGTATTTACGGTAAACTGCCCACTTGGCAGTACATCAAGTGTATCATATGCCAAGTACG1206
CCCCCTATTGACGTCAATGACGGTAAATGGCCCGCCTGGCATTATGCCCAGTACATGACCTTATGGGACTTTCCTACTTGGCAG1207
TACATCTACGTATTAGTCATCGCTATTACCATGGTGATGCGGTTTTGGCAGTACATCAATGGGCGTGGATAGCGGTTTGACTCA1208
CGGGGATTTCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGACTTTCCAAAATGTCGT1209
AACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTGTACGGTGGGAGGTCTATATAAGCAGAGCTCTCTGGCTAACTAG1210
CCACCATGGGCAAACGCCCAGCAGCTACAAAGAAGGCAGGTCAAGCCAAGAAAAAGAAAggaGCCCCAAAGAAGAAGCGGAAG1211
GTCGGTtccAAGCGGAACTACATCCTGGGCCTGGACATCGGCATCACCAGCGTGGGCTACGGCATCATCGACTACGAGACACG1212
GGACGTGATCGATGCCGGCGTGCGGCTGTTCAAAGAGGCCAACGTGGAAAACAACGAGGGCAGGCGGAGCAAGAGAGGCGC1213
CAGAAGGCTGAAGCGGCGGAGGCGGCATAGAATCCAGAGAGTGAAGAAGCTGCTGTTCGACTACAACCTGCTGACCGACCAC1214
AGCGAGCTGAGCGGCATCAACCCCTACGAGGCCAGAGTGAAGGGCCTGAGCCAGAAGCTGAGCGAGGAAGAGTTCTCTGCC1215
GCCCTGCTGCACCTGGCCAAGAGAAGAGGCGTGCACAACGTGAACGAGGTGGAAGAGGACACCGGCAACGAGCTGTCCACC1216
AAAGAGCAGATCAGCCGGAACAGCAAGGCCCTGGAAGAGAAATACGTGGCCGAACTGCAGCTGGAACGGCTGAAGAAAGACG1217
GCGAAGTGCGGGGCAGCATCAACAGATTCAAGACCAGCGACTACGTGAAAGAAGCCAAACAGCTGCTGAAGGTGCAGAAGGC1218
CTACCACCAGCTGGACCAGAGCTTCATCGACACCTACATCGACCTGCTGGAAACCCGGCGGACCTACTATGAGGGACCTGGC1219
GAGGGCAGCCCCTTCGGCTGGAAGGACATCAAAGAATGGTACGAGATGCTGATGGGCCACTGCACCTACTTCCCCGAGGAAC1220
TGCGGAGCGTGAAGTACGCCTACAACGCCGACCTGTACAACGCCCTGAACGACCTGAACAATCTCGTGATCACCAGGGACGA1221
GAACGAGAAGCTGGAATATTACGAGAAGTTCCAGATCATCGAGAACGTGTTCAAGCAGAAGAAGAAGCCCACCCTGAAGCAGA1222
TCGCCAAAGAAATCCTCGTGAACGAAGAGGATATTAAGGGCTACAGAGTGACCAGCACCGGCAAGCCCGAGTTCACCAACCTG1223
AAGGTGTACCACGACATCAAGGACATTACCGCCCGGAAAGAGATTATTGAGAACGCCGAGCTGCTGGATCAGATTGCCAAGAT1224
CCTGACCATCTACCAGAGCAGCGAGGACATCCAGGAAGAACTGACCAATCTGAACTCCGAGCTGACCCAGGAAGAGATCGAG1225
CAGATCTCTAATCTGAAGGGCTATACCGGCACCCACAACCTGAGCCTGAAGGCCATCAACCTGATCCTGGACGAGCTGTGGCA1226
CACCAACGACAACCAGATCGCTATCTTCAACCGGCTGAAGCTGGTGCCCAAGAAGGTGGACCTGTCCCAGCAGAAAGAGATCC1227
CCACCACCCTGGTGGACGACTTCATCCTGAGCCCCGTCGTGAAGAGAAGCTTCATCCAGAGCATCAAAGTGATCAACGCCATC1228
ATCAAGAAGTACGGCCTGCCCAACGACATCATTATCGAGCTGGCCCGCGAGAAGAACTCCAAGGACGCCCAGAAAATGATCAA1229
CGAGATGCAGAAGCGGAACCGGCAGACCAACGAGCGGATCGAGGAAATCATCCGGACCACCGGCAAAGAGAACGCCAAGTAC1230
CTGATCGAGAAGATCAAGCTGCACGACATGCAGGAAGGCAAGTGCCTGTACAGCCTGGAAGCCATCCCTCTGGAAGATCTGCT1231
GAACAACCCCTTCAACTATGAGGTGGACCACATCATCCCCAGAAGCGTGTCCTTCGACAACAGCTTCAACAACAAGGTGCTCGT1232
GAAGCAGGAAGAAAACAGCAAGAAGGGCAACCGGACCCCATTCCAGTACCTGAGCAGCAGCGACAGCAAGATCAGCTACGAA1233
ACCTTCAAGAAGCACATCCTGAATCTGGCCAAGGGCAAGGGCAGAATCAGCAAGACCAAGAAAGAGTATCTGCTGGAAGAACG1234
GGACATCAACAGGTTCTCCGTGCAGAAAGACTTCATCAACCGGAACCTGGTGGATACCAGATACGCCACCAGAGGCCTGATGA1235
ACCTGCTGCGGAGCTACTTCAGAGTGAACAACCTGGACGTGAAAGTGAAGTCCATCAATGGCGGCTTCACCAGCTTTCTGCGG1236
CGGAAGTGGAAGTTTAAGAAAGAGCGGAACAAGGGGTACAAGCACCACGCCGAGGACGCCCTGATCATTGCCAACGCCGATT1237
TCATCTTCAAAGAGTGGAAGAAACTGGACAAGGCCAAAAAAGTGATGGAAAACCAGATGTTCGAGGAAAAGCAGGCCGAGAGC1238
ATGcctgagatcgagacagagcaggaatacaaggaaattttcatcacccctcatcagattaaacacataaaggacttcaaagactataaatactctcatagggtggacaaaaaaccca1239
atcgcaagctcattaatgacaccctgtactcaacacggaaggatgataaaggtaataccttgattgtgaataatcttaatggattgtatgacaaagataacgacaagctcaagaagctgatc1240
aacaagtctccagagaagctccttatgtatcaccacgacccacagacttatcagaaattgaaactgatcatggagcaatacggggatgagaagaacccactctacaaatattatgagga1241
aacaggtaattacctgaccaagtactccaagaaggataacggaccagtgatcaaaaagataaagtactatggcaacaaacttaatgcgcatttggacataactgacgattaccccaattc1242
tcgaaacaaggttgtgaagctctccctgaagccttatagatttgacgtgtacctggataatggggtttataaattcgtcaccgtgaaaaatctggacgtgatcaaaaaggagaactattatgaa1243
gtaaactcaaagtgctatgaggaggcgaagaagctgaagaagatctccaatcaggccgagttcatcgcttccttctataagaacgatctcatcaagatcaatggagagctttatcgcgtcat1244
tggtgtgaacaatgacttgctgaacaggatcgaagtcaatatgatagacattacctaccgggagtatctcgaaaacatgaatgataaacggccgcctcacatcatcaagacaatcgcatct1245
aaaactcagtcaataaaaaagtactctaccgatatcctggggaatctctatgaagtgaagtcaaagaagcacccacaaatcattaaaaaaggtAAAAGGCCGGCGGCCAC1246
GAAAAAGGCCGGCCAGGCAAAAAAGAAAAAGGGATCCTACCCATACGATGTTCCAGATTACGCTTAAGAATTCGCTGATCAGCC1247
TCGACTGTGCCTTCTAGTTGCCAGCCATCTGTTGTTTGCCCCTCCCCCGTGCCTTCCTTGACCCTGGAAGGTGCCACTCCCACT1248
GTCCTTTCCTAATAAAATGAGGAAATTGCATCGCATTGTCTGAGTAGGTGTCATTCTATTCTGGGGGGTGGGGTGGGGCAGGAC1249
AGCAAGGGGGAGGATTGGGAAGAGAATAGCAGGCATGCTGGGGAGGTACCGAGGGCCTATTTCCCATGATTCCTTCATATTTG1250
CATATACGATACAAGGCTGTTAGAGAGATAATTGGAATTAATTTGACTGTAAACACAAAGATATTAGTACAAAATACGTGACGTA1251
GAAAGTAATAATTTCTTGGGTAGTTTGCAGTTTTAAAATTATGTTTTAAAATGGACTATCATATGCTTACCGTAACTTGAAAGTATT1252
TCGATTTCTTGGCTTTATATATCTTGTGGAAAGGACGAAACACCGAAGCAAAAATGTGCTAGTTCgttAtagtactctgtaatgaaaattacaga1253
atctactaTaacaaggcaaaatgccgtgtttatctcgtcaacttgttggcgagaTTTTTTTGCGGCCGCAGGAACCCCTAGTGATGGAGTTGGCCACTCCC1254
TCTCTGCGCGCTCGCTCGCTCACTGAGGCCGGGCGACCAAAGGTCGCCCGACGCCCGGGCTTTGCCCGGGCGGCCTCAGTG1255
AGCGAGCGAGCGCGCAGCTGCCTGCAGG 1256
1257
ITR, CMV Promoter, NLSs, KKH-saCas9 coding sequence, HA Tag, PloyA, U6 Promoter, sgRNA 1258
1259
Supplementary Fig. 1 | DNA sequence of AAV-CMV-KKH-saCas9-sgRNA-4. 1260
1261
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
AAV-U1A-spCas9-PA: 1262
1263
CCTGCAGGCAGCTGCGCGCTCGCTCGCTCACTGAGGCCGCCCGGGCGTCGGGCGACCTTTGGTCGCCCGGCCTCAGTGAGC1264
GAGCGAGCGCGCAGAGAGGGAGTGGCCAACTCCATCACTAGGGGTTCCTGCGGCCTCTAGAATGGAGGCGGTACTATGTAGA1265
TGAGAATTCAGGAGCAAACTGGGAAAAGCAACTGCTTCCAAATATTTGTGATTTTTACAGTGTAGTTTTGGAAAAACTCTTAGCC1266
TACCAATTCTTCTAAGTGTTTTAAAATGTGGGAGCCAGTACACATGAAGTTATAGAGTGTTTTAATGAGGCTTAAATATTTACCGT1267
AACTATGAAATGCTACGCATATCATGCTGTTCAGGCTCCGTGGCCACGCAACTCATACTACCGGtgCCACCATGGGCAAACGCC1268
CAGCAGCTACAAAGAAGGCAGGTCAAGCCAAGAAAAAGAAAggaGCCCCAAAGAAGAAGCGGAAGGTCGGTGGAtccGACAAGA1269
AGTACAGCATCGGCCTGGACATCGGCACCAACTCTGTGGGCTGGGCCGTGATCACCGACGAGTACAAGGTGCCCAGCAAGAA1270
ATTCAAGGTGCTGGGCAACACCGACCGGCACAGCATCAAGAAGAACCTGATCGGAGCCCTGCTGTTCGACAGCGGCGAAACA1271
GCCGAGGCCACCCGGCTGAAGAGAACCGCCAGAAGAAGATACACCAGACGGAAGAACCGGATCTGCTATCTGCAAGAGATCT1272
TCAGCAACGAGATGGCCAAGGTGGACGACAGCTTCTTCCACAGACTGGAAGAGTCCTTCCTGGTGGAAGAGGATAAGAAGCAC1273
GAGCGGCACCCCATCTTCGGCAACATCGTGGACGAGGTGGCCTACCACGAGAAGTACCCCACCATCTACCACCTGAGAAAGA1274
AACTGGTGGACAGCACCGACAAGGCCGACCTGCGGCTGATCTATCTGGCCCTGGCCCACATGATCAAGTTCCGGGGCCACTT1275
CCTGATCGAGGGCGACCTGAACCCCGACAACAGCGACGTGGACAAGCTGTTCATCCAGCTGGTGCAGACCTACAACCAGCTG1276
TTCGAGGAAAACCCCATCAACGCCAGCGGCGTGGACGCCAAGGCCATCCTGTCTGCCAGACTGAGCAAGAGCAGACGGCTGG1277
AAAATCTGATCGCCCAGCTGCCCGGCGAGAAGAAGAATGGCCTGTTCGGAAACCTGATTGCCCTGAGCCTGGGCCTGACCCC1278
CAACTTCAAGAGCAACTTCGACCTGGCCGAGGATGCCAAACTGCAGCTGAGCAAGGACACCTACGACGACGACCTGGACAAC1279
CTGCTGGCCCAGATCGGCGACCAGTACGCCGACCTGTTTCTGGCCGCCAAGAACCTGTCCGACGCCATCCTGCTGAGCGACA1280
TCCTGAGAGTGAACACCGAGATCACCAAGGCCCCCCTGAGCGCCTCTATGATCAAGAGATACGACGAGCACCACCAGGACCT1281
GACCCTGCTGAAAGCTCTCGTGCGGCAGCAGCTGCCTGAGAAGTACAAAGAGATTTTCTTCGACCAGAGCAAGAACGGCTACG1282
CCGGCTACATTGACGGCGGAGCCAGCCAGGAAGAGTTCTACAAGTTCATCAAGCCCATCCTGGAAAAGATGGACGGCACCGA1283
GGAACTGCTCGTGAAGCTGAACAGAGAGGACCTGCTGCGGAAGCAGCGGACCTTCGACAACGGCAGCATCCCCCACCAGATC1284
CACCTGGGAGAGCTGCACGCCATTCTGCGGCGGCAGGAAGATTTTTACCCATTCCTGAAGGACAACCGGGAAAAGATCGAGAA1285
GATCCTGACCTTCCGCATCCCCTACTACGTGGGCCCTCTGGCCAGGGGAAACAGCAGATTCGCCTGGATGACCAGAAAGAGC1286
GAGGAAACCATCACCCCCTGGAACTTCGAGGAAGTGGTGGACAAGGGCGCTTCCGCCCAGAGCTTCATCGAGCGGATGACCA1287
ACTTCGATAAGAACCTGCCCAACGAGAAGGTGCTGCCCAAGCACAGCCTGCTGTACGAGTACTTCACCGTGTATAACGAGCTG1288
ACCAAAGTGAAATACGTGACCGAGGGAATGAGAAAGCCCGCCTTCCTGAGCGGCGAGCAGAAAAAGGCCATCGTGGACCTGC1289
TGTTCAAGACCAACCGGAAAGTGACCGTGAAGCAGCTGAAAGAGGACTACTTCAAGAAAATCGAGTGCTTCGACTCCGTGGAA1290
ATCTCCGGCGTGGAAGATCGGTTCAACGCCTCCCTGGGCACATACCACGATCTGCTGAAAATTATCAAGGACAAGGACTTCCT1291
GGACAATGAGGAAAACGAGGACATTCTGGAAGATATCGTGCTGACCCTGACACTGTTTGAGGACAGAGAGATGATCGAGGAAC1292
GGCTGAAAACCTATGCCCACCTGTTCGACGACAAAGTGATGAAGCAGCTGAAGCGGCGGAGATACACCGGCTGGGGCAGGCT1293
GAGCCGGAAGCTGATCAACGGCATCCGGGACAAGCAGTCCGGCAAGACAATCCTGGATTTCCTGAAGTCCGACGGCTTCGCC1294
AACAGAAACTTCATGCAGCTGATCCACGACGACAGCCTGACCTTTAAAGAGGACATCCAGAAAGCCCAGGTGTCCGGCCAGGG1295
CGATAGCCTGCACGAGCACATTGCCAATCTGGCCGGCAGCCCCGCCATTAAGAAGGGCATCCTGCAGACAGTGAAGGTGGTG1296
GACGAGCTCGTGAAAGTGATGGGCCGGCACAAGCCCGAGAACATCGTGATCGAAATGGCCAGAGAGAACCAGACCACCCAGA1297
AGGGACAGAAGAACAGCCGCGAGAGAATGAAGCGGATCGAAGAGGGCATCAAAGAGCTGGGCAGCCAGATCCTGAAAGAACA1298
CCCCGTGGAAAACACCCAGCTGCAGAACGAGAAGCTGTACCTGTACTACCTGCAGAATGGGCGGGATATGTACGTGGACCAG1299
GAACTGGACATCAACCGGCTGTCCGACTACGATGTGGACCATATCGTGCCTCAGAGCTTTCTGAAGGACGACTCCATCGACAA1300
CAAGGTGCTGACCAGAAGCGACAAGAACCGGGGCAAGAGCGACAACGTGCCCTCCGAAGAGGTCGTGAAGAAGATGAAGAAC1301
TACTGGCGGCAGCTGCTGAACGCCAAGCTGATTACCCAGAGAAAGTTCGACAATCTGACCAAGGCCGAGAGAGGCGGCCTGA1302
GCGAACTGGATAAGGCCGGCTTCATCAAGAGACAGCTGGTGGAAACCCGGCAGATCACAAAGCACGTGGCACAGATCCTGGA1303
CTCCCGGATGAACACTAAGTACGACGAGAATGACAAGCTGATCCGGGAAGTGAAAGTGATCACCCTGAAGTCCAAGCTGGTGT1304
CCGATTTCCGGAAGGATTTCCAGTTTTACAAAGTGCGCGAGATCAACAACTACCACCACGCCCACGACGCCTACCTGAACGCC1305
GTCGTGGGAACCGCCCTGATCAAAAAGTACCCTAAGCTGGAAAGCGAGTTCGTGTACGGCGACTACAAGGTGTACGACGTGC1306
GGAAGATGATCGCCAAGAGCGAGCAGGAAATCGGCAAGGCTACCGCCAAGTACTTCTTCTACAGCAACATCATGAACTTTTTCA1307
AGACCGAGATTACCCTGGCCAACGGCGAGATCCGGAAGCGGCCTCTGATCGAGACAAACGGCGAAACCGGGGAGATCGTGTG1308
GGATAAGGGCCGGGATTTTGCCACCGTGCGGAAAGTGCTGAGCATGCCCCAAGTGAATATCGTGAAAAAGACCGAGGTGCAG1309
ACAGGCGGCTTCAGCAAAGAGTCTATCCTGCCCAAGAGGAACAGCGATAAGCTGATCGCCAGAAAGAAGGACTGGGACCCTAA1310
GAAGTACGGCGGCTTCGACAGCCCCACCGTGGCCTATTCTGTGCTGGTGGTGGCCAAAGTGGAAAAGGGCAAGTCCAAGAAA1311
CTGAAGAGTGTGAAAGAGCTGCTGGGGATCACCATCATGGAAAGAAGCAGCTTCGAGAAGAATCCCATCGACTTTCTGGAAGC1312
CAAGGGCTACAAAGAAGTGAAAAAGGACCTGATCATCAAGCTGCCTAAGTACTCCCTGTTCGAGCTGGAAAACGGCCGGAAGA1313
GAATGCTGGCCTCTGCCGGCGAACTGCAGAAGGGAAACGAACTGGCCCTGCCCTCCAAATATGTGAACTTCCTGTACCTGGCC1314
AGCCACTATGAGAAGCTGAAGGGCTCCCCCGAGGATAATGAGCAGAAACAGCTGTTTGTGGAACAGCACAAGCACTACCTGGA1315
CGAGATCATCGAGCAGATCAGCGAGTTCTCCAAGAGAGTGATCCTGGCCGACGCTAATCTGGACAAAGTGCTGTCCGCCTACA1316
ACAAGCACCGGGATAAGCCCATCAGAGAGCAGGCCGAGAATATCATCCACCTGTTTACCCTGACCAATCTGGGAGCCCCTGCC1317
GCCTTCAAGTACTTTGACACCACCATCGACCGGAAGAGGTACACCAGCACCAAAGAGGTGCTGGACGCCACCCTGATCCACCA1318
GAGCATCACCGGCCTGTACGAGACACGGATCGACCTGTCTCAGCTGGGAGGCGACAAAAGGCCGGCGGCCACGAAAAAGGC1319
CGGCCAGGCAAAAAAGAAAAAGTGAGGAtccAATAAAGGAAATTTATTTTCATTGCAATAGTGTGTTGGAATTTTTTGTGTCTCTCA1320
GCAGGAACCCCTAGTGATGGAGTTGGCCACTCCCTCTCTGCGCGCTCGCTCGCTCACTGAGGCCGGGCGACCAAAGGTCGCC1321
CGACGCCCGGGCTTTGCCCGGGCGGCCTCAGTGAGCGAGCGAGCGCGCAGCTGCCTGCAGG 1322
1323
ITR, U1A Promoter, NLSs, spCas9 coding sequence, PloyA 1324
1325
Supplementary Fig. 2 | DNA sequence of AAV-U1A-spCas9-PA. 1326
1327
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
AAV-sgmir96-Master: 1328
1329
CCTGCAGGCAGCTGCGCGCTCGCTCGCTCACTGAGGCCGCCCGGGCGTCGGGCGACCTTTGGTCGCCCGGCCTCAGTGAGC1330
GAGCGAGCGCGCAGAGAGGGAGTGGCCAACTCCATCACTAGGGGTTCCTGCGGCCGCACGCGTGAGGGCCTATTTCCCATG1331
ATTCCTTCATATTTGCATATACGATACAAGGCTGTTAGAGAGATAATTGGAATTAATTTGACTGTAAACACAAAGATATTAGTACA1332
AAATACGTGACGTAGAAAGTAATAATTTCTTGGGTAGTTTGCAGTTTTAAAATTATGTTTTAAAATGGACTATCATATGCTTACCGT1333
AACTTGAAAGTATTTCGATTTCTTGGCTTTATATATCTTGTGGAAAGGACGAAACACCGAATGTGCTAGTTCCAAAATGTTTCAGA1334
GCTATGCTGGAAACAGCATAGCAAGTTGAAATAAGGCTAGTCCGTTATCAACTTGAAAAAGTGGCACCGAGTCGGTGCTTTTTT1335
TCTAGAGGGTACCGGGGCCCGGTCGACTAGTTATTAATAGTAATCAATTACGGGGTCATTAGTTCATAGCCCATATATGGAGTT1336
CCGCGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGACCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATG1337
TTCCCATAGTAACGCCAATAGGGACTTTCCATTGACGTCAATGGGTGGAGTATTTACGGTAAACTGCCCACTTGGCAGTACATC1338
AAGTGTATCATATGCCAAGTACGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGCCTGGCATTATGCCCAGTACATGACCT1339
TATGGGACTTTCCTACTTGGCAGTACATCTACGTATTAGTCATCGCTATTACCATGGTGATGCGGTTTTGGCAGTACATCAATGG1340
GCGTGGATAGCGGTTTGACTCACGGGGATTTCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCA1341
ACGGGACTTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTGTACGGTGGGAGGTCTATATAA1342
GCAGAGCTGGTTTAGTGAACCGTCAGATCCGCTAGCGCTACCGGTCGCCACCATGGTGAGCAAGGGCGAGGAGCTGTTCACC1343
GGGGTGGTGCCCATCCTGGTCGAGCTGGACGGCGACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGGGCGAGGGCGAT1344
GCCACCTACGGCAAGCTGACCCTGAAGTTCATCTGCACCACCGGCAAGCTGCCCGTGCCCTGGCCCACCCTCGTGACCACCC1345
TGACCTACGGCGTGCAGTGCTTCAGCCGCTACCCCGACCACATGAAGCAGCACGACTTCTTCAAGTCCGCCATGCCCGAAGG1346
CTACGTCCAGGAGCGCACCATCTTCTTCAAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGGCGACACC1347
CTGGTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGACGGCAACATCCTGGGGCACAAGCTGGAGTACAACTACA1348
ACAGCCACAACGTCTATATCATGGCCGACAAGCAGAAGAACGGCATCAAGGTGAACTTCAAGATCCGCCACAACATCGAGGAC1349
GGCAGCGTGCAGCTCGCCGACCACTACCAGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTGCCCGACAACCACTAC1350
CTGAGCACCCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGCGATCACATGGTCCTGCTGGAGTTCGTGACCGCCGCCG1351
GGATCACTCTCGGCATGGACGAGCTGTACAAGTCCGGACTCAGATCTCGATAACCTGCAGCGAATTCGATATCAAGCTTATCGA1352
TACCGAGCGCTGCTCGAGAGATCTACGGGTGGCATCCCTGTGACCCCTCCCCAGTGCCTCTCCTGGCCCTGGAAGTTGCCACT1353
CCAGTGCCCACCAGCCTTGTCCTAATAAAATTAAGTTGCATCATTTTGTCTGACTAGGTGTCCTTCTATAATATTATGGGGTGGA1354
GGGGGGTGGTATGGAGCAAGGGGCAAGTTGGGAAGACAACCTGTAGGGCCTGCGGGGTCTATTGGGAACCAAGCTGGAGTG1355
CAGTGGCACAATCTTGGCTCACTGCAATCTCCGCCTCCTGGGTTCAAGCGATTCTCCTGCCTCAGCCTCCCGAGTTGTTGGGAT1356
TCCAGGCATGCATGACCAGGCTCAGCTAATTTTTGTTTTTTTGGTAGAGACGGGGTTTCACCATATTGGCCAGGCTGGTCTCCA1357
ACTCCTAATCTCAGGTGATCTACCCACCTTGGCCTCCCAAATTGCTGGGATTACAGGCGTGAACCACTGCTCCCTTCCCTGTCC1358
TTCTGATTTTGTAGGTAACCACGCAGGAAGAGGGCCTATTTCCCATGATTCCTTCATATTTGCATATACGATACAAGGCTGTTAG1359
AGAGATAATTGGAATTAATTTGACTGTAAACACAAAGATATTAGTACAAAATACGTGACGTAGAAAGTAATAATTTCTTGGGTAGT1360
TTGCAGTTTTAAAATTATGTTTTAAAATGGACTATCATATGCTTACCGTAACTTGAAAGTATTTCGATTTCTTGGCTTTATATATCTT1361
GTGGAAAGGACGAAACACCGAATGTGCTAGTGTCAAAATGTTTCAGAGCTATGCTGGAAACAGCATAGCAAGTTGAAATAAGGC1362
TAGTCCGTTATCAACTTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTTCAGGAAACAGCTATGACACGCGTGAGGGCCTATTTC1363
CCATGATTCCTTCATATTTGCATATACGATACAAGGCTGTTAGAGAGATAATTGGAATTAATTTGACTGTAAACACAAAGATATTA1364
GTACAAAATACGTGACGTAGAAAGTAATAATTTCTTGGGTAGTTTGCAGTTTTAAAATTATGTTTTAAAATGGACTATCATATGCTT1365
ACCGTAACTTGAAAGTATTTCGATTTCTTGGCTTTATATATCTTGTGGAAAGGACGAAACACCGAATGTGCTAGAGCCAAAATGT1366
TTCAGAGCTATGCTGGAAACAGCATAGCAAGTTGAAATAAGGCTAGTCCGTTATCAACTTGAAAAAGTGGCACCGAGTCGGTGC1367
TTTTTTGTGCGGACCGAGCGGCCGCAGGAACCCCTAGTGATGGAGTTGGCCACTCCCTCTCTGCGCGCTCGCTCGCTCACTGA1368
GGCCGGGCGACCAAAGGTCGCCCGACGCCCGGGCTTTGCCCGGGCGGCCTCAGTGAGCGAGCGAGCGCGCAGCTGCCTGC1369
AGG 1370
1371
ITR, CMV Promoter, EGFP, PloyA, U6 Promoter, sg14C, sg13G, sg15A 1372
1373
Supplementary Fig. 3 | DNA sequence of AAV- sgmir96-Master. 1374
1375
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
1376
1377
1378
1379
Supplementary Fig. 4 | Uncropped images of western blots. 1380
1381
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
Name
Sequence (5'-3')
rtmri96
CTCAACTGGTGTCGTGGAGTCGGCAATTCAGTTGAGCAAAAATGTG
qmiR96-F
TCGGCAGGTTTGGAACTAGCAC
qmiR96-R
CTCAACTGGTGTCGTGGA
P1
ACAGAGCAGAGACAGATCCGCGAG
P2
TGAGACACACAGACCCTGGGATG
P3
GCTTCAACAACAAGGTGCTCGTG
P4
CTGTTGATGTCCCGTTCTTCCAG
AAV ITR
GGAACCCCTAGTGATGGAGTT
mus-mir96-NGS-F
ACAGAGCAGAGACAGATCCGCGAG
mus-mir96-NGS-R
TGAGACACACAGACCCTGGGATG
hu-mir96-NGS-F
TCCGGAGCACCTTACCCACTTCTG
hu-mir96-NGS-R
GCTGGGGCCCTGACACAAGGATG
1382
Supplementary Table. 1 | Primers used in this study. 1383
1384
1385
Primer
Sequence (5'-3')
Primer
Sequence (5'-3')
hsp-OT1-F
CGGGATTTCACCATATTGGTCAGG
hsp-OT1-R
TGGTCTCGAACTCCTGACCTCAAG
hsp-OT2-F
AAGTGATGCATAAAACCTCGTGGG
hsp-OT2-R
TGAGGTTTCACCATGTTGGCCAGG
hsp-OT3-F
TGATTCCTAGGTCCTCTTTCACAG
hsp-OT3-R
GAGAGTGACAAGAAAGCTTCTATG
hsp-OT4-F
GTCCAAAGTCTCATCTGAGACAAG
hsp-OT4-R
AGCGTACCGTGGGTGTGAGACATG
hsp-OT5-F
AATAGCAGCAAACACCTAATAGGG
hsp-OT5-R
CATCAGGACTTTCAGATGACACAG
hsp-OT6-F
GCAAATAAATGATACACAAAGATG
hsp-OT6-R
TAAACATGATTGTCAATTTGGAGG
hsp-OT7-F
ACACAAAGACGACACGCCTGACAG
hsp-OT7-R
CATCTTAATCTGTTAGGCTGATGG
hsp-OT8-F
TTCAAAACAAAGGGTGTTTGGGGC
hsp-OT8-R
ATGTTCTTTTCCTGTGACCTCCTG
hsp-OT9-F
TACATTCACACACATACACACACG
hsp-OT10-F
ACACTGTAGAAACTCTGTTATTAG
hsp-OT10-F
ACACTGTAGAAACTCTGTTATTAG
hsp-OT10-R
GAGCCATAATATACTGAAGATCAG
msa-OT1-F
TGCACTGGGAAACTGAGCAAATAG
msa-OT1-R
TCTTGAATCCCAGTCACATCATTC
msa-OT2-F
GCTGGGATCTAAGTTTGATTGCTG
msa-OT2-R
AACACTAGGATGATGTTTAGAGGC
msa -OT3-F
AAGGTGGTCAGAACTTAGAGTTTG
msa -OT3-R
AAAGCTCATTTCAAACAAAGATCG
msa-OT4-F
TTTGGGCTCCTGCCATACTCTGTG
msa-OT4-R
GGAGAGTGGCCAGGAAAGCTAGTG
msa-OT5-F
AGTACCTAAAATATATACCCTAAG
msa-OT5-R
CAGTGGGTCCCCACATGCTCAGAG
msa-OT6-F
CTGACTTCATACTATGAATATGGG
msa-OT6-R
TTGCTACTGTTATGAATCGTAATG
msa-OT7-F
ATCTACAAAGCGTTGAACCTCGGG
msa-OT7-R
GATGTCTCTCTAAGATCCAGTGAG
msa-OT8-F
TCACTGTAGCCATTTCTCATGCAG
msa-OT8-R
CTGGTCTGTCTATAATTTAGTTGG
msa-OT9-F
ACTGGTACTAGAGCTATCTCTTCC
msa-OT9-R
ATGTAAGTGTATATATGTATATCTG
msa-OT10-F
CAGGCAGGCTGGCTGGAGCAGCAG
msa-OT10-R
CCTGAGCAATTATTAACTAGGCTG
1386
Supplementary Table. 2 | Primers used for offtarget analysis in this study. 1387
1388
1389
Name
Sequence (5'-3')
sgRNA-1
AAATGTGCTAGTTCCAAAAT CGG
sgRNA-2
AAAATGTGCTAGTTCCAAAA TCG
sgRNA-3
AAAATGTGCTAGTTCCAAAA TCGG
sgRNA-4
CAAGCAAAAATGTGCTAGTTC CAAAAT
sgtdT-1
CAGACATGATAAGATACATTG ATGAGT
sgtdT-2
GTATGGCTGATTATGATCCTC TAGAGT
sg13
AAATGTGCTAGTGTCAAAAT CGG
sg15
AAATGTGCTAGAGCCAAAAT CGG
1390
Supplementary Table. 3 | sgRNA protospacer sequences used in this study. Red fonts indicate the 1391
PAM sequence. 1392
1393
1394
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted October 28, 2023. ; https://doi.org/10.1101/2023.10.26.564008doi: bioRxiv preprint
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Mutations in Atp2b2, an outer hair cell gene, cause dominant hearing loss in humans. Using a mouse model Atp2b2Obl/+, with a dominant hearing loss mutation (Oblivion), we show that liposome-mediated in vivo delivery of CRISPR-Cas9 ribonucleoprotein complexes leads to specific editing of the Obl allele. Large deletions encompassing the Obl locus and indels were identified as the result of editing. In vivo genome editing promotes outer hair cell survival and restores their function, leading to hearing recovery. We further show that in a double-dominant mutant mouse model, in which the Tmc1 Beethoven mutation and the Atp2b2 Oblivion mutation cause digenic genetic hearing loss, Cas9/sgRNA delivery targeting both mutations leads to partial hearing recovery. These findings suggest that liposome-RNP delivery can be used as a strategy to recover hearing with dominant mutations in OHC genes and with digenic mutations in the auditory hair cells, potentially expanding therapeutics of gene editing to treat hearing loss.
Article
Full-text available
Patients with mutations in the TMPRSS3 gene suffer from recessive deafness DFNB8/DFNB10. For these patients, cochlear implantation is the only treatment option. Poor cochlear implantation outcomes are seen in some patients. To develop biological treatment for TMPRSS3 patients, we generated a knockin mouse model with a frequent human DFNB8 TMPRSS3 mutation. The Tmprss3A306T/A306T homozygous mice display delayed onset progressive hearing loss similar to human DFNB8 patients. Using AAV2 as a vector to carry a human TMPRSS3 gene, AAV2-hTMPRSS3 injection in the adult knockin mouse inner ear results in TMPRSS3 expression in the hair cells and the spiral ganglion neurons. A single AAV2-hTMPRSS3 injection in Tmprss3A306T/A306T mice of an average age of 18.5 months leads to sustained rescue of the auditory function to a level similar to wild-type mice. AAV2-hTMPRSS3 delivery rescues the hair cells and the spiral ganglions neurons. This study demonstrates successful gene therapy in an aged mouse model of human genetic deafness. It lays the foundation to develop AAV2-hTMPRSS3 gene therapy to treat DFNB8 patients, as a standalone therapy or in combination with cochlear implantation.
Article
Full-text available
Gene therapy would benefit from the effective editing of targeted cells with CRISPR/Cas9 tools. However, it is difficult to precisely assess the editing performance in vivo because the tissues contain many non-targeted cells, which is one of the major barriers to clinical translation. Here, in the Atoh1-GFP;Kcnq4+/G229D mice, recapitulating a novel mutation we identified in a hereditary hearing loss pedigree, the high-efficiency editing of CRISPR/Cas9 in hair cells (34.10% on average) was precisely detected by sorting out labeled cells compared with only 1.45% efficiency in the whole cochlear tissue. After injection of the developed AAV_SaCas9-KKH_sgRNA agents, the Kcnq4+/G229D mice showed significantly lower ABR and DPOAE thresholds, shorter ABR wave I latencies, higher ABR wave I amplitudes, increased number of surviving outer hair cells (OHCs) and more hyperpolarized resting membrane potentials of OHCs. These findings provide an innovative approach to accurately assess the underestimated editing efficiency of CRISPR/Cas9 in vivo and offer a promising strategy for the treatment of KCNQ4-related deafness.
Article
Full-text available
Outer hair cell (OHC) degeneration is a major cause of progressive hearing loss and presbycusis. Despite the high prevalence of these disorders, targeted therapy is currently not available. Methods: We generated a mouse model harboring Kcnq4W276S/+ to recapitulate DFNA2, a common genetic form of progressive hearing loss accompanied by OHC degeneration. After comprehensive optimization of guide RNAs, Cas9s, vehicles, and delivery routes, we applied in vivo gene editing strategy to disrupt the dominant-negative allele in Kcnq4 and prevent progressive hearing loss. Results: In vivo gene editing using a dual adeno-associated virus package targeting OHCs significantly improved auditory thresholds in auditory brainstem response and distortion-product otoacoustic emission. In addition, we developed a new live-cell imaging technique using thallium ions to investigate the membrane potential of OHCs and successfully demonstrated that mutant allele disruption resulted in more hyperpolarized OHCs, indicating elevated KCNQ4 channel activity. Conclusion: These findings can facilitate the development of targeted therapies for DFNA2 and support the use of CRISPR-based gene therapy to rectify defects in OHCs.
Article
Full-text available
Prime editing enables the installation of virtually any combination of point mutations, small insertions or small deletions in the DNA of living cells. A prime editing guide RNA (pegRNA) directs the prime editor protein to the targeted locus and also encodes the desired edit. Here we show that degradation of the 3′ region of the pegRNA that contains the reverse transcriptase template and the primer binding site can poison the activity of prime editing systems, impeding editing efficiency. We incorporated structured RNA motifs to the 3′ terminus of pegRNAs that enhance their stability and prevent degradation of the 3′ extension. The resulting engineered pegRNAs (epegRNAs) improve prime editing efficiency 3–4-fold in HeLa, U2OS and K562 cells and in primary human fibroblasts without increasing off-target editing activity. We optimized the choice of 3′ structural motif and developed pegLIT, a computational tool to identify non-interfering nucleotide linkers between pegRNAs and 3′ motifs. Finally, we showed that epegRNAs enhance the efficiency of the installation or correction of disease-relevant mutations.
Article
Full-text available
Background Aging, noise, infection, and ototoxic drugs are the major causes of human acquired sensorineural hearing loss, but treatment options are limited. CRISPR/Cas9 technology has tremendous potential to become a new therapeutic modality for acquired non-inherited sensorineural hearing loss. Here, we develop CRISPR/Cas9 strategies to prevent aminoglycoside-induced deafness, a common type of acquired non-inherited sensorineural hearing loss, via disrupting the Htra2 gene in the inner ear which is involved in apoptosis but has not been investigated in cochlear hair cell protection. Results The results indicate that adeno-associated virus (AAV)-mediated delivery of CRISPR/SpCas9 system ameliorates neomycin-induced apoptosis, promotes hair cell survival, and significantly improves hearing function in neomycin-treated mice. The protective effect of the AAV–CRISPR/Cas9 system in vivo is sustained up to 8 weeks after neomycin exposure. For more efficient delivery of the whole CRISPR/Cas9 system, we also explore the AAV–CRISPR/SaCas9 system to prevent neomycin-induced deafness. The in vivo editing efficiency of the SaCas9 system is 1.73% on average. We observed significant improvement in auditory brainstem response thresholds in the injected ears compared with the non-injected ears. At 4 weeks after neomycin exposure, the protective effect of the AAV–CRISPR/SaCas9 system is still obvious, with the improvement in auditory brainstem response threshold up to 50 dB at 8 kHz. Conclusions These findings demonstrate the safe and effective prevention of aminoglycoside-induced deafness via Htra2 gene editing and support further development of the CRISPR/Cas9 technology in the treatment of non-inherited hearing loss as well as other non-inherited diseases.
Article
Full-text available
The microRNA miR-96 is important for hearing, as point mutations in humans and mice result in dominant progressive hearing loss. Mir96 is expressed in sensory cells along with Mir182 and Mir183, but the roles of these closely-linked microRNAs are as yet unknown. Here we analyse mice carrying null alleles of Mir182, and of Mir183 and Mir96 together to investigate their roles in hearing. We found that Mir183/96 heterozygous mice had normal hearing and homozygotes were completely deaf with abnormal hair cell stereocilia bundles and reduced numbers of inner hair cell synapses at four weeks old. Mir182 knockout mice developed normal hearing then exhibited progressive hearing loss. Our transcriptional analyses revealed significant changes in a range of other genes, but surprisingly there were fewer genes with altered expression in the organ of Corti of Mir183/96 null mice compared with our previous findings in Mir96 Dmdo mutants, which have a point mutation in the miR-96 seed region. This suggests the more severe phenotype of Mir96 Dmdo mutants compared with Mir183/96 mutants, including progressive hearing loss in Mir96 Dmdo heterozygotes, is likely to be mediated by the gain of novel target genes in addition to the loss of its normal targets. We propose three mechanisms of action of mutant miRNAs; loss of targets that are normally completely repressed, loss of targets whose transcription is normally buffered by the miRNA, and gain of novel targets. Any of these mechanisms could lead to a partial loss of a robust cellular identity and consequent dysfunction.
Article
Gene therapy focuses on genetic modification to produce therapeutic effects or treat diseases by repairing or reconstructing genetic material, thus being expected to be the most promising therapeutic strategy for genetic disorders. Due to the growing attention to hearing impairment, an increasing amount of research is attempting to utilize gene therapy for hereditary hearing loss (HHL)-an important monogenic disease and the most common type of congenital deafness. Several gene therapy clinical trials for HHL have recently been approved, and additionally, CRISPR/Cas tools have been attempted for HHL treatment. Therefore, in order to further advance the development of inner ear gene therapy and promote its broad application in other forms of genetic disease, it is imperative to review the progress of gene therapy for HHL. Herein, we address three main gene therapy strategies-gene replacement, gene suppression, and gene editing, summarizing which strategy is most appropriate for particular monogenic diseases based on different pathogenic mechanisms and then focusing on their successful applications for HHL in preclinical trials. Finally, we elaborate on the challenges and outlooks of gene therapy for HHL.
Article
Programmable gene-editing tools have transformed the life sciences and have shown potential for the treatment of genetic disease. Among the CRISPR-Cas technologies that can currently make targeted DNA changes in mammalian cells, prime editors offer an unusual combination of versatility, specificity and precision. Prime editors do not require double-strand DNA breaks and can make virtually any substitution, small insertion and small deletion within the DNA of living cells. Prime editing minimally requires a programmable nickase fused to a polymerase enzyme, and an extended guide RNA that both specifies the target site and templates the desired genome edit. In this Review, we summarize prime editing strategies to generate programmed genomic changes, highlight their limitations and recent developments that circumvent some of these bottlenecks, and discuss applications and future directions.
Article
Myosin VI is an unconventional myosin that is vital for auditory and vestibular function. Pathogenic variants in the human MYO6 gene cause autosomal dominant or recessive forms of hearing loss. Effective treatments for Myo6 mutation causing hearing loss are limited. We studied whether adeno-associated virus (AAV)-PHP.eB vector-mediated in vivo delivery of Staphylococcus aureus Cas9 (SaCas9-KKH)-single-guide (sg)RNA complexes could ameliorate hearing loss in Myo6WT/C442Y mouse model which recapitulated the phenotypes of human patients. The in vivo editing efficiency of the AAV-SaCas9-KKH-Myo6-g2 system on Myo6C442Y is 4.05% on average in Myo6WT/C442Y mice, which was ∼17-fold greater than editing efficiency of Myo6WT alleles. Rescue of auditory function was observed up to 5 months post AAV-SaCas9-KKH-Myo6-g2 injection in Myo6WT/C442Y mice. Meanwhile, shorter latencies of ABR wave I, lower distortion product otoacoustic emission (DPOAE) thresholds, increased cell survival rates, more regular hair bundle morphology, recovery of inward calcium levels were also observed in the AAV-SaCas9-KKH-Myo6-g2 treated ears compared to untreated ears. These findings provide further reference for in vivo genome editing as a therapeutic treatment for various semi-dominant forms of hearing loss and other semi-dominant diseases.