Abstract
Genetic hearing loss has emerged as a significant public health concern that demands attention. Among the various treatment strategies, gene therapy based on gene editing technology is considered the most promising approach for addressing genetic hearing loss by repairing or eliminating mutated genes. The advent of the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas system has revolutionized gene therapy through its remarkable gene editing capabilities. This system has been extensively employed in mammalian gene editing and is currently being evaluated through clinical trials. Against this backdrop, this review aims to provide an overview of recent advances in utilizing the CRISPR-Cas system to treat genetic hearing loss. Additionally, we delve into the primary challenges and prospects associated with the current application of this system in addressing genetic hearing loss.
Keywords: Genetic hearing loss, gene editing, gene therapy, clustered regularly interspaced short palindromic repeats (CRISPR), Cas9, Cas13
Introduction
Hearing loss is prevalent across all human life cycle stages, with environmental and genetic factors playing crucial roles in its onset. As of 2019, the global population affected by hearing loss reached a staggering 1.57 billion people, accounting for one-fifth of the total population. Alarmingly, it is projected that by 2050, this number will soar to 2.45 billion, representing a significant increase of 56.1% from 2019 [1]. Given that approximately 1 in 500 births are affected by genetic hearing loss and genetic factors contribute directly or indirectly to about 50-60% of cases of hearing loss [2], hearing loss caused by genetic factors has emerged as an urgent public health concern that demands attention. Presently, the primary clinical treatments for hearing loss involve hearing aids and cochlear implants. However, their effectiveness is contingent upon the number of residual spiral ganglion cells in the patient and does not fully restore natural hearing [3]. Gene therapy, conversely, holds the potential to address these limitations, as mentioned above, at a fundamental level [4]. Gene therapy encompasses three prominent approaches: gene replacement, gene suppression, and gene editing. Gene replacement involves the introduction of normal genes to synthesize functional proteins, while gene suppression tackles gain-of-function mutations by dampening the expression of mutated genes. Gene editing, on the other hand, addresses diseases through base addition, deletion, and replacement [5]. Of particular interest is gene editing, which can be applied to both dominant and recessive genetic hearing loss and has the potential to yield long-lasting effects by permanently modifying cellular genes. Encouraging breakthroughs have already been achieved in gene editing for other genetically related diseases, as exemplified by ongoing clinical trials of CRISPR therapies targeting conditions like β-thalassemia and sickle cell disease [6]. These success stories pave the way for the potential application of CRISPR-based gene editing therapies to treat genetic hearing loss.
CRISPR-Cas systems
The CRISPR locus was initially discovered in Escherichia coli and serves as a crucial component of bacterial and archaeal immune systems, acting as an adaptive defense mechanism [7]. The transcription and translation products of this locus exhibit specific recognition and binding capabilities towards exogenous nucleic acids. In the presence of Cas proteins, they efficiently degrade these nucleic acids, thereby preventing viral attacks on bacteria and archaea. In vitro studies conducted in 2012 demonstrated that CRISPR-Cas9 can cleave target DNA under the guidance of CRISPR RNA (crRNA) and trans-activating CRISPR RNA (tracrRNA) [8]. Subsequently, in 2013, CRISPR-Cas9 showcased its potential for gene editing in mammalian cells [9]. Since then, the CRISPR-Cas system has undergone significant advancements, with various CRISPR/Cas variants emerging. Consequently, this system has found application in gene therapy for diverse genetic disorders [10-13]. Nucleases employed in gene editing can be classified into two groups based on their DNA recognition patterns. The first group includes zinc-finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs), which achieve DNA binding through protein-DNA interactions. The second group comprises Cas proteins, which target specific nucleic acid sequences via guide RNA that forms base pairs directly with the target nucleic acid [14]. Compared to the other three classes of nucleases, the CRISPR-Cas system offers greater simplicity and versatility. By simply modifying the base sequence of the guide RNA, it can effectively target different nucleic acid sequences. The CRISPR-Cas system is extensive and can be classified into two classes, six types and thirty-three subtypes [15]. Within this large family, this review will primarily focus on type II CRISPR-Cas9 and type VI CRISPR-Cas13, belonging to class 2. These two types treat hearing loss and represent the most prominent CRISPR-Cas systems.
CRISPR-Cas9, which belongs to type II, consists of three components: the CRISPR RNA (crRNA), the trans-activating crRNA (tracrRNA), and the Cas protein [16], which together form the ribonucleoprotein (RNP) complex to cleave DNA efficiently. The crRNA contains a sequence that can bind to the target DNA base pairing sequence that directs Cas9 RNP to a specific location to form an R-loop. The formation of the R-loop activates the DNA cleavage structural domain of Cas9, RuvC, and His-Asn-His (HNH) to cleave the DNA to form DSBs [17]. The gRNA in CRISPR-Cas9 is formed as a chimeric molecule consisting of tracrRNA and crRNA anteceded by an 18-20-nt spacer sequence complementary to the target DNA adjacent to the protospacer adjacent motif (PAM). The PAM is a 3-nt (NGG) sequence located immediately downstream of the single-guide RNA (sgRNA) target site and together with the first 20-nt of the chimeric sgRNA, which determines the target specificity of the Cas9 endonuclease [18]. The process of gene editing by CRISPR-Cas9 can be summarized as a two-step process: the first step is the expression of nuclear-localized Cas9 protein and gRNA in the target cell and the formation of an RNP complex. The second step is that the RNP complex recognizes the PAM site located near the 3’ end of the target site and, under the guidance of the sgRNA Cas9, generates DSBs approximately three bp upstream of the PAM site (Figure 1A).
Figure 1.
CRISPR-Cas-mediated gene editing. A. CRISPR-Cas9-mediated gene editing. Cas9 produces DSBs by shearing the target DNA in the presence of sgRNA. Repair of DSBs through the NHEJ pathway is likely to result in indel mutations, whereas through the HDR pathway, in the presence of donor DNA template, can perform precise repair. B. Combining CRISPR-Cas9 with CBE for single base editing of DNA. C. CRISPR-Cas13-mediated RNA editing. D. Combining CRISPR-Cas13 with ABE for single-base editing of RNA. CRISPR, clustered regularly interspaced short palindromic repeat; sgRNA, single-guide RNA; HDR, homology-directed repair; NHEJ, non-homologous end-joining; DSBs, double-strand breaks; indel, insertions or deletions; ABE, adenine base editor; CBE, cytosine base editors.
CRISPR-Cas13, which belongs to type VI, differs from type II Cas9 in that it does not require a tracrRNA and consists mainly of crRNA and Cas proteins. The DR region of the crRNA forms a single hairpin flanked on one side by a roughly 20-30-nt target RNA specific to a particular target RNA and mediates its recognition of spacer sequence. In this review, crRNA is referred to as guide RNA or gRNA. After the RNP complex forms a ternary complex with the target RNA, the conformation is rearranged so that the crRNA: target RNA hybrid is surrounded by the nuclease core, and the catalytic nuclease is activated [19,20]. Unlike CRISPR-Cas9, which cleaves explicitly double-stranded DNA sequences complementary to the crRNA spacer sequence (i.e., target DNA), the Cas13-crRNA complex nonspecifically cleaves its bound activator RNA, as well as any surrounding single-stranded RNA encountered from both phage and host (also called collateral cleavage, collateral cleavage) [21]. The Cas13 protein does not require Protospacer Adjacent Motif (PAM) sequences to recognize its targets and distinguish between autologous and non-autologous nucleic acids. Still, some preference for nucleotides flanking the 3’ regions of the protospacer has been observed and has been termed the protospacer flanking site (PFS) [22,23]. We can also summarize the process of gene editing by CRISPR-Cas13 as a two-step process: the first step is to express Cas13 proteins and gRNAs in target cells and form RNP complexes. The second step is for the RNP complexes to form a ternary complex with the target RNAs under the guidance of the sgRNAs, and to cleave the target RNAs (Figure 1C).
CRISPR-Cas delivery forms, vectors, and route
The delivery forms of the CRISPR-Cas system can be categorized into DNA, RNA, and RNP complexes. The DNA form refers to the delivery of DNA into the target cell’s nucleus, where it is transcribed into mRNA and then translated into Cas proteins, which permits sustained Cas expression [24]. Still, the prolonged-expression also increases the likelihood of off-target effects [25], and the molecular weight of this type of DNA exceeds 6400 kDa to hinder its delivery efficiency [26]. RNA form refers to the delivery of Cas mRNA and gRNA into the cell; compared to the DNA form, it does not require transcription to express Cas protein faster. The stability of mRNA/gRNA is not as good as DNA, so it is difficult to ensure the continuous expression of the Cas protein. RNP is a complex formed by Cas protein and gRNA that enters the nucleus and performs genome editing.
The delivery vectors of the CRISPR-Cas system can be categorized into biological, chemical, and physical methods according to the delivery mechanism into the cells. Biological methods use viruses or cellular components; viral vectors include adeno-associated virus (AAV), AdV, LV, VLP, etc. The main advantage of viral vectors is that they can target a specific type of cell, and their structure can primarily protect the CRISPR-Cas system from degradation by intracellular host enzymes. Still, viruses have disadvantages such as mutation, high immunogenicity [27,28], and hepatotoxicity [29]. To avoid the risk of immunogenicity of protein components targeting the delivery system, a chemical approach using artificially synthesized materials also seems feasible, with commonly used chemical components including polymers, lipids, or metal. Still, they are incompetent for in vivo use due to the high toxicity of cationic lipids and low transfection efficiency of in vivo tissues because of massive interaction with anionic cellular membranes [26]. There are also physical methods that rely on the physical energy of electricity or ultrasound to deliver the genes into cells, such as electroporation, sonoporation, and microinjection. However, physical methods are mainly limited to in vitro and surface delivery [30-32], and it is difficult to ensure the safety and effectiveness of in vivo delivery to deep organs such as the cochlea, so it may be possible to achieve better delivery results if physical methods are combined with chemical or biological methods.
The primary delivery routes for the CRISPR-Cas system include the round window membrane (RWM), canalostomy, cochleostomy, and RWM combined with canal fenestration (CF) [33]. The RWM is currently the most established and preferred method for inner ear drug delivery, and cochlear implantation is also performed through the round window. However, studies have demonstrated that virus delivered by RWM tends to occur in a base-to-apex gradient in adult mice [34]. Canalostomy is an alternative to RWM, allowing the drug to enter the cochlea and vestibule by injecting it through a window in the posterior semicircular canals. Nevertheless, it remains challenging to determine whether the drug enters the exolymph or endolymph after injection. In contrast, cochleostomy involves directly injecting the drug into the endolymph by creating a hole between the basal turn of the cochlea and the round window. This technique offers better delivery efficiency by allowing the drug to be injected into the scala media. However, it is more traumatic to the ear compared to the previous two options. As previously mentioned, transduction following injection via the RWM route tends to occur in a base-to-apex gradient in adult mice. Increasing the injection volume only leads to additional hearing loss. Therefore, combining RWM with CF, which enables the drug to flow longitudinally in the inner ear while ensuring uniform distribution, presents a promising approach if transient vestibular dysfunction associated with canal fenestration in the posterior semicircular canals [34] can be effectively avoided. Additionally, Wang et al. demonstrated good safety and delivery efficiency by introducing the drug into the scala media of the cochlea through injection in the lateral wall [35].
CRISPR-mediated hearing loss therapy
As mentioned above, the CRISPR-Cas system is a well-suited technology for application in gene therapy for genetic hearing loss. This section will discuss the CRISPR-Cas systems used for in vivo gene therapy for genetic hearing loss according to CRISPR-Cas targeting DNA and RNA, respectively.
DNA-targeted gene editing
In 2015, CRISPR-Cas9 showed the first in vitro and in vivo gene editing ability for inner ear hair cells [36], laying the foundation for gene editing therapy for genetic hearing loss. Subsequently, CRISPR-Cas9 was applied to treat hearing loss associated with genetic factors such as Cdh23, Klhl18, Tmc1, Kcnq4, and Myo6 (Table 1) [37-41].
Table 1.
Summary of CRISPR-Cas gene therapy for the treatment of genetic hearing loss
Gene | Mutation | Expression site | Forms | Vector | Route | Injection age | CRISPR | Reference |
---|---|---|---|---|---|---|---|---|
TMC1 | p.M412K, c.T1253A | HCs | Plasmid | Lipofectamine 2000 | PSCC | P0-2 | Cas9 | [39] |
HCs | Plasmid | AAV-Anc80 | Inner ear | P1-P2 | SaCas9-KKH | [67] | ||
HCs | Plasmid | AAV9-PHP.B | Utricle | P1 | SpCas9 | [68] | ||
p.Y182C, c.A545G | HCs | Plasmid | AAV2/Anc80 L65 | Inner ear | P1 | SpCas9-based AID-BE4max | [62] | |
MYO6 | p.C442Y, c.1325G > A | HCs | Plasmid | AAV-PHP.eB | Scala media | P0-P2 | SaCas9-KKH | [41] |
KCNQ4 | p.W276S, c.830G > C | OHCs | Plasmid | AAV2/Anc80 L | Utricle, PSCC, RWM, scala media | P1-P3 | SpCas9 | [40] |
p.G228D, c.683G > A | OHCs | Plasmid | AAV-PHP.eB | Scala media | P1-P2 | SaCas9-KKH | [69] | |
PCDH15 | av3j “A” insertion | HCs | Plasmid | AAV2/9 | Scala media | P0-P2 | SpCas9 | [44] |
KLHL18 | p.V55F, Chr9: 110455454 C > A | IHCs | Plasmid | AAV9, AAV-PHP.eB | Inner ear | P1 | SaCas9-KKH | [38] |
CDH23 | 753A > G | HCs | Plasmid | Microinjected | One-cell-stage mouse embryos | Cas9 | [37] | |
MYO6 | p.C442Y, c.1325G > A | HCs | Plasmid | AAV-PHP.eB | Scala media | P0-P2 | Mini dCas13X-based adenine base editor (mxABE) | [66] |
TMC1 | p.M412K, c.T1253A | HCs | Plasmid | AAV-PHP.eB | RWM | P1-P2 | RfxCas13d | [63] |
Double-stranded breaks (DSBs) of DNA generated by CRISPR-Cas9 cleavage of mutant genes rely primarily on the homology-directed repair (HDR) or non-homologous DNA end joining (NHEJ) pathways for rehabilitation [14,42] (Figure 1A). The Cdh23ahl allele is a synonymous single nucleotide polymorphism (SNP) affecting the last nucleotide of the seventh coding exon of the Cdh23 gene (c.753). The presence of an adenine (A) but not guanine (G) at this position results in an increased frequency of exon 7 skipping, predisposing inbred mice carrying the A allele to age-related hearing loss [43]. The researchers injected offset-nicking Cas9 nickase with paired RNA guides and a single-stranded oligonucleotide donor template in C57BL/6NTac zygotes to repair a point mutation in the Cdh23 gene via the HDR pathway. Encouragingly, mice that underwent successful mutation repair exhibited normal hearing thresholds and maintained hair cells and hair bundles at 36 weeks. However, the survival rate of the mice in this study was meager, with only 104 out of the 456 fertilized eggs injected surviving, and only four mice were successfully repaired with mutations [37]. Treatment of Cdh23 mutant mice achieved salvage of hearing loss in mice, but the HDR pathway hardly occurs in nondividing, terminally differentiated cells/organs [14]. It is only carried out in dividing cells, so if it is desired to achieve efficient gene repair in nondividing mammalian cells (including hair cells), efficient gene repair needs to be completed by the HMEJ-based system. The researchers devised a new HMEJ-based strategy using CRISPR/Cas9-mediated cleavage of the transgene donor vector, which contains guide RNA target sites and a homology arm, and division of the targeted site in the genome. The CRISPR/Cas9 knock-in system delivered into the inner ear using AAV successfully corrected the hair cells mutation in Klhl18lowf mice, restored the inner hair cell static cilia morphology, and significantly improved the hearing loss of the mice for up to 6 months after treatment [38].
The NHEJ pathway is the primary DSB repair mechanism but tends to introduce random insertions or deletions without the donor vector, so it is widely used in gene disruption [44-48]. Tmc1 (transmembrane channel-like 1) protein is essential to mechanotransduction channels in mammalian hair cells. Dominant-negative mutation in TMC1 (p.M418K, c.T1253A) cause progressive post-lingual sensorineural hearing loss in humans [49-51]. Beethoven mouse model carrying a homozygous mutation (p.M412K, c.T1235A) in the human Tmc1 gene exhibits progressively elevated auditory response thresholds and progressive loss of hair cells starting at one month [52]. In 2017, researchers used Lipofectamine 2000 to deliver Cas9-sgRNA RNP complexes into the inner ear, disrupting dominant deafness-associated alleles in a Bth mouse model. They succeeded in partially alleviating hearing loss in Tmc1Bth/+ mice [39]. The study provides evidence of the efficacy of in vivo disruption of target genes using the CRISPR-Cas system to treat specific autosomal dominant hearing loss disorders.
To achieve more precise targeting and improve the therapeutic efficiency of CRISPR-Cas9, researchers have also tried to use AAV as a delivery vector. Myosin VI (MYO6) is an unconventional myosin vital for auditory and vestibular function. The MYO6 p.C442Y mutation causes DFNA22. Carriers of the Myo6 p.C442Y mutation begin to develop progressive hearing loss during childhood and show profound sensorineural hearing loss by middle age [53]. The researchers used AAV-PHP.eB as a vector to deliver Cas9 (SaCas9-KKH)-sgRNA complexes into the Myo6WT/C442Y mouse model that recapitulated the phenotypes of human patients. At week 15 post-treatment, auditory brainstem response (ABR) was restored by approximately 17 dB at 16 kHz in AAV-SaCas9-KKH-Myo6-gRNA-treated ears compared to untreated ears, with lower distortion product otoacoustic emission (DPOAE) thresholds, increased outer hair cell survival, and more regular hair bundle morphology. Recovery of auditory function was observed up to 5 months after treatment [41].
The aforementioned studies provide compelling evidence to support the potential of utilizing CRISPR-Cas9 gene editing technology to treat dominant and semi-dominant inherited hearing loss. Furthermore, recent findings have shed light on the non-random nature of CRISPR-Cas9 cleavage-induced insertions/deletions (indels) introduced through the NHEJ pathway in dividing cells, which appear to be dependent on the specific guide RNA sequence [54-59]. The PCDH15 protein is one of two components that form the tip link to gate the mechanotransduction channel in hair cells [60], a key component for Hair cells to detect mechanical force. DFNB23-Pcdh15 av-3J is a code shift mutation in a single adenine (A) nucleobase insertion. This mutation results in the premature appearance of the stop codon to form a truncated PCDH15 protein lacking transmembrane and intracellular structural domains [61]. The researchers introduced non-random CRISPR-Cas9 cleavage-induced insertions/deletions (indels) via the NHEJ pathway to block the early appearance of the stop codon to achieve repair of the shifted-code mutation, and 30 of 52 injected mice developed ABRs at 4-5 weeks; however, no response was observed in mice that were not injected [44].
For hearing loss due to recessive loss-of-function mutations, in addition to precise gene repair under the guidance of a donor, single-base editing offers a therapeutic option that can efficiently and permanently correct the causative mutation without forming DSBs (Figure 1B). Using a dual AAV base editing delivery system, researchers delivered cytosine base editing (CBE) into the inner ear of Baringo (Tmc1Y182C/Y182C; Tmc2+/+) mice, which have a T-A to C-G mutation in the Tmc1 (c.A545G) gene, and succeeded in restoring sensory transduction and hair cell morphology in inner hair cells, and four weeks after injection at 5-35 kHz temporarily rescued 10-20 dB of hearing [62]. Although the degree of hearing salvage is very limited, the experimental results demonstrate that it is entirely feasible to utilize in vivo base editing to restore recessively inherited hearing loss.
Gene editing that targets DNA will cause irreversible genetic changes if it edits non-targeted normal genes, so the off-target effect of gene editing is an essential aspect of judging the safety of gene editing therapy, and it is encouraging to note that no off-target mutations were detected or reported, or only very low off-target mutations were demonstrated in these research (Table 2), suggesting that Cas9-based editing has a very good safety profile.
Table 2.
Hearing and hair cell rescue and off-target analysis
Gene | ABR threshold recovery (compared to the untreated group) | DPOAE threshold recovery (compared to the untreated group) | The hair cell | Off-target analysis | Reference |
---|---|---|---|---|---|
TMC1 | Recovered 20 dB~0 dB at 5.66 kHz to 45.24 kHz at 4 weeks | DPOAE thresholds were slightly elevated in the injected ears at 4 weeks | Preserved part of stereocilia of IHCs at 8 weeks | Modification of only one off-target site (off-T1, 1.2% indels) was detected in primary fibroblasts from Tmc1Bth/+ mice | [39] |
The mean threshold increased by 40 dB at 8 kHz at 12 weeks | Recovered 40 dB~0 dB at 5 kHz to 30 kHz at 12 weeks | Preserved normal hair bundle at 24 weeks | Did not detect off-target effects in cultures of primary fibroblasts from Tmc1Bth/WT mice | [67] | |
Recovered 40 dB~0 dB at 5 kHz to 35 kHz from no response at 4 weeks | Recovered 40 dB~0 dB at 5 kHz to 35 kHz from no response at 4 weeks | IHCs: Rescued: 3.3 in apex, 5.4 in middle, 4.7 in base; OHCs: 15.8 in apex, 10.8 in middle: 2.5 in base at 24 weeks | - | [68] | |
Recovered 20 dB~10 dB at 5 kHz to 35 kHz at 4 weeks | None of the treated mice showed recovery of DPOAE | Promoted preservation of normal hair bundle morphology at 4 weeks | No off-target editing at any protospacer position above that of an untreated control sample (≤ 0.1% mutation frequency above the untreated control) at any of the nine tested off-target sites tested | [62] | |
MYO6 | Recovered 30 dB~5 dB at 4 kHz to 32 kHz at 10 weeks | Recovered 10 dB~5 dB at 4 kHz to 32 kHz at 12 weeks | IHCs: Rescued 5.22 in middle, 4.89 in basal; OHCs: 20.34 in middle at 10 months; Observed more organized hair bundles in OHCs and IHCs | No obvious indel mutations were observed in 15 off-target sites in mESCs | [41] |
KCNQ4 | Recovered 30 dB~10 dB at 6 kHz to 30 kHz at 7 weeks | Recovered 10 dB~0 dB at 6 kHz to 30 kHz at 7 weeks | OHC viability in treated mice was not significantly different from that in non-treated mice at 7 weeks | No off-target editing at any protospacer position above that of an untreated control sample (≤ 0.1% mutation frequency above the untreated control) at any of the nine tested off-target sites tested | [40] |
Recovered 20 dB~10 dB at 5.6 kHz to 32 kHz at 8 weeks | Recovered 15 dB~5 dB at 5.6 kHz to 30 kHz at 8 weeks | The treatment promoted survival of OHCs at middle and basal turns at 12 weeks | No significant editing occurred at the four sites in treated mice compared with the control | [69] | |
PCDH15 | Recovered 20 dB~10 dB at 4 kHz to 32 kHz at 4-5 weeks from no response | - | Restored PCDH15 expression and mechanotransduction of HCS | Based on inner ears collected from treatment, 2 of 16 selected off-target sites showed detectable editing, with average indel frequencies of 0.7% and 0.3% | [44] |
KLHL18 | Recovered 20 dB~15 dB at 4 kHz to 32 kHz at 12 weeks | Normal DPOAE thresholds for this Klhl18 mutation | Rescued stereocilia morphology in some inner hair cells at 12 weeks | No obvious indel mutations for the off-target sites were detected in homozygous Klhl18lowf fibroblasts | [38] |
CDH23 | Normal ABR thresholds at 36 weeks | - | Restored normal hair cells and hair bundles | No induced sequence changes were identified at assessed the ‘off-target’ sites predicted for the guide RNAs (≤ 4 nucleotide mismatches) | [37] |
MYO6 | Recovered 25 dB~10 dB at 4 kHz to 32 kHz at 12 weeks | Recovered 30 dB~0 dB at 4 kHz to 32 kHz at 12 weeks | OHCs: Rescued 13.20 in middle turn, 29.6 in basal turn | Inducing A-to-I conversion without substantial off-target edits in vivo | [66] |
TMC1 | Recovered 20 dB~10 dB at 4 kHz to 32 kHz at 4 weeks | Recovered 20 dB~0 dB at 4 kHz to 32 kHz at 4 weeks | OHCs: 10.8 in apical turn, 17.2 in middle turn; IHCs: 10.2 in middle turn at 10 weeks; Preserved hair bundles in the middle turn | No difference in RNA expression for 9 of the top 10 most likely off-target genes, one gene was not detected in vivo compared with untreated mice | [63] |
RNA-targeted gene editing
The exploration of utilizing the CRISPR/Cas13 RNA editing system to treat genetic hearing loss is also underway. To evaluate the therapeutic efficacy of CRISPR/Cas13 researchers evaluated the therapeutic efficacy of CRISPR/Cas13 and compared the editing specificity and efficiency of the PspCas13b and CasRx systems. The CasRx and sgRNA were delivered into the inner ear of postnatal mice using AAV-PHP.eB43 as a vector and successfully reduced the transcript of Tmc1 mutants by 70.2% within two weeks and recovered ABR thresholds by 20 to 2 dB at 4 kHz~32 kHz within eight weeks after treatment, with few off-target effects across the transcriptome [63]. This research suggests that CRISPR-CasRx-based RNA editing is a promising therapeutic approach for treating autosomal dominant hearing loss.
In addition to using Cas13 to cleave target RNAs, RNA single-base editors developed based on Cas13 can realize single-base changes at the RNA level by targeted RNA editing techniques (Figure 1D). Minibase editors developed based on compact Cas13 proteins that exhibit high targeting and low off-targeting efficiency in A to I and C to U substitutions [64,65] provide for in vivo treatment of genetic disorders using the CRISPR-Cas13 system only via a single AAV vector. Myo6C442Y/+ mice have a C-G to T-A change in the mutant gene Myo6 (NM_001039546.1: c.1325G > A), so the researchers used AAV-PHP.eB to deliver the minibase editor, mxABE, into Myo6C442Y/+ mice to induce the Myo6C442Y A-to-G conversion of RNA. The treatment rescued auditory function, with ABR at 12 weeks showing 20-10 dB of hearing loss rescued at 4-32 kHz, and DPOAE also showing 20-0 dB of hearing loss rescued at 4-32 kHz [66]. The results demonstrate the potential of RNA base editing therapies to treat autosomal dominant hearing loss associated with hair cell dysfunction and provide complementary strategies to other approaches using RNA interference and DNA alteration.
In conclusion, RNA-targeted gene editing exhibits comparable effectiveness to DNA-targeted gene editing in addressing the manifestations of genetic hearing loss. Throughout the experimental duration, RNA-targeted gene editing demonstrated therapeutic efficacy on par with DNA-targeted gene editing (Table 2).
Conclusion
Gene therapy based on the CRISPR-Cas system holds great promise for treating genetic hearing loss as it enables the restoration of regular gene expression at the genetic level. Different Cas proteins in the CRISPR-Cas system are targeted to edit DNA and target RNA, so we discuss recent studies on CRISPR-Cas gene editing technology in inner ear gene therapy in the order of targeting DNA and RNA. The application of the CRISPR-Cas system in the treatment of both dominantly inherited hearing loss and recessive inherited hearing loss has shown surprising results. Still, at the same time, we also found that the delivery forms, vectors, and pathways of the CRISPR-Cas system in the inner ear limit its therapeutic effect, so we also briefly introduce the delivery forms, vectors, and pathways.
The research advancements cited in this review demonstrate the potential of CRISPR-Cas-based gene editing therapies for genetically related hearing loss (Table 2). However, several challenges need to be addressed. Currently, most gene therapies based on the CRISPR-Cas system focus on mutations in inner ear hair cells (Table 1), and treatments in supporting cells and the stria vascularis remain unexplored. For instance, GJB2, the primary causative mutant gene for genetic hearing loss, is predominantly expressed in non-sensory epithelial cells such as supportive cells. Although attempts at gene therapy for GJB2-associated genetic hearing loss have been made, they mainly involve gene replacement.
In addition, current experiments are modeled on mice. Still, there are significant species-related physiological and anatomical differences between mice and humans. Thus the CRISPR-Cas system will need to be validated for efficacy and safety in animals with genetic mutation patterns and disease phenotypes closer to those of humans, such as non-human primates before it can be applied to humans. Although non-human primates are more comparable to humans regarding genetic mutation patterns and disease phenotypes, they are not the best choice because of their long breeding cycles and high prices. The genetic hearing loss pig model being constructed by our lab and the Army Medical University has a cochlea size, thickness, and strength of the inner ear bone wall, genetic mutation patterns, and disease phenotypes that are highly similar to those of humans, and has a shorter breeding cycle than that of non-human primates. It is reasonably priced and has potential value for evaluating human gene therapy.
CRISPR-Cas system-based gene therapy had a limited rescue effect on hearing function, could not restore the hearing of mutant mice to the level of wild-type, and the rescue effect decreased significantly with the age of the mice, which indicated that the editing efficiency of CRISPR-Cas system-based gene therapy needed to be further improved. Finally, since the ultimate goal of the research is to apply the CRISPR-Cas-based system to humans, its safety is of paramount importance, and there is a need for rational ways to detect off-target effects of gene editing in the human cochlea, as well as the development of new tools to enable the timely termination of off-target gene editing.
Although the current gene therapy based on CRISPR-Cas system has some limitations, with the application of more new and expanded CRISPR-Cas systems and delivery vectors, the gene transduction efficiency and targeting can be further improved, and it is reasonable to believe that the application scenarios of CRISPR-Cas system in the treatment of genetic hearing loss will be more extensive.
Acknowledgements
This work was funded by the National Nature Science Foundation of China (No. 82071058) and the National Key Research and Development Program of China (No. 2021YFF0702303).
Disclosure of conflict of interest
None.
References
- 1.Haile LM, Kamenov K, Briant PS, Orji AU, Steinmetz JD, Abdoli A, Abdollahi M, Abu-Gharbieh E, Afshin A, Ahmed H, Ahmed Rashid T, Akalu Y, Alahdab F, Alanezi FM, Alanzi TM, Al Hamad H, Ali L, Alipour V, Al-Raddadi RM, Amu H, Arabloo J, Arab-Zozani M, Arulappan J, Ashbaugh C, Atnafu DD, Babar ZUD, Baig AA, Banik PC, Bärnighausen TW, Barrow A, Bender RG, Bhagavathula AS, Bhardwaj N, Bhardwaj P, Bibi S, Bijani A, Burkart K, Cederroth CR, Charan J, Choudhari SG, Chu DT, Couto RAS, Dagnew AB, Dagnew B, Dahlawi SMA, Dai X, Dandona L, Dandona R, Desalew A, Dhamnetiya D, Dhimal ML, Dhimal M, Doyle KE, Duncan BB, Ekholuenetale M, Filip I, Fischer F, Franklin RC, Gaidhane AM, Gaidhane S, Gallus S, Ghamari F, Ghashghaee A, Ghozali G, Gilani SA, Glavan IR, Golechha M, Goulart BNG, Gupta VB, Gupta VK, Hamidi S, Hammond BR, Hay SI, Hayat K, Heidari G, Hoffman HJ, Hopf KP, Hosseinzadeh M, Househ M, Hussain R, Hwang B-F, Iavicoli I, Ibitoye SE, Ilesanmi OS, Irvani SSN, Islam SMS, Iwagami M, Jacob L, Jayapal SK, Jha RP, Jonas JB, Kalhor R, Kameran Al-Salihi N, Kandel H, Kasa AS, Kayode GA, Khalilov R, Khan EA, Khatib MN, Kosen S, Koyanagi A, Kumar GA, Landires I, Lasrado S, Lim SS, Liu X, Lobo SW, Lugo A, Makki A, Mendoza W, Mersha AG, Mihretie KM, Miller TR, Misra S, Mohamed TA, Mohammadi M, Mohammadian-Hafshejani A, Mohammed A, Mokdad AH, Moni MA, Neupane Kandel S, Nguyen HLT, Nixon MR, Noubiap JJ, Nuñez-Samudio V, Oancea B, Oguoma VM, Olagunju AT, Olusanya BO, Olusanya JO, Orru H, Owolabi MO, Padubidri JR, Pakshir K, Pardhan S, Pashazadeh Kan F, Pasovic M, Pawar S, Pham HQ, Pinheiro M, Pourshams A, Rabiee N, Rabiee M, Radfar A, Rahim F, Rahimi-Movaghar V, Rahman MHU, Rahman M, Rahmani AM, Rana J, Rao CR, Rao SJ, Rashedi V, Rawaf DL, Rawaf S, Renzaho AMN, Rezapour A, Ripon RK, Rodrigues V, Rustagi N, Saeed U, Sahebkar A, Samy AM, Santric-Milicevic MM, Sathian B, Satpathy M, Sawhney M, Schlee W, Schmidt MI, Seylani A, Shaikh MA, Shannawaz M, Shiferaw WS, Siabani S, Singal A, Singh JA, Singh JK, Singhal D, Skryabin VY, Skryabina AA, Sotoudeh H, Spurlock EE, Taddele BW, Tamiru AT, Tareque MI, Thapar R, Tovani-Palone MR, Tran BX, Ullah S, Valadan Tahbaz S, Violante FS, Vlassov V, Vo B, Vongpradith A, Vu GT, Wei J, Yadollahpour A, Yahyazadeh Jabbari SH, Yeshaw Y, Yigit V, Yirdaw BW, Yonemoto N, Yu C, Yunusa I, Zamani M, Zastrozhin MS, Zastrozhina A, Zhang ZJ, Zhao JT, Murray CJL, Davis AC, Vos T, Chadha S. Hearing loss prevalence and years lived with disability, 1990-2019: findings from the Global Burden of Disease Study 2019. Lancet. 2021;397:996–1009. doi: 10.1016/S0140-6736(21)00516-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Smith RJ, Bale JF Jr, White KR. Sensorineural hearing loss in children. Lancet. 2005;365:879–890. doi: 10.1016/S0140-6736(05)71047-3. [DOI] [PubMed] [Google Scholar]
- 3.Kral A, O’Donoghue GM. Profound deafness in childhood. N Engl J Med. 2010;363:1438–1450. doi: 10.1056/NEJMra0911225. [DOI] [PubMed] [Google Scholar]
- 4.Ahmed H, Shubina-Oleinik O, Holt JR. Emerging gene therapies for genetic hearing loss. J Assoc Res Otolaryngol. 2017;18:649–670. doi: 10.1007/s10162-017-0634-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Yoshimura H, Nishio SY, Usami SI. Milestones toward cochlear gene therapy for patients with hereditary hearing loss. Laryngoscope Investig Otolaryngol. 2021;6:958–967. doi: 10.1002/lio2.633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Arnold C. What’s new in clinical CRISPR? Nat Med. 2021;27:184–185. doi: 10.1038/s41591-020-01222-4. [DOI] [PubMed] [Google Scholar]
- 7.Ishino Y, Shinagawa H, Makino K, Amemura M, Nakata A. Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene product. J Bacteriol. 1987;169:5429–5433. doi: 10.1128/jb.169.12.5429-5433.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337:816–821. doi: 10.1126/science.1225829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini LA, Zhang F. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013;339:819–823. doi: 10.1126/science.1231143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ma H, Marti-Gutierrez N, Park SW, Wu J, Lee Y, Suzuki K, Koski A, Ji D, Hayama T, Ahmed R, Darby H, Van Dyken C, Li Y, Kang E, Park AR, Kim D, Kim ST, Gong J, Gu Y, Xu X, Battaglia D, Krieg SA, Lee DM, Wu DH, Wolf DP, Heitner SB, Belmonte JCI, Amato P, Kim JS, Kaul S, Mitalipov S. Correction of a pathogenic gene mutation in human embryos. Nature. 2017;548:413–419. doi: 10.1038/nature23305. [DOI] [PubMed] [Google Scholar]
- 11.Schwank G, Koo BK, Sasselli V, Dekkers JF, Heo I, Demircan T, Sasaki N, Boymans S, Cuppen E, van der Ent CK, Nieuwenhuis EE, Beekman JM, Clevers H. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell. 2013;13:653–658. doi: 10.1016/j.stem.2013.11.002. [DOI] [PubMed] [Google Scholar]
- 12.Amoasii L, Hildyard JCW, Li H, Sanchez-Ortiz E, Mireault A, Caballero D, Harron R, Stathopoulou TR, Massey C, Shelton JM, Bassel-Duby R, Piercy RJ, Olson EN. Gene editing restores dystrophin expression in a canine model of Duchenne muscular dystrophy. Science. 2018;362:86–91. doi: 10.1126/science.aau1549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Yin H, Xue W, Chen S, Bogorad RL, Benedetti E, Grompe M, Koteliansky V, Sharp PA, Jacks T, Anderson DG. Genome editing with Cas9 in adult mice corrects a disease mutation and phenotype. Nat Biotechnol. 2014;32:551–553. doi: 10.1038/nbt.2884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Cox DB, Platt RJ, Zhang F. Therapeutic genome editing: prospects and challenges. Nat Med. 2015;21:121–131. doi: 10.1038/nm.3793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Makarova KS, Wolf YI, Iranzo J, Shmakov SA, Alkhnbashi OS, Brouns SJJ, Charpentier E, Cheng D, Haft DH, Horvath P, Moineau S, Mojica FJM, Scott D, Shah SA, Siksnys V, Terns MP, Venclovas C, White MF, Yakunin AF, Yan W, Zhang F, Garrett RA, Backofen R, van der Oost J, Barrangou R, Koonin EV. Evolutionary classification of CRISPR-Cas systems: a burst of class 2 and derived variants. Nat Rev Microbiol. 2020;18:67–83. doi: 10.1038/s41579-019-0299-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jinek M, Jiang F, Taylor DW, Sternberg SH, Kaya E, Ma E, Anders C, Hauer M, Zhou K, Lin S, Kaplan M, Iavarone AT, Charpentier E, Nogales E, Doudna JA. Structures of Cas9 endonucleases reveal RNA-mediated conformational activation. Science. 2014;343:1247997. doi: 10.1126/science.1247997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mir A, Alterman JF, Hassler MR, Debacker AJ, Hudgens E, Echeverria D, Brodsky MH, Khvorova A, Watts JK, Sontheimer EJ. Heavily and fully modified RNAs guide efficient SpyCas9-mediated genome editing. Nat Commun. 2018;9:2641. doi: 10.1038/s41467-018-05073-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Mao Y, Botella JR, Zhu JK. Heritability of targeted gene modifications induced by plant-optimized CRISPR systems. Cell Mol Life Sci. 2017;74:1075–1093. doi: 10.1007/s00018-016-2380-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Liu L, Li X, Ma J, Li Z, You L, Wang J, Wang M, Zhang X, Wang Y. The molecular architecture for RNA-guided RNA cleavage by Cas13a. Cell. 2017;170:714–726. e10. doi: 10.1016/j.cell.2017.06.050. [DOI] [PubMed] [Google Scholar]
- 20.Liu L, Li X, Wang J, Wang M, Chen P, Yin M, Li J, Sheng G, Wang Y. Two distant catalytic sites are responsible for C2c2 RNase activities. Cell. 2017;168:121–134. e12. doi: 10.1016/j.cell.2016.12.031. [DOI] [PubMed] [Google Scholar]
- 21.Perčulija V, Lin J, Zhang B, Ouyang S. Functional features and current applications of the RNA-targeting type VI CRISPR-Cas systems. Adv Sci (Weinh) 2021;8:2004685. doi: 10.1002/advs.202004685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Abudayyeh OO, Gootenberg JS, Essletzbichler P, Han S, Joung J, Belanto JJ, Verdine V, Cox DBT, Kellner MJ, Regev A, Lander ES, Voytas DF, Ting AY, Zhang F. RNA targeting with CRISPR-Cas13. Nature. 2017;550:280–284. doi: 10.1038/nature24049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Collias D, Beisel CL. CRISPR technologies and the search for the PAM-free nuclease. Nat Commun. 2021;12:555. doi: 10.1038/s41467-020-20633-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Yip BH. Recent advances in CRISPR/Cas9 delivery strategies. Biomolecules. 2020;10:839. doi: 10.3390/biom10060839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ishida K, Gee P, Hotta A. Minimizing off-target mutagenesis risks caused by programmable nucleases. Int J Mol Sci. 2015;16:24751–24771. doi: 10.3390/ijms161024751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Taha EA, Lee J, Hotta A. Delivery of CRISPR-Cas tools for in vivo genome editing therapy: trends and challenges. J Control Release. 2022;342:345–361. doi: 10.1016/j.jconrel.2022.01.013. [DOI] [PubMed] [Google Scholar]
- 27.Manno CS, Pierce GF, Arruda VR, Glader B, Ragni M, Rasko JJ, Ozelo MC, Hoots K, Blatt P, Konkle B, Dake M, Kaye R, Razavi M, Zajko A, Zehnder J, Rustagi PK, Nakai H, Chew A, Leonard D, Wright JF, Lessard RR, Sommer JM, Tigges M, Sabatino D, Luk A, Jiang H, Mingozzi F, Couto L, Ertl HC, High KA, Kay MA. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat Med. 2006;12:342–347. doi: 10.1038/nm1358. [DOI] [PubMed] [Google Scholar]
- 28.Boutin S, Monteilhet V, Veron P, Leborgne C, Benveniste O, Montus MF, Masurier C. Prevalence of serum IgG and neutralizing factors against adeno-associated virus (AAV) types 1, 2, 5, 6, 8, and 9 in the healthy population: implications for gene therapy using AAV vectors. Hum Gene Ther. 2010;21:704–712. doi: 10.1089/hum.2009.182. [DOI] [PubMed] [Google Scholar]
- 29.Hinderer C, Katz N, Buza EL, Dyer C, Goode T, Bell P, Richman LK, Wilson JM. Severe toxicity in nonhuman primates and piglets following high-dose intravenous administration of an adeno-associated virus vector expressing human SMN. Hum Gene Ther. 2018;29:285–298. doi: 10.1089/hum.2018.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Xu H, Wang B, Ono M, Kagita A, Fujii K, Sasakawa N, Ueda T, Gee P, Nishikawa M, Nomura M, Kitaoka F, Takahashi T, Okita K, Yoshida Y, Kaneko S, Hotta A. Targeted disruption of HLA genes via CRISPR-Cas9 generates iPSCs with enhanced immune compatibility. Cell Stem Cell. 2019;24:566–578. e7. doi: 10.1016/j.stem.2019.02.005. [DOI] [PubMed] [Google Scholar]
- 31.Stadtmauer EA, Fraietta JA, Davis MM, Cohen AD, Weber KL, Lancaster E, Mangan PA, Kulikovskaya I, Gupta M, Chen F, Tian L, Gonzalez VE, Xu J, Jung IY, Melenhorst JJ, Plesa G, Shea J, Matlawski T, Cervini A, Gaymon AL, Desjardins S, Lamontagne A, Salas-Mckee J, Fesnak A, Siegel DL, Levine BL, Jadlowsky JK, Young RM, Chew A, Hwang WT, Hexner EO, Carreno BM, Nobles CL, Bushman FD, Parker KR, Qi Y, Satpathy AT, Chang HY, Zhao Y, Lacey SF, June CH. CRISPR-engineered T cells in patients with refractory cancer. Science. 2020;367:eaba7365. doi: 10.1126/science.aba7365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lentacker I, De Cock I, Deckers R, De Smedt SC, Moonen CT. Understanding ultrasound induced sonoporation: definitions and underlying mechanisms. Adv Drug Deliv Rev. 2014;72:49–64. doi: 10.1016/j.addr.2013.11.008. [DOI] [PubMed] [Google Scholar]
- 33.Omichi R, Shibata SB, Morton CC, Smith RJH. Gene therapy for hearing loss. Hum Mol Genet. 2019;28:R65–R79. doi: 10.1093/hmg/ddz129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Yoshimura H, Shibata SB, Ranum PT, Smith RJH. Enhanced viral-mediated cochlear gene delivery in adult mice by combining canal fenestration with round window membrane inoculation. Sci Rep. 2018;8:2980. doi: 10.1038/s41598-018-21233-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Wang Y, Sun Y, Chang Q, Ahmad S, Zhou B, Kim Y, Li H, Lin X. Early postnatal virus inoculation into the scala media achieved extensive expression of exogenous green fluorescent protein in the inner ear and preserved auditory brainstem response thresholds. J Gene Med. 2013;15:123–133. doi: 10.1002/jgm.2701. [DOI] [PubMed] [Google Scholar]
- 36.Zuris JA, Thompson DB, Shu Y, Guilinger JP, Bessen JL, Hu JH, Maeder ML, Joung JK, Chen ZY, Liu DR. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat Biotechnol. 2015;33:73–80. doi: 10.1038/nbt.3081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Mianne J, Chessum L, Kumar S, Aguilar C, Codner G, Hutchison M, Parker A, Mallon AM, Wells S, Simon MM, Teboul L, Brown SD, Bowl MR. Correction of the auditory phenotype in C57BL/6N mice via CRISPR/Cas9-mediated homology directed repair. Genome Med. 2016;8:16. doi: 10.1186/s13073-016-0273-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Gu X, Hu X, Wang D, Xu Z, Wang F, Li D, Li GL, Yang H, Li H, Zuo E, Shu Y. Treatment of autosomal recessive hearing loss via in vivo CRISPR/Cas9-mediated optimized homology-directed repair in mice. Cell Res. 2022;32:699–702. doi: 10.1038/s41422-022-00624-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Gao X, Tao Y, Lamas V, Huang M, Yeh WH, Pan B, Hu YJ, Hu JH, Thompson DB, Shu Y, Li Y, Wang H, Yang S, Xu Q, Polley DB, Liberman MC, Kong WJ, Holt JR, Chen ZY, Liu DR. Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents. Nature. 2018;553:217–221. doi: 10.1038/nature25164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Noh B, Rim JH, Gopalappa R, Lin H, Kim KM, Kang MJ, Gee HY, Choi JY, Kim HH, Jung J. In vivo outer hair cell gene editing ameliorates progressive hearing loss in dominant-negative Kcnq4 murine model. Theranostics. 2022;12:2465–2482. doi: 10.7150/thno.67781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Xue Y, Hu X, Wang D, Li D, Li Y, Wang F, Huang M, Gu X, Xu Z, Zhou J, Wang J, Chai R, Shen J, Chen ZY, Li GL, Yang H, Li H, Zuo E, Shu Y. Gene editing in a Myo6 semi-dominant mouse model rescues auditory function. Mol Ther. 2022;30:105–118. doi: 10.1016/j.ymthe.2021.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Chandrasegaran S, Carroll D. Origins of programmable nucleases for genome engineering. J Mol Biol. 2016;428:963–989. doi: 10.1016/j.jmb.2015.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Johnson KR, Zheng QY, Noben-Trauth K. Strain background effects and genetic modifiers of hearing in mice. Brain Res. 2006;1091:79–88. doi: 10.1016/j.brainres.2006.02.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Liu L, Zou L, Li K, Hou H, Hu Q, Liu S, Li J, Song C, Chen J, Wang S, Wang Y, Li C, Du H, Li JL, Chen F, Xu Z, Sun W, Sun Q, Xiong W. Template-independent genome editing in the Pcdh15(av-3j) mouse, a model of human DFNB23 nonsyndromic deafness. Cell Rep. 2022;40:111061. doi: 10.1016/j.celrep.2022.111061. [DOI] [PubMed] [Google Scholar]
- 45.Long C, Amoasii L, Mireault AA, McAnally JR, Li H, Sanchez-Ortiz E, Bhattacharyya S, Shelton JM, Bassel-Duby R, Olson EN. Postnatal genome editing partially restores dystrophin expression in a mouse model of muscular dystrophy. Science. 2016;351:400–403. doi: 10.1126/science.aad5725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yang Y, Wang L, Bell P, McMenamin D, He Z, White J, Yu H, Xu C, Morizono H, Musunuru K, Batshaw ML, Wilson JM. A dual AAV system enables the Cas9-mediated correction of a metabolic liver disease in newborn mice. Nat Biotechnol. 2016;34:334–338. doi: 10.1038/nbt.3469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Yin H, Song CQ, Dorkin JR, Zhu LJ, Li Y, Wu Q, Park A, Yang J, Suresh S, Bizhanova A, Gupta A, Bolukbasi MF, Walsh S, Bogorad RL, Gao G, Weng Z, Dong Y, Koteliansky V, Wolfe SA, Langer R, Xue W, Anderson DG. Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo. Nat Biotechnol. 2016;34:328–333. doi: 10.1038/nbt.3471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Komor AC, Badran AH, Liu DR. CRISPR-based technologies for the manipulation of eukaryotic genomes. Cell. 2017;168:20–36. doi: 10.1016/j.cell.2016.10.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhao Y, Wang D, Zong L, Zhao F, Guan L, Zhang P, Shi W, Lan L, Wang H, Li Q, Han B, Yang L, Jin X, Wang J, Wang J, Wang Q. A novel DFNA36 mutation in TMC1 orthologous to the Beethoven (Bth) mouse associated with autosomal dominant hearing loss in a Chinese family. PLoS One. 2014;9:e97064. doi: 10.1371/journal.pone.0097064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kurima K, Peters LM, Yang Y, Riazuddin S, Ahmed ZM, Naz S, Arnaud D, Drury S, Mo J, Makishima T, Ghosh M, Menon PS, Deshmukh D, Oddoux C, Ostrer H, Khan S, Riazuddin S, Deininger PL, Hampton LL, Sullivan SL, Battey JF Jr, Keats BJ, Wilcox ER, Friedman TB, Griffith AJ. Dominant and recessive deafness caused by mutations of a novel gene, TMC1, required for cochlear hair-cell function. Nat Genet. 2002;30:277–284. doi: 10.1038/ng842. [DOI] [PubMed] [Google Scholar]
- 51.Kawashima Y, Kurima K, Pan B, Griffith AJ, Holt JR. Transmembrane channel-like (TMC) genes are required for auditory and vestibular mechanosensation. Pflugers Arch. 2015;467:85–94. doi: 10.1007/s00424-014-1582-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Vreugde S, Erven A, Kros CJ, Marcotti W, Fuchs H, Kurima K, Wilcox ER, Friedman TB, Griffith AJ, Balling R, Hrabé De Angelis M, Avraham KB, Steel KP. Beethoven, a mouse model for dominant, progressive hearing loss DFNA36. Nat Genet. 2002;30:257–258. doi: 10.1038/ng848. [DOI] [PubMed] [Google Scholar]
- 53.Melchionda S, Ahituv N, Bisceglia L, Sobe T, Glaser F, Rabionet R, Arbones ML, Notarangelo A, Di Iorio E, Carella M, Zelante L, Estivill X, Avraham KB, Gasparini P. MYO6, the human homologue of the gene responsible for deafness in Snell’s waltzer mice, is mutated in autosomal dominant nonsyndromic hearing loss. Am J Hum Genet. 2001;69:635–640. doi: 10.1086/323156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Allen F, Crepaldi L, Alsinet C, Strong AJ, Kleshchevnikov V, De Angeli P, Palenikova P, Khodak A, Kiselev V, Kosicki M, Bassett AR, Harding H, Galanty Y, Munoz-Martinez F, Metzakopian E, Jackson SP, Parts L. Predicting the mutations generated by repair of Cas9-induced double-strand breaks. Nat Biotechnol. 2018:10.1038/nbt.4317. doi: 10.1038/nbt.4317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Chakrabarti AM, Henser-Brownhill T, Monserrat J, Poetsch AR, Luscombe NM, Scaffidi P. Target-specific precision of CRISPR-mediated genome editing. Mol Cell. 2019;73:699–713. e6. doi: 10.1016/j.molcel.2018.11.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Leenay RT, Aghazadeh A, Hiatt J, Tse D, Roth TL, Apathy R, Shifrut E, Hultquist JF, Krogan N, Wu Z, Cirolia G, Canaj H, Leonetti MD, Marson A, May AP, Zou J. Large dataset enables prediction of repair after CRISPR-Cas9 editing in primary T cells. Nat Biotechnol. 2019;37:1034–1037. doi: 10.1038/s41587-019-0203-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Roman-Rodriguez FJ, Ugalde L, Alvarez L, Diez B, Ramirez MJ, Risueno C, Corton M, Bogliolo M, Bernal S, March F, Ayuso C, Hanenberg H, Sevilla J, Rodriguez-Perales S, Torres-Ruiz R, Surralles J, Bueren JA, Rio P. NHEJ-mediated repair of CRISPR-Cas9-induced DNA breaks efficiently corrects mutations in HSPCs from patients with fanconi anemia. Cell Stem Cell. 2019;25:607–621. e7. doi: 10.1016/j.stem.2019.08.016. [DOI] [PubMed] [Google Scholar]
- 58.Shen MW, Arbab M, Hsu JY, Worstell D, Culbertson SJ, Krabbe O, Cassa CA, Liu DR, Gifford DK, Sherwood RI. Predictable and precise template-free CRISPR editing of pathogenic variants. Nature. 2018;563:646–651. doi: 10.1038/s41586-018-0686-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.van Overbeek M, Capurso D, Carter MM, Thompson MS, Frias E, Russ C, Reece-Hoyes JS, Nye C, Gradia S, Vidal B, Zheng J, Hoffman GR, Fuller CK, May AP. DNA repair profiling reveals nonrandom outcomes at Cas9-mediated breaks. Mol Cell. 2016;63:633–646. doi: 10.1016/j.molcel.2016.06.037. [DOI] [PubMed] [Google Scholar]
- 60.Kazmierczak P, Sakaguchi H, Tokita J, Wilson-Kubalek EM, Milligan RA, Müller U, Kachar B. Cadherin 23 and protocadherin 15 interact to form tip-link filaments in sensory hair cells. Nature. 2007;449:87–91. doi: 10.1038/nature06091. [DOI] [PubMed] [Google Scholar]
- 61.Alagramam KN, Murcia CL, Kwon HY, Pawlowski KS, Wright CG, Woychik RP. The mouse Ames waltzer hearing-loss mutant is caused by mutation of Pcdh15, a novel protocadherin gene. Nat Genet. 2001;27:99–102. doi: 10.1038/83837. [DOI] [PubMed] [Google Scholar]
- 62.Yeh WH, Shubina-Oleinik O, Levy JM, Pan B, Newby GA, Wornow M, Burt R, Chen JC, Holt JR, Liu DR. In vivo base editing restores sensory transduction and transiently improves auditory function in a mouse model of recessive deafness. Sci Transl Med. 2020;12:eaay9101. doi: 10.1126/scitranslmed.aay9101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zheng Z, Li G, Cui C, Wang F, Wang X, Xu Z, Guo H, Chen Y, Tang H, Wang D, Huang M, Chen ZY, Huang X, Li H, Li GL, Hu X, Shu Y. Preventing autosomal-dominant hearing loss in Bth mice with CRISPR/CasRx-based RNA editing. Signal Transduct Target Ther. 2022;7:79. doi: 10.1038/s41392-022-00893-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Xu C, Zhou Y, Xiao Q, He B, Geng G, Wang Z, Cao B, Dong X, Bai W, Wang Y, Wang X, Zhou D, Yuan T, Huo X, Lai J, Yang H. Programmable RNA editing with compact CRISPR-Cas13 systems from uncultivated microbes. Nat Methods. 2021;18:499–506. doi: 10.1038/s41592-021-01124-4. [DOI] [PubMed] [Google Scholar]
- 65.Kannan S, Altae-Tran H, Jin X, Madigan VJ, Oshiro R, Makarova KS, Koonin EV, Zhang F. Compact RNA editors with small Cas13 proteins. Nat Biotechnol. 2022;40:194–197. doi: 10.1038/s41587-021-01030-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Xiao Q, Xu Z, Xue Y, Xu C, Han L, Liu Y, Wang F, Zhang R, Han S, Wang X, Li GL, Li H, Yang H, Shu Y. Rescue of autosomal dominant hearing loss by in vivo delivery of mini dCas13X-derived RNA base editor. Sci Transl Med. 2022;14:eabn0449. doi: 10.1126/scitranslmed.abn0449. [DOI] [PubMed] [Google Scholar]
- 67.György B, Nist-Lund C, Pan B, Asai Y, Karavitaki KD, Kleinstiver BP, Garcia SP, Zaborowski MP, Solanes P, Spataro S, Schneider BL, Joung JK, Géléoc GSG, Holt JR, Corey DP. Allele-specific gene editing prevents deafness in a model of dominant progressive hearing loss. Nat Med. 2019;25:1123–1130. doi: 10.1038/s41591-019-0500-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wu J, Solanes P, Nist-Lund C, Spataro S, Shubina-Oleinik O, Marcovich I, Goldberg H, Schneider BL, Holt JR. Single and dual vector gene therapy with AAV9-PHP.B rescues hearing in Tmc1 mutant mice. Mol Ther. 2021;29:973–988. doi: 10.1016/j.ymthe.2020.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Cui C, Wang D, Huang B, Wang F, Chen Y, Lv J, Zhang L, Han L, Liu D, Chen ZY, Li GL, Li H, Shu Y. Precise detection of CRISPR-Cas9 editing in hair cells in the treatment of autosomal dominant hearing loss. Mol Ther Nucleic Acids. 2022;29:400–412. doi: 10.1016/j.omtn.2022.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]