Next Article in Journal
Stage-Dependent Activity and Pro-Chondrogenic Function of PI3K/AKT during Cartilage Neogenesis from Mesenchymal Stromal Cells
Next Article in Special Issue
CRISPR-Cas9 Technology for the Creation of Biological Avatars Capable of Modeling and Treating Pathologies: From Discovery to the Latest Improvements
Previous Article in Journal
HIV-Associated Insults Modulate ADAM10 and Its Regulator Sirtuin1 in an NMDA Receptor-Dependent Manner
Previous Article in Special Issue
Recent Advances in Improving Gene-Editing Specificity through CRISPR–Cas9 Nuclease Engineering
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CRISPR-Based Therapeutic Gene Editing for Duchenne Muscular Dystrophy: Advances, Challenges and Perspectives

1
Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
2
Department of Anesthesiology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
3
Shanghai Institute of Precision Medicine, Shanghai 200125, China
4
HUIGENE Therapeutics Co., Ltd., Shanghai 200131, China
5
Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
6
School of Medicine, Wayne State University, Detroit, MI 48201, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2022, 11(19), 2964; https://doi.org/10.3390/cells11192964
Submission received: 11 August 2022 / Revised: 6 September 2022 / Accepted: 9 September 2022 / Published: 22 September 2022
(This article belongs to the Special Issue CRISPR-Based Genome Editing in Translational Research)

Abstract

:
Duchenne muscular dystrophy (DMD) is a severe neuromuscular disease arising from loss-of-function mutations in the dystrophin gene and characterized by progressive muscle degeneration, respiratory insufficiency, cardiac failure, and premature death by the age of thirty. Albeit DMD is one of the most common types of fatal genetic diseases, there is no curative treatment for this devastating disorder. In recent years, gene editing via the clustered regularly interspaced short palindromic repeats (CRISPR) system has paved a new path toward correcting pathological mutations at the genetic source, thus enabling the permanent restoration of dystrophin expression and function throughout the musculature. To date, the therapeutic benefits of CRISPR genome-editing systems have been successfully demonstrated in human cells, rodents, canines, and piglets with diverse DMD mutations. Nevertheless, there remain some nonignorable challenges to be solved before the clinical application of CRISPR-based gene therapy. Herein, we provide an overview of therapeutic CRISPR genome-editing systems, summarize recent advancements in their applications in DMD contexts, and discuss several potential obstacles lying ahead of clinical translation.

1. Introduction

DMD, the most prevalent genetic muscular disease in man, is attributed to diverse mutations in the X-chromosome-resident dystrophin gene and affects approximately 1 in 3500 to 5000 newborn boys worldwide [1]. The dystrophin gene is the largest known human gene encompassing 2.6 million base pairs and contains 79 exons that encode a massive 427 kDa dystrophin protein [2,3]. The dystrophin protein, located underneath the sarcolemma, functions as a key mechanical anchor to connect the intracellular cytoskeleton to the inner surface of the muscle fiber membrane, maintaining sarcolemmal integrity and supporting muscle structure. Additionally, dystrophin plays a crucial role as the molecular scaffold to coordinate the assembly of numerous signaling molecules (e.g., nitric oxide synthase and ion channels), which work in concert to ensure the normal functioning of muscles [4,5]. However, the absence of dystrophin protein in DMD patients leads to muscle membrane fragility, myocyte necrosis, inflammatory infiltration, myocardial fibrosis, and progressive muscle weakness [2,6]. Due to the huge size of the dystrophin gene, more than 7000 pathological mutations, ranging from deletions, duplications, and point mutations to other small gene arrangements, have been discovered in DMD patients [7]. Most of these mutations (~75%) are intragenic deletions or duplications of one or multiple exons and primarily cluster into two hotspot regions between exons 2–22 and exons 43–55, whereas other small mutations (e.g., insertions and nonsense mutations) randomly occur throughout the dystrophin gene [7,8]. Pathologically, the majority of DMD mutations destroy the open reading frames (ORFs) or create a premature stop codon in the transcripts, which leads to aberrant translation and the generation of nonfunctional dystrophin. With the progression of DMD, dystrophin deficiency eventually results in the loss of ambulation, respiratory failure, cardiomyopathy, and premature death in early adulthood [6]. Unfortunately, although DMD is devastating, there exists no curative therapy for this lethal disease. At present, both corticosteroid and antisense oligonucleotide (AON) treatments are available for the mitigation of the symptoms of this disease [9,10,11], but they fail to remove the underlying genetic mutations from the dystrophin gene. It has been suggested that the current therapeutic approaches are offered mainly only for the alleviation of secondary manifestations of DMD, such as inflammation, fibrosis, mitochondrial dysfunction, impaired angiogenesis, or calcium dyshomeostasis [12,13,14,15,16,17,18,19]. Moreover, the long-term use of corticosteroids has been found to minimally ameliorate DMD phenotypes and cause many adverse effects, including growth delay and bone weakness [12,20,21]. Through the induction of the skipping of exon 45, 51, or 53 in dystrophin transcripts, four AON medicines have been approved to treat DMD patients with particular mutations in the exon 43–55 hotspot, but they usually restore dystrophin protein expression to less than 1% of the normal level after a year of continuous administration [22,23,24,25,26]. The broad application of AON medicines is restrained by various factors, including reduplicative administration, high cost, and poor delivery efficiency, particularly in the heart, probably due to their short half-lives [27,28]. Hence, there remains a great unmet need to develop innovative therapeutic strategies for correcting genetic mutations and restoring functional dystrophin generation in DMD individuals.
Based on its simplicity and precision, CRISPR-mediated genome engineering offers a promising therapeutic approach to restoring dystrophin expression and muscular functions in DMD individuals via eliminating pathological mutations at the genomic level [29,30]. Thus, a single administration of CRISPR genome-editing components can cure DMD in theory. The CRISPR system is composed of two major components, one CRISPR-associated (Cas) endonuclease and the other a single-guide RNA (sgRNA) complementary to the target genomic sequence [31,32]. Under the guidance of the sgRNA, Cas endonuclease directly binds to the target genomic sites adjacent to the protospacer-adjacent motif (PAM), creating DNA double-strand breaks (DSBs). In mammalian cells, these site-specific DSBs are typically resolved by endogenous cellular repair pathways, either non-homologous end joining (NHEJ) or homology-directed repair (HDR) [33]. The choice of DSB repair pathway depends on the cell type, cellular proliferation status, and the absence or presence of an exogenous DNA template. Thus far, CRISPR systems have been widely employed to correct diverse DMD-causing mutations not only in human DMD myoblasts and induced pluripotent stem cells (iPSCs) but also in preclinical DMD animal models such as mice, dogs, and pigs [34,35,36,37,38,39,40,41,42]. After the single systemic administration of Cas9 system components, both genomic editing and dystrophin restoration have been shown to persist for at least 18 months in the mdx mice harboring a point mutation in exon 23 and the DMD mouse model with an exon 44 deletion (ΔEx44) mutation [35,43,44], highlighting the durability of CRISPR therapeutics in dystrophic mice. Since adult human cardiomyocytes have an extremely low turnover rate over time [45], CRISPR gene therapy will most likely lead to lifelong benefits when used to treat DMD patients. However, clinical studies assessing the efficacy and safety of CRISPR gene therapy in DMD patients are still missing. Therefore, to facilitate the clinical translation of therapeutic CRISPR gene editing for DMD, more efforts are imminently necessary to address the multiple challenges, including but not limited to safe dosage, the in vivo delivery strategy, immunogenicity, and the extent of dystrophin restoration.
In this paper, we overview recent advances in the knowledge of CRISPR gene therapy for DMD and discuss several challenges in the clinical application of therapeutic CRISPR gene editing.

2. Dystrophin

There exist multiple different isoforms of dystrophin transcripts originating from seven unique promoters and alternative splicing [46]. Remarkably, the splicing patterns of 79 exons are highly conserved across vertebrate species [47], which aids the development of therapeutic medicines via evaluating the efficacy and mechanisms of potential medicines in preclinical DMD animals. Among these isoforms, the largest transcript driven by the promoter upstream of exon 1 is 14 kb in size and encodes for the 427 kDa dystrophin protein with 3684 amino acids. This full-length dystrophin is expressed in all skeletal muscles, heart, vascular and visceral smooth muscles, as well as some neural cells [48]. The dystrophin protein consists of four distinct functional domains in the following order: an actin-binding domain at the N-terminus, a central rod region with 24 successive spectrin-like repeats (SLRs), a cysteine-rich domain binding β-dystroglycan, and a C-terminal domain interacting with dystrobrevin and syntrophin [2,41]. Thus, dystrophin serves as an organizing center for the dystrophin-glycoprotein complex (DGC), linking the intracellular cytoskeleton to the extracellular matrix across the sarcolemma. Beyond dystrophin, other DGC components comprise an extracellular α-dystroglycan binding laminin-2, a trans-membrane β-dystroglycan, a membrane-resident sarcospan, and four trans-membrane sarcoglycans (α-, β-, γ-, and δ-sarcoglycan) [49]. Meanwhile, dystrophin has a direct association with neuronal nitric oxide synthase (nNOS) via two SLRs (R16 and R17) [5], localizing the nNOS near the sarcolemma and modulating nitric oxide signaling in muscle cells. It has been observed that DGC may associate with the proteins engaged in calcium homeostasis, such as plasma membrane calcium ATPase and calcium channels [50,51]. It is likely that the modulation of DGC on calcium signaling happens at the sarcoplasmic membrane level in dystrophic cells [50].
The deficiency of functional dystrophin results in the mislocalization of DGC components, dysregulation of calcium balance, abnormal nitric oxide signaling, mitochondrial dysfunctions, increased oxidative stress, defective energy metabolism, impaired autophagy, insufficient angiogenesis, and aberrant inflammation [51,52,53,54,55,56,57,58,59]. Intracellular calcium is known to be abnormally elevated in dystrophic cells owing to the activation of calcium channels in the sarcoplasmic membrane and the decreased calcium handling [51,52,53]. This elevation of cytosolic free calcium leads to an overload of calcium in the mitochondria and other organelles such as the endoplasmic reticulum, which in turn conduces to the augmentation of oxidative stress and the impairment of mitochondrial respiration and ATP production [54,55,56,60]. Consistent with these scenarios, defective energy metabolism is present in the dystrophic cells, partially evidenced by insufficient glucose utilization and fatty acid oxidation [57]. On the other hand, excessive cytosolic calcium and disturbed cellular homeostasis not only trigger the activation of calcium-sensitive proteases (e.g., calpains and phospholipase A2) but also promote the release of diverse cytokines and chemokines into the extracellular space. Such aberrant events provoke the infiltration of immune cells (e.g., macrophages and neutrophils) into dystrophic muscles [61], contributing to myofiber necrosis and muscle destruction. As time goes on, continuous cycles of muscle damage and regeneration progressively allow the replacement of dystrophic muscles with fibro-fatty connective tissue, accompanied by the loss of muscle mass and functions. It is noteworthy that muscle damage and membrane leakage are obligated to the unnatural elevations of serum creatine kinase and lactate dehydrogenase in DMD individuals [62]. These myriad dysfunctions have been well documented in the skeletal and cardiac muscles of both DMD patients and animal models [6,63,64,65].
Both the N terminus linking the actin filaments and the C terminus binding the DGC components are essential for dystrophin functions [66], whose mutations lead to a complete loss of dystrophin protein. On the contrary, the median domain having redundant rod repeats could be shortened and generates the internally truncated dystrophin forms with partial functions. It has been shown that a central domain with as few as 4 spectrin-like repeats enables truncated dystrophin to be partially functional [67]. Under natural conditions, internally shortened dystrophin proteins can be observed in patients with Becker muscular dystrophy (BMD) resulting from the in-frame mutations of the dystrophin gene [68]. Thus, BMD patients show relatively mild symptoms in general, and some patients may remain asymptomatic until later life [69,70]. Likewise, micro-dystrophin, a truncated but partially functional protein lacking redundant rod repeats, only contains a minimal number of functional domains and has been shown to ameliorate DMD symptoms in many preclinical animal models [71,72,73,74]. The adeno-associated virus (AAV)-mediated single administration of micro-dystrophin (e.g., SRP-9001, PF-06939926, and SGT-001) is still being evaluated in several clinical studies involving DMD boys (http://clinicaltrials.gov). Despite the tolerance of some DMD patients to micro-dystrophin therapy [49,75], the spatiotemporal expression of micro-dystrophin is actually modulated by an exogenous promoter within the AAV, which may lead to uncontrollable protein localization and level. On the contrary, the expression patterns of CRISPR-corrected dystrophin transcripts and proteins are controlled by endogenous promoters, representing an attractive advantage of CRISPR genome engineering.

3. CRISPR Systems

The CRISPR-Cas system was originally discovered as an adaptive defense system in bacteria and archaea against foreign viral pathogens [76,77,78] and has been harnessed for genomic editing in eukaryotic cells. Based on Cas type and number, six CRISPR systems (I–VI) are grouped into two distinct classes: the class 1 system, comprising types I, III and IV, needs multiple Cas effectors at once; the class 2 system, containing types II, V and VI, utilizes one single Cas endonuclease [79]. Most type-II Cas9 and type-V Cas12 proteins act on DNA and introduce the DSBs in a programmable manner, whereas type-VI Cas13 proteins cleave the RNA transcripts specifically via their RNA-targeting nuclease activity [32,80]. As for the manner of cleavage, Cas9 proteins with RuvC and HNH nuclease domains primarily create blunt-end DSBs in the protospacer sequence 3 nucleotides upstream of the PAM [76], while Cas12 variants possessing a single RuvC-like nuclease domain typically make the sticky ends with five-nucleotide overhangs in the PAM-distal regions of the protospacer [81]. It has been reported that Streptococcus pyogenes Cas9 (SpCas9) may cut DNA in a staggered orientation and leave a single-nucleotide overhang at the broken point [82,83]. Following DNA cleavage, the HDR pathway is activated to repair the DSBs in the presence of an exogenous donor template and introduces the desired modification into the genome at the target locus. Nonetheless, HDR is only active in the proliferative cells owing to its requirement of some proteins expressed in the S and G2 cell-cycle phases [84,85], so it has a very low efficiency (less than 2%) in post-mitotic cells such as cardiomyocytes and myofibers. On the contrary, the NHEJ machinery turns to fix the DSBs in the absence of an exogenous template and often generates small insertions or deletions around the broken site. Unlike HDR, NHEJ is functional throughout the cell cycle and operates at high efficiency not only in dividing cells but also in post-mitotic cells [86]. Thus, it is believed that NHEJ serves as the predominant repair pathway in most mammalian cells.
The diversification of Cas proteins is extremely remarkable in terms of the bacterial source, protein size, PAM sequence, suitable spacer length, and editing efficiency and specificity. For example, the most often used SpCas9 with 1368 amino acids specifically recognizes the tri-nucleotide PAM sequences of 5′-NGG or -NAG that are common in the human genome [76,77,87]. By comparison, Staphylococcus aureus Cas9 (SaCas9, 1053 amino acids) and Campylobacter jejuni Cas9 (CjCas9, 984 amino acids) utilize the longer but relatively restrictive PAM sequence of 5′-NNGRRT or 5′-NNNVRYM, respectively [88,89]. It should be noted that both SaCas9 (~3.2 kb) and CjCas9 (~2.95 kb) are much smaller than the genome packaging limit of AAV vectors (~4.7 kb) [90], enabling them to be efficiently delivered in vivo by conventional AAV vectors. In the cases of Cas12 effectors, Lachnospiraceae bacterium Cas12a (LbCas12a, 1228 amino acids) and Acidaminococcus sp. Cas12a (AsCas12a, 1307 amino acids) are more efficient and widely adopted than other Cas12 variants in mammalian cells via targeting the T-rich PAM of 5′-TTN [81,91,92]. Beyond these five Cas proteins, an enormous variety of Cas9 and Cas12 enzymes have been discovered and characterized from diverse bacterial species, including Neisseria meningitidis Cas9 (NmeCas9), Streptococcus thermophilus Cas9 (StCas9), Francisella novicida Cas9 (FnCas9), Alicyclobacillus acidoterrestris Cas12b (AacCas12b), Bacillus hisashii Cas12b (BhCas12b), Oleiphilus sp. Cas12c (OspCas12c), Deltaproteobacteria Cas12e (DpbCas12e), and Planctomycetes Cas12e (PlmCas12e) [79,93,94,95,96,97,98,99,100,101]. Additionally, many naturally occurring Cas proteins have been engineered to improve their PAM availability, enhance on-target efficiency, and/or reduce off-target activity [32,79]. Hitherto, a great deal of engineered Cas9 and Cas12 proteins with high targetability and specificity have already been made, such as eSpCas9, Spy-mac Cas9, HypaCas9, evoCas9, HeFSpCas9, xCas9, HiFiCas9, Sniper-Cas9, SpCas9-HF1, SpCas9-NG, FnCas9-RHA, SaCas9-KKH, AsCas12a-RVR, and enAsCas12a [102,103,104,105,106,107,108,109,110,111,112,113,114]. As new Cas9 and Cas12 variants unceasingly emerge, the toolkit of CRISPR gene-editing systems is getting expanded by both natural and engineered Cas proteins, thus offering more and more choices for CRISPR therapeutics.
An important engineering direction is to modify the nuclease domains of Cas enzymes to generate catalytically impaired nickase Cas (nCas) or catalytically inactivated dead Cas (dCas) variants while retaining their programmable DNA-binding ability [32,115]. In this regard, both nCas and dCas proteins have been fused with a cytidine deaminase or adenosine deaminase for single-base conversions or with an engineered reverse transcriptase for short insertions and deletions [116,117,118]. The fusion of nCas or dCas proteins with the cytidine deaminase APOBEC1 causes the precise transition from C•G to T•A base pairs [119], while the combination of nCas or dCas variants with the adenosine deaminase TadA accurately converts the targeted A•T base pairs to G•C [120]. Fusing an nCas effector with a reverse transcriptase can induce all base pair transitions, small insertions, and/or short deletions in a targeted and precise way [118,121]. The fusion protein between dCas9 and transcriptional activator VP160 could drastically increase the expression level of dystrophin homolog utrophin via targeting its promoters [122,123], which represents a promising compensatory approach for DMD treatment. Unlike conventional Cas enzymes, both nCas and dCas proteins are unable to introduce DSBs into the genome, making these nCas- or dCas-based CRISPR tools especially safe for genome editing and disease therapy.

4. CRISPR-Driven Therapeutic Strategies

The first demonstration of CRISPR gene-editing therapy is in the mdx mice through the direct zygote injection of SpCas9, sgRNAs, and single-stranded oligodeoxynucleotide (ssODN) [124]. Due to ethical issues and public policies, this germline editing approach is likely unavailable for DMD treatment in humans. Therefore, accumulating evidence instead validates the in vivo therapeutic benefits of CRISPR gene editing systems in postnatal DMD animals [36,37,39,40,43,44,125,126,127,128,129]. The leading strategies for the CRISPR-mediated therapeutic correction of DMD mutations are exon excision, exon skipping, exon reframing, exon knockin, base editing, and prime editing (Table 1 and Table 2).

5. Double-Cut Exon Excision

The removal of one or more exons by CRISPR gene editing applies to approximately 85% of all DMD patients bearing exon duplications, deletions, or point mutations. Two sgRNAs flanking either side of the mutant exons are designed to simultaneously cut the target genomic sites in the presence of Cas proteins, resulting in the complete excision of mutant exons (Figure 1A). Consequently, this kind of exon excision restores the dystrophin reading frame and the expression of functionally truncated dystrophin protein. It has been shown that this exon excision strategy is particularly suitable for correcting exon duplication mutations [122,125,150,151,152,153]. Exonic duplications are estimated to comprise 10–15% of all DMD mutations [152,164,165], making them the third most common cause of DMD. Under the guidance of two sgRNAs targeting a duplicated intronic region, SpCas9 precisely excludes a tandem duplication of exon 2. It repairs the expression of 7–11% of full-length dystrophin protein in human DMD myoblasts with an exon 2 duplication [151]. Likewise, the SpCas9 system has been employed to remove exon 18-30 duplication and induce full-length dystrophin restoration in human DMD myoblasts with exon 18-30 duplications (Dup18-30) [122]. Intravenously administrating AAV-SaCas9 system in DMD Dup18-30 mice could ensure full-length dystrophin protein expression in cardiac and skeletal muscles ranging from 4% to 25% of the normal levels [125]. These levels of functional dystrophin restoration ameliorate dystrophic pathology, enhance muscle strength, and improve open-filed activity in SaCas9-corrected Dup18-30 mice [125]. This observation aligns with the notion that as little as 3–14% of full-length dystrophin protein significantly benefits muscle functions [166,167]. Intriguingly, the elimination of a duplication event can be achieved readily with one sgRNA targeting the duplicated intronic region.
CRISPR-mediated exon excision is useful for correcting exonic deletion mutations, especially multi-exon deletions, in the dystrophin gene. This exon excision approach is supported in nature by the asymptomatic or mild symptoms of BMD patients with small in-frame deletions in the exon 45-55 mutation hotspot region [154]. It should be noted that the excision of the exon 45-55 hotspot region could be used as a treatment for more than 60% of DMD patients regardless of mutation type. For example, two different sgRNAs, one targeting intron 44 and the other targeting intron 55, are utilized to remove the entire exon 45-55 region in the presence of SpCas9, which in turn generates an internally truncated dystrophin protein in human DMD myoblasts with an exon 48-50 deletion (ΔEx48-50) [154]. This large excision of the exon 45-55 region efficiently repairs dystrophin protein expression, stabilizes the DGC complex, and improves membrane integrity in both cardiomyocytes and skeletal muscles from human DMD iPSCs harboring an exon 46-51 deletion (ΔEx46-51) mutation [38]. The restoration of functional dystrophin transcripts and protein following the excision of the exon 45-55 region has also been demonstrated in human DMD myoblasts with an exon 51 deletion (ΔEx51) mutation and a humanized DMD mouse model carrying the exon 45-deleted human dystrophin gene (hDMDΔ45/mdxD2) [130,155]. Notably, the efficiencies of the SpCas9 system when reframing the ΔEx51 mutation and restoring dystrophin expression are comparable to that of the AsCas12a system in human DMD ΔEx51 myoblasts and a patient-derived xenograft DMD mouse model [155]. Likewise, the combination of either SpCas9 or eSpCas9(1.1) with two sgRNAs targeting introns 43 and 54 is found to abscise the exon 44-54 region and generate a functional dystrophin protein in human DMD myoblasts with ΔEx48-50 or ΔEx45-52 mutation [156,157,158]. Intriguingly, the utilization of SaCas9 and two sgRNAs targeting exons 47 and 58 leads to the formation of a hybrid exon 47-58 lacking their internal large region and the expression of 360 kDa dystrophin protein in human DMD myoblasts with ΔEx49-50, ΔEx50-52, ΔEx51-53, or ΔEx51-56 mutation [40]. The systemic AAV delivery of SaCas9 components is further reported to restore functional dystrophin expression in the heart of humanized DMD mice with an exon 52-deleted human dystrophin gene (hDMDΔ52/mdx) [40]. Apart from the above multi-exon excision, the elimination of single exon 51 or 53 by the SpCas9 system has also been proved to repair the dystrophin reading frame in human DMD ΔEx48-50 or ΔEx45-52 myoblasts, respectively [154,156,157]. Moreover, the single systemic administration of SpCas9 components by AAV9 vectors in DMD ΔEx52 pigs restores dystrophin protein expression throughout muscle tissues, ranging from 12% to 54% of normal levels, which gives rise to the alleviation of muscle pathology, the improvement of skeletal muscle and cardiac functions, and the extension of porcine lifespan [39]. Notably, SpCas9-driven exon excision has been taken to handle the deletion mutation in the N-terminal exon 2-20 hotspot [159]. Three distinct excision approaches are designed to separately remove exons 3-7, 6-7, or 7-11 in human DMD iPSCs with an exon 8-9 deletion (ΔEx8-9) mutation. The exon 3-9 excision is the most effective strategy in restoring the contractility and calcium transits of DMD ΔEx8-9 iPSC-derived cardiomyocytes, whereas the exon 7-11 excision causes the minimal recovery of cardiomyocyte functionality due to the generation of a structurally unstable dystrophin protein [159]. When facing the mutations in N- and C-terminal domains, specific considerations need to be paid to retain the essential amino acid residues for functional dystrophin protein restoration.
Beyond exon duplication or deletion mutations, the exon excision strategy has been adopted to reframe the out-of-frame point mutations comprising ~27% of all DMD cases both in vitro and in vivo [34,35,36,37,43,126,127,128,129,131,160,168]. For example, AAVrh74-delivered SaCas9 and two sgRNAs targeting introns 20 and 23 in neonatal mdx mice are found to remove the exon 21-23 region with a nonsense mutation, prevent cardiomyopathy, and improve cardiac functions [43]. These benefits following systemic AAV-SaCas9 therapy can be sustained for up to 19 months without the occurrence of tumorigenicity and organ toxicity [43]. Likewise, the systemic administration of either SaCas9 or SpCas9 with two sgRNAs targeting introns 51 and 53 produces widespread dystrophin restoration in the cardiac, diaphragmatic and skeletal muscles of DMD mdx4Cv mice harboring a point mutation in exon 53 [128,129]. The genomic editing efficiency of systemic AAV6-Cas9 treatment is stable in mouse cardiomyocytes rather than their skeletal muscles [129]. Intramuscularly administrating SpCas9 or SaCas9 components in DMD mdx4Cv mice repairs dystrophin protein expression in up to 68% of skeletal myofibers, improves skeletal muscle structure, and boosts muscle forces [128]. Thus far, a single exon 23 excision using either SaCas9 or SpCas9 system has been validated in neonatal and adult mdx mice [34,35,36,37,127]. In neonatal mdx mice, systemically infused SaCas9 or SpCas9 components are capable of rescuing dystrophin protein expression in body-wide muscle tissues [34,36,37,127]. The local administration of the AAV-SaCas9 system in adult mdx mice leads to improved muscle morphology, ameliorated nNOS localization, and enhanced skeletal muscle force [36,127]. An interesting finding is that genomic editing efficiency and the extent of dystrophin restoration in the heart of systemically treated mdx mice increase as the mice age [37], which is probably due to the survival disadvantage of dystrophic cardiomyocytes.
Two major concerns limit the clinical application of double-cut exon excision at its current iterations. The first is its low editing efficiency, which may be attributed to the indispensability of two cooperative cutting across large genomic intervening regions. Another is the generation of diversely unpredictable genome modifications such as DNA inversion and AAV integration [37].

6. Single-Cut Exon Skipping and Reframing

Single-cut gene editing has emerged as a promising alternative strategy for the efficient and safe correction of diverse DMD mutations [41,169]. In this approach, one single sgRNA is designed to target the vicinity of the intron-exon boundary and splice signal sequences. Its utilization, together with the Cas enzyme, performs one single cutting. This single DSB is rejoined later by endogenous NHEJ pathways, introducing small insertions or deletions into the target loci. There are two repair outcomes: (1) exon skipping happens as small deletions abolish the splice consensus sites of out-of-frame exons (Figure 1B); (2) exon reframing occurs when an appropriate number of nucleotide deletions or insertions appear in the exonic region (Figure 1C). In theory, approximately one-third of single-cut editing events hold the promise to put the dystrophin gene back in the frame [42]. Indeed, SpCas9-mediated single cutting in exon 51 generates a large fraction of exon reframing events among all indels via preferentially inserting one single adenosine [138], while the reframing of exon 51 by the SaCas9-KKH system may even account for ~80% of all editing events [133]. Regardless of whether conducting exon skipping or exon reframing, the permanent restoration of the dystrophin reading frame and protein expression is eventually achieved in muscle cells. More than 80% of DMD patients are estimated to benefit from this therapeutic strategy [42]. Compared to double-cut exon excision, both exon skipping and exon reframing via single-cut gene editing possess many advantages, including but not limited to a high editing efficiency, low frequency of off-target events, and minimum genomic modifications.
Single-cut editing strategy is very efficient for reconstituting the dystrophin reading frame and expression in DMD mice and dogs with an exon 50 deletion (ΔEx50) mutation, representing one of the most common single exonic deletions in humans [133,135,138,139,140]. The AAV9-based intramuscular delivery of SpCas9 and one sgRNA in young DMD ΔEx50 mice and dogs can rescue dystrophin expression in nearly all skeletal myofibers and prevent the onset of skeletal muscle pathology [138,139,140]. In neonatal DMD ΔEx50 mice and young DMD ΔEx50 dogs, the systemic administration of SpCas9 components results in widespread dystrophin restoration throughout body muscles, improves the expression of DGC complex, repairs muscle structure and histology, and enhances muscle strength [138,139,140]. The restoration of dystrophin expression can be up to 92% of the normal level in the heart of systemically injected DMD ΔEx50 mice, in which ~21% of the genome-editing efficiency is yielded durably [138]. Likewise, a single intraperitoneal administration of the SaCas9-KKH system by AAV9 vector in neonatal DMD ΔEx50 mice has been shown to efficiently repair dystrophin expression in skeletal and cardiac muscles, ameliorate muscle structure and membrane integrity, and improve muscle functions such as contractility [133]. Beyond SpCas9 and SaCas9-KKH, both LbCas12a and AsCas12a systems have been shown to rescue dystrophin expression and increase mitochondrial number and oxygen consumption rate in human DMD ΔEx48-50 iPSCs-derived cardiomyocytes [133,142,150]. Recently, single-cut gene editing using the SpCas9 system has been extended to correct other single exonic deletions in human DMD iPSCs and DMD mouse models with ΔEx43, ΔEx44, ΔEx45, or ΔEx52 mutation [44,132,134,136]. The systemic AAV delivery of SpCas9 components in neonatal DMD ΔEx44 mice restores dystrophin expression in nearly all skeletal and cardiac muscles, consequently preventing muscle damage and improving muscle histology and force in the treated mice [44,132,136]. Notably, single-cut editing events in skeletal muscles of 18-month-old DMD ΔEx44 mice are around 15% more than in 1-month-old corrected ΔEx44 skeletal muscles [44], providing evidence for the lifelong benefits of single-cut gene therapy in DMD contexts.
Single-cut gene editing also provides an effective means of treating point mutations, small insertions, and short deletions in the exonic and intronic regions of the dystrophin gene [137,141,142,150]. Intramuscularly delivered CjCas9 and an sgRNA targeting the mutant exon 23 are sufficient to restore dystrophin expression and increase skeletal muscle force in the DMD mice with a 1-bp insertion or 14-bp deletion in exon 23 [137]. Likewise, SpCas9-driven single cutting in mutant intron 13 or 19 rescues dystrophin expression in the skeletal and cardiac muscles of WCMD or LRMD DMD canines bearing a small insertion in the intron 13 or 19, respectively [141].
Though single-cut editing is highly efficient in most DMD cases, its efficiency in genomic editing and dystrophin restoration varies dramatically from mutation to mutation. As an example, SpCas9 and one sgRNA targeting the splice donor site of exon 44 induces ~60% of dystrophin-positive myofibers in skeletal muscles of DMD ΔEx45 mice, but the same SpCas9 system merely restores dystrophin expression in ~36% of myofibers of a DMD ΔEx43 mouse model [134]. Meanwhile, the genome-editing efficiency, dystrophin restoration, and the extent of pathological amelioration depend on the dosage ratio of sgRNA to Cas9 protein [132,136]. Thus, the sgRNA sequence and its ratio to Cas protein must be well designed to achieve high therapeutic efficacy.

7. Exon Knockin

Despite its inefficiency in the post-mitotic cells, HDR-mediated gene editing has the capacity to produce full-length dystrophin protein regardless of DMD mutations. This therapeutic strategy is particularly useful for mutations in the essential N- and C-terminus regions of dystrophin. Thus far, the HDR-based knockin strategy has been exploited to handle either point mutations in DMD animal models or a single exonic deletion in human DMD iPSCs (Figure 1D) [124,128,142,143,144,170]. The intramuscular delivery of AAV6-encoded SpCas9, sgRNA and a donor template sequence in adult mdx4Cv mice rescues dystrophin generation and improves skeletal muscle morphology, but its gene-editing efficiency is only about 0.18% [128]. In mdx mice, the application of 180-nt ssODN, sgRNA, and SpCas9 or LbCas12a by zygote injection could repair dystrophin expression in various muscle tissues at different restoration rates [124,142]. The gene-editing efficiencies of 17% to 41% in SpCas9-corrected mice and 8% to 50% in LbCas12a-treated mice probably come from the relatively high activity of HDR in zygotes [124,142]. Moreover, the SpCas9 system and a donor template have been harnessed to insert the missing human exon 44 back in DMD ΔEx44 iPSCs and generate full-length human dystrophin protein in their derivate cells [161]. Nonetheless, the HDR-based knockin strategy is greatly restrained by its low efficiency, the risk of inverted integration, and the allowable length of donor DNA template in certain delivery vectors, making it problematic for large dystrophin deletion mutations.
The homology-independent targeted integration (HITI), an NHEJ-based knockin approach, has no limitations regarding cell type, cellular proliferation status, and off-target integration [171,172]. This HITI technique uses an exogenous donor template containing the desired DNA sequence, which is flanked by the Cas9 cleavage sites. Once Cas9 protein cleaves both the genomic target sequence and the donor template, NHEJ repair machinery ensures the precise and efficient integration of a donor DNA sequence into the genomic locus. Notably, Cas9 protein repeatedly acts on the cleavage sites in the case of inverted integration until the occurrence of the desired insertion of the donor sequence. Recently, an HITI-mediated exon knockin strategy has been used to insert the missing human exon 52 in the hDMDΔ52/mdx mice (Figure 1E) [145]. The exogenous donor template lacks homology arms but contains either human exon 52 or the superexon encompassing the last 28 exons of the human dystrophin gene. Both intramuscular and systemic injections of the SaCas9 system and exogenous DNA template by AAV vectors have been found to effectively restore full-length dystrophin expression in the skeletal and cardiac muscles of hDMDΔ52/mdx mice. The restoration rate of cardiac dystrophin expression in systemically treated hDMDΔ52/mdx mice is 10% to 50% of the normal levels [145]. HITI-mediated superexon knockin approach has great potential for treating more than 20% of global DMD patients.

8. Base Editing

The base editing system, whose editing events do not rely on DSB generation and endogenous NHEJ machinery, offers a powerful strategy for safely correcting DMD mutations, especially point mutations [116]. Two major categories of DNA base editing tools exist: cytosine base editors (CBEs) catalyzing the C•G to T•A transitions, and adenine base editors (ABEs) converting the A•T to G•C base pairs [116,173]. Mechanistically, base editing-driven mutation corrections are accomplished either by direct base conversions at the mutational position or by exon skipping (Figure 1F). Through inducing G-to-A conversion at the splice site of a mutant or surrounding exon, the CBE fused between nSaCas9 and cytidine deaminase AID is employed to skip exon 50 in the cardiomyocytes from human DMD ΔEx51 iPSCs or trigger exon 4 skipping in the DmdE4* mice harboring a 4-bp deletion within exon 4 [149,162]. The editing efficiency of this CBE in human DMD ΔEx51 iPSCs can be up to ~90% at the genomic level, which brings about nearly complete restorations of dystrophin and β-dystroglycan in the corrected cardiomyocytes [162]. The systemic administration of AAV9-encoded CBE and sgRNA in neonatal DmdE4* mice not only leads to the highly efficient restoration of dystrophin protein in cardiac and skeletal muscles but also prevents the onset of DMD symptoms throughout the mouse lifespan [149]. Because CBE-induced dystrophin restoration in the heart can be durable for at least 12 months, a single AAV9-CBE treatment is sufficient to extend the life span of corrected DmdE4* mice [149]. Since off-target editing events of CBEs have been described at both the genomic and transcriptomic levels [174,175,176], there is an urgent need to examine whether the in vivo application of CBEs may cause unpredictably detrimental outcomes (e.g., oncogenesis) in several DMD animal models.
It has been suggested that ABEs are safer than CBEs when rewriting the genome due to their high specificity and low off-target activity [176,177]. In support of their therapeutic potential, multiple ABE variants have been shown to be effective in DMD mice carrying single exonic deletion or nonsense mutations [146,147,148]. By directly introducing an A-to-G substitution at the point mutation site, the nSpCas9-ABE7.10 system has been found to cause genomic correction at ~3.3% efficiency and restore dystrophin expression in up to 17% of skeletal myofibers in DMD mice with a nonsense mutation in exon 20 [147]. Similarly, the systemic infusion of the nSpCas9-miniABE(GG) system in adult mdx4cv mice leads to restored dystrophin expression, ameliorated muscle pathology, and improved muscle functions in both cardiac and skeletal muscles [148]. Genomic editing efficiency and therapeutic benefits can last for at least 9 months in the ABE-corrected mdx4cv mice [148]. As an instance of ABE-triggered exon skipping, the nSpCas9-ABEmax system efficiently rescues the dystrophin reading frame in both human DMD ΔEx51 iPSCs and neonatal DMD ΔEx51 mice via inducing exon 50 skipping [146]. The local delivery of nSpCas9-ABEmax components into the skeletal muscles of DMD ΔEx51 mice can induce dystrophin restoration in nearly all skeletal myofibers and thereby prevent muscle pathology [146].

9. Prime Editing

Besides inducing the transitions of C•G to T•A and A•T to G•C as observed in BE systems, the versatile prime editing is capable of installing all other base substitutions, small insertions, and/or small deletions at the target locus. The prime editing system needs at least a prime editor fused between a reverse transcriptase and an nCas9 protein, and a prime editing guide RNA (pegRNA) [118,178]. PegRNA comprises a spacer complementary to the target site, an sgRNA scaffold, a primer binding site, and a reverse transcription template encoding the desired genomic sequence. Upon binding to the target site, the primer editor nicks the PAM-contained DNA strand, initiates reverse transcription, and synthesizes a new 3′ DNA flap containing the desired modification [118]. This newly synthesized 3′ DNA flap is eventually incorporated into the genome with the aid of endogenous DNA repair pathways. Since coordinating multiple pegRNA components is an essential prerequisite for precise genomic modification, prime editing is thought to trigger negligible byproduct events around the off-target sites.
The prime editing system has been demonstrated to rescue the dystrophin reading frame and protein expression in both human DMD ΔEx51 iPSCs-derived cardiomyocytes (Figure 1G) and human DMD myoblasts with an Ex6 mutant [146,163]. Prime editing-mediated insertion of two nucleotides within downstream exon 52 is found to reframe the ORF of dystrophin transcripts and generate a functional dystrophin protein reaching up to 39.7% of the normal level in the corrected cardiomyocytes [146]. Moreover, this reframing of exon 52 ameliorates abnormal calcium handling and improves the contractility in prime editing-corrected cardiomyocytes [146]. Given that the ΔEx51 mutation accounts for ~8% of all DMD patients [8], it will be valuable to assess the in vivo efficacy and durability of therapeutic prime editing in DMD animal models.

10. DMD Animal Models

More than 60 different animal models of DMD have been identified or generated in Caenorhabditis elegans, Drosophila, zebrafish, rodents, rabbits, dogs, pigs, and nonhuman primates [47,179,180,181,182,183,184,185,186,187,188,189,190]. These DMD animal models either naturally occur or are genetically engineered for the studies of disease mechanisms and clinical translation, and each animal model has its advantages and limitations. Although they are easily reproductive and relatively inexpensive, DMD rodent models generally exhibit mild clinical features of DMD patients owing to the complementary utrophin upregulation and their robust muscle regeneration capacity [191,192]. For example, the most frequently utilized mdx mice do not have moderate pathological signs until they are 15 months old and show just a 25% reduction in lifespan [179,180,181]. Unlike small rodent models, DMD pigs can develop severe disease phenotypes, but they die prematurely before breeding age [183,184], making them difficult to breed on a large scale. Conversely, DMD dogs, another typical large-animal model, display a 75% shortened lifespan similar to human DMD patients and can be bred relatively easily [185,193]. Meanwhile, canine DMD models closely resemble the disease progression and severity experienced by DMD patients, including limb muscle fibrosis and cardiomyopathy [64,65,194]. It seems that DMD dogs may be more suitable for preclinical translational studies than other large animal models. Nonetheless, their broad application in therapeutic translation is restrained by the heavy economic burden and the long time it takes to breed them in sufficient numbers [195]. Albeit both DMD rabbits and monkeys are already established via direct injection of SpCas9 and sgRNAs [186,187], they have not been applied in therapeutic testing due possibly to genetic mosaicism. It is important to note that a single animal model cannot fully mimic all pathological symptoms of human DMD patients. Given the unique characteristics of each DMD animal model, different animal models are proposed to recapitulate specific stages of human DMD progression [182,195]. Murine models are similar to the neonatal to the 3-year-old stage of DMD patients, the canine models represent the 5- to 10-year-old stage, and the porcine models resemble the later stage with cardiac defects. Therefore, it is necessary to insightfully consider and choose the optimal animal model for assessing therapeutic strategies.

11. Future Challenges and Prospects

11.1. Safety

The AAV-based delivery system is the most widely used vehicle for in vivo gene therapy in preclinical and clinical studies, mainly due to its high transduction efficiency, low immunogenicity, and durable therapeutic benefits [196,197]. Notably, the tissue tropism of multiple AAV serotypes (e.g., AAV6, AAV8, and AAV9) to skeletal and cardiac muscles makes these vectors particularly suitable for DMD gene therapy. Since mammalian muscle accounts for approximate 40% of total body mass [198], achieving durable and efficient genome editing in DMD animals requires high AAV vector doses, normally ranging from 5.5 × 1014 to 1.8 × 1015 vector genomes (vg)/kg [35,37,128,132]. This requirement of a high vector dosage poses formidable challenges for clinical-grade AAV manufacturing and the safety of AAV-based gene therapies. The single intravenous administration of high-dose AAV9 (at least 1.5 × 1014 vg/kg) could trigger notable adverse events in multiple organs, such as liver toxicity and kidney injury in dogs, piglets, and nonhuman primates [199,200]. In some clinical trials involving DMD patients, the systemic delivery of the AAV9 vector carrying the human micro-dystrophin gene at high doses (5 × 1013 to 3 × 1014 vg/kg) is reported to cause serious adverse events, including cardiopulmonary insufficiency and thrombocytopenia [201], which might be responsible for the recently described patient death. Therefore, it is crucial to determine the optimal vector dosage before the clinical application of systemic AAV therapeutics.
Given the packaging limitation of AAV vectors, the dual-AAV vector system is most often utilized for in vivo delivery of CRISPR gene-editing components. One major drawback of this dual-AAV system is the essential high dosage of AAV vectors for efficient gene editing. As an optimization strategy to reduce the viral dose, the self-complementary AAV (scAAV) vector with a double-strand viral genome has been developed to transport the sgRNA expression cassette into the skeletal and cardiac muscles of DMD ΔEx44 mice [136]. Unlike single-stranded AAV (ssAAV) vectors, the scAAV bypasses the rate-limiting second-strand synthesis and is resistant to degradation [202,203]. The dosage of scAAV to achieve an efficient genome modification is at least 20-fold lower than that of ssAAV [136]. Thus, the combination of sgRNA-expressed scAAV and Cas-packaged ssAAV may act as an attractive dual-vector system to ensure durable therapeutic efficacy in DMD individuals. However, there remains a need to further optimize the AAV delivery system and design all-in-one AAV vectors to accelerate the clinical translation of CRISPR gene therapy.

11.2. Immunogenicity

Another major concern of AAV-CRISPR gene therapy is the innate and adaptive immune responses evoked by AAV vectors and Cas proteins. Pre-existing anti-AAV antibodies are found in a large proportion of the human population [204,205], so some DMD patients with abundant AAV-neutralizing antibodies may be ineligible for AAV gene therapy. In this circumstance, either plasmapheresis or immunosuppressant needs to be administrated prior to AAV gene therapy for the reduction of anti-AAV antibody titer or the decline of immune system activity in the host patients [169,206]. Beyond the immunogenicity of AAV vectors, Cas-specific immune responses have been documented in murines, canines, and humans, owing to the bacterial and archaeal sources of Cas proteins [37,141,207,208,209]. For example, SaCas9- or SpCas9-specific antibodies and T cells have been ascertained in around 78% or 58–67% of healthy human populations [208]. On the contrary, the sgRNAs are merely reported to stimulate innate immune responses within human cells in vitro [210,211], but whether they are immunogenic in vivo remains to be determined. It is noteworthy that both cellular and humoral immune responses against AAV vectors and SaCas9 are not observed in neonatal mice after the systemic administration of AAV-SaCas9 components [37], suggesting that host immune responses to AAV vectors and Cas proteins might be avoided by treatment at the juvenile stage. This observation has been largely attributed to the lower essential AAV dosage, the more preserved muscles, and the absence of pre-existing immunity against AAV-CRISPR components at young ages [30,37]. Alternatively, the optimized forms of Cas9 proteins without immunogenic epitopes have been developed [212], and their in vivo application holds great potential to reduce the activity of the host immune system. Regardless, since immunosuppressant corticosteroids are normally used to dampen inflammation in DMD patients, one feasible solution to address the above immunogenicity is administrating corticosteroids with CRISPR gene therapy.

11.3. Off-Target Activity

The potential off-target activity of CRISPR systems poses an obstacle to their clinical application. In this regard, SpCas9 has been found to have a relatively high off-target activity owing to its tolerance of up to five mismatches between the guide sequence and target genome site [213,214,215]. Because most off-target cleavage events are present in highly proliferating cells in culture, the off-target genome editing of CRISPR systems is thought to be very low in animal models, especially in post-mitotic skeletal and cardiac cells [30,66]. Nonetheless, it cannot exclude the possible occurrence of deleterious off-target mutations in a specialized cell in vivo. Thus far, several different approaches have been developed to minimize such off-target mutagenesis. An attractive strategy is to use high-fidelity Cas enzymes (e.g., HypaCas9, evoCas9, SpCas9-HF1, and enAsCas12a-HF1) possessing high on-target specificity but low off-target activity [103,104,105,106,114,216]. It has been shown that Cas12 proteins generally exhibit much lower off-target editing activity than Cas9 variants [92,217], so their application is a relatively safe choice for CRISPR therapeutics. As for the second strategy, the sgRNAs can be elaborately optimized by either truncation or extension in a specific manner [218,219]. The truncated sgRNAs of less than 20 nucleotides in length can decrease genome-wide off-target events by up to five-fold [218]. In the third method, the utilization of muscle-specific promoters to specifically drive CRISPR component expressions in muscles can prevent the appearance of off-target editing events in undesirable tissues such as liver and kidney [128,138,220].

11.4. Durability

It should be noted that the skeletal muscles of both DMD patients and animals maintain high turnover rates throughout their lives [221,222]. As a result, dystrophin-positive myofibers following CRISPR therapeutics may be gradually diluted out of existence by dystrophic myofibers in the long term. Because muscle satellite cells undergoing de novo myogenesis are responsible for muscle regeneration [223,224], the delivery of CRISPR components into satellite cells could theoretically ensure durable therapeutic benefits in dystrophic muscles. Several studies have reported that systemic and intramuscular administrations of AAV9 vectors carrying SpCas9 or SaCas9 system in DMD mouse models induce Cas9 component expression and genomic editing in muscle satellite cells [127,225,226,227]. This achievement of satellite cell gene editing following systemic AAV9-CRISPR therapy can maintain restored dystrophin expression for 18 months in the skeletal muscles of mdx mice [225]. Therefore, the effective genome editing of muscle satellite cells is likely a valuable strategy for yielding lifelong therapeutic benefits, but its efficiency needs to be further improved. In the second strategy, micro-dystrophin is co-delivered with the SaCas9 system to achieve durable dystrophin restoration in skeletal muscles [129]. The systemic administration of the AAV6-encoded micro-dystrophin gene not only stabilizes skeletal myofibers but also halts CRISPR component loss, consequently allowing for persistent gene correction and ensuring lifelong skeletal dystrophin expression in DMD mdx4Cv mice [129]. Considering the broad application of micro-dystrophin gene therapy in clinical trials, AAV-based co-delivery of micro-dystrophin and the CRISPR system shows great promise for the efficient treatment of diverse DMD patients.

12. Conclusions

The development and application of CRISPR-Cas technologies provide new opportunities for treating various genomic mutations at the source, enabling the durable restoration of protein expression and functions in the correct tissues. To date, the lifelong benefits of one-time CRISPR therapeutics have been manifested in some DMD mouse models at the preclinical level [35,37,43,44,148,149]. Although the in vivo therapeutic efficacy of CRISPR systems is promising without doubt, their safety profiles, especially concerning immunogenicity, AAV delivery and off-target issues, are imminently necessary to be addressed in several DMD animal models. As the challenges prior to clinical translation are overcome in the near future, the lessons from CRISPR therapeutics in DMD should apply to other devastating genetic diseases that lack effective therapies.

Author Contributions

Conceptualization, H.L.; Literature, G.C., T.W., G.L., H.L.; Writing—original draft preparation, G.C., H.L.; Visualization, G.C., T.W., H.L.; Writing—review and editing, T.W., H.Y., H.L.; Supervision: H.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the National Natural Science Foundation of China (82273475), the Natural Science Foundation of Shanghai (22ZR1449100), the Strategic Priority Research Program of the Chinese Academy of Science (XDB32060000), and the National Natural Science Foundation of China (31871502, 31925016, 91957122, 82021001).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Zhaoming Chen for his assistance in the literature collection.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mendell, J.R.; Shilling, C.; Leslie, N.D.; Flanigan, K.M.; al-Dahhak, R.; Gastier-Foster, J.; Kneile, K.; Dunn, D.M.; Duval, B.; Aoyagi, A.; et al. Evidence-based path to newborn screening for Duchenne muscular dystrophy. Ann. Neurol. 2012, 71, 304–313. [Google Scholar] [CrossRef] [PubMed]
  2. Muntoni, F.; Torelli, S.; Ferlini, A. Dystrophin and mutations: One gene, several proteins, multiple phenotypes. Lancet Neurol. 2003, 2, 731–740. [Google Scholar] [CrossRef]
  3. Hamosh, A.; Scott, A.F.; Amberger, J.S.; Bocchini, C.A.; McKusick, V.A. Online Mendelian Inheritance in Man (OMIM), a knowledgebase of human genes and genetic disorders. Nucleic Acids Res. 2005, 33, D514–D517. [Google Scholar] [CrossRef] [PubMed]
  4. Constantin, B. Dystrophin complex functions as a scaffold for signalling proteins. Biochim. Biophys. Acta 2014, 1838, 635–642. [Google Scholar] [CrossRef] [PubMed]
  5. Lai, Y.; Zhao, J.; Yue, Y.; Duan, D. α2 and α3 helices of dystrophin R16 and R17 frame a microdomain in the α1 helix of dystrophin R17 for neuronal NOS binding. Proc. Natl. Acad. Sci. USA 2013, 110, 525–530. [Google Scholar] [CrossRef] [PubMed]
  6. Mercuri, E.; Muntoni, F. Muscular dystrophies. Lancet 2013, 381, 845–860. [Google Scholar] [CrossRef]
  7. Bladen, C.L.; Salgado, D.; Monges, S.; Foncuberta, M.E.; Kekou, K.; Kosma, K.; Dawkins, H.; Lamont, L.; Roy, A.J.; Chamova, T.; et al. The TREAT-NMD DMD Global Database: Analysis of more than 7000 Duchenne muscular dystrophy mutations. Hum. Mutat. 2015, 36, 395–402. [Google Scholar] [CrossRef]
  8. Echigoya, Y.; Lim, K.R.Q.; Nakamura, A.; Yokota, T. Multiple Exon Skipping in the Duchenne Muscular Dystrophy Hot Spots: Prospects and Challenges. J. Pers. Med. 2018, 8, 41. [Google Scholar] [CrossRef]
  9. Gloss, D.; Moxley, R.T., 3rd; Ashwal, S.; Oskoui, M. Practice guideline update summary: Corticosteroid treatment of Duchenne muscular dystrophy: Report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology 2016, 86, 465–472. [Google Scholar] [CrossRef]
  10. Birnkrant, D.J.; Bushby, K.; Bann, C.M.; Apkon, S.D.; Blackwell, A.; Colvin, M.K.; Cripe, L.; Herron, A.R.; Kennedy, A.; Kinnett, K.; et al. Diagnosis and management of Duchenne muscular dystrophy, part 3: Primary care, emergency management, psychosocial care, and transitions of care across the lifespan. Lancet Neurol. 2018, 17, 445–455. [Google Scholar] [CrossRef] [Green Version]
  11. Wilton-Clark, H.; Yokota, T. Antisense and Gene Therapy Options for Duchenne Muscular Dystrophy Arising from Mutations in the N-Terminal Hotspot. Genes 2022, 13, 257. [Google Scholar] [CrossRef]
  12. Zhang, T.; Kong, X. Recent advances of glucocorticoids in the treatment of Duchenne muscular dystrophy (Review). Exp. Ther. Med. 2021, 21, 447. [Google Scholar] [CrossRef]
  13. Ciafaloni, E.; Moxley, R.T. Treatment options for Duchenne muscular dystrophy. Curr. Treat. Options Neurol. 2008, 10, 86–93. [Google Scholar] [CrossRef]
  14. Dubinin, M.V.; Talanov, E.Y.; Tenkov, K.S.; Starinets, V.S.; Belosludtseva, N.V.; Belosludtsev, K.N. The Effect of Deflazacort Treatment on the Functioning of Skeletal Muscle Mitochondria in Duchenne Muscular Dystrophy. Int. J. Mol. Sci. 2020, 21, 8763. [Google Scholar] [CrossRef]
  15. Hörster, I.; Weigt-Usinger, K.; Carmann, C.; Chobanyan-Jürgens, K.; Köhler, C.; Schara, U.; Kayacelebi, A.A.; Beckmann, B.; Tsikas, D.; Lücke, T. The L-arginine/NO pathway and homoarginine are altered in Duchenne muscular dystrophy and improved by glucocorticoids. Amino Acids 2015, 47, 1853–1863. [Google Scholar] [CrossRef]
  16. Podkalicka, P.; Mucha, O.; Dulak, J.; Loboda, A. Targeting angiogenesis in Duchenne muscular dystrophy. Cell. Mol. Life Sci. 2019, 76, 1507–1528. [Google Scholar] [CrossRef]
  17. Miyatake, S.; Shimizu-Motohashi, Y.; Takeda, S.; Aoki, Y. Anti-inflammatory drugs for Duchenne muscular dystrophy: Focus on skeletal muscle-releasing factors. Drug Des. Dev. Ther. 2016, 10, 2745–2758. [Google Scholar]
  18. Shimizu-Motohashi, Y.; Miyatake, S.; Komaki, H.; Takeda, S.; Aoki, Y. Recent advances in innovative therapeutic approaches for Duchenne muscular dystrophy: From discovery to clinical trials. Am. J. Transl. Res. 2016, 8, 2471–2489. [Google Scholar]
  19. Goemans, N.; Buyse, G. Current treatment and management of dystrophinopathies. Curr. Treat. Options Neurol. 2014, 16, 287. [Google Scholar] [CrossRef]
  20. Ricotti, V.; Ridout, D.A.; Scott, E.; Quinlivan, R.; Robb, S.A.; Manzur, A.Y.; Muntoni, F.; NorthStar Clinical, N. Long-term benefits and adverse effects of intermittent versus daily glucocorticoids in boys with Duchenne muscular dystrophy. J. Neurol. Neurosurg. Psychiatry 2013, 84, 698–705. [Google Scholar] [CrossRef]
  21. Hammers, D.W.; Hart, C.C.; Patsalos, A.; Matheny, M.K.; Wright, L.A.; Nagy, L.; Sweeney, H.L. Glucocorticoids counteract hypertrophic effects of myostatin inhibition in dystrophic muscle. JCI Insight 2020, 5, e133276. [Google Scholar] [CrossRef]
  22. Schneider, A.E.; Aartsma-Rus, A. Developments in reading frame restoring therapy approaches for Duchenne muscular dystrophy. Expert Opin. Biol. Ther. 2021, 21, 343–359. [Google Scholar] [CrossRef]
  23. Charleston, J.S.; Schnell, F.J.; Dworzak, J.; Donoghue, C.; Lewis, S.; Chen, L.; Young, G.D.; Milici, A.J.; Voss, J.; DeAlwis, U.; et al. Eteplirsen treatment for Duchenne muscular dystrophy: Exon skipping and dystrophin production. Neurology 2018, 90, e2146–e2154. [Google Scholar] [CrossRef]
  24. Frank, D.E.; Schnell, F.J.; Akana, C.; El-Husayni, S.H.; Desjardins, C.A.; Morgan, J.; Charleston, J.S.; Sardone, V.; Domingos, J.; Dickson, G.; et al. Increased dystrophin production with golodirsen in patients with Duchenne muscular dystrophy. Neurology 2020, 94, e2270–e2282. [Google Scholar] [CrossRef]
  25. Clemens, P.R.; Rao, V.K.; Connolly, A.M.; Harper, A.D.; Mah, J.K.; Smith, E.C.; McDonald, C.M.; Zaidman, C.M.; Morgenroth, L.P.; Osaki, H.; et al. Safety, Tolerability, and Efficacy of Viltolarsen in Boys With Duchenne Muscular Dystrophy Amenable to Exon 53 Skipping: A Phase 2 Randomized Clinical Trial. JAMA Neurol. 2020, 77, 982–991. [Google Scholar] [CrossRef]
  26. Wagner, K.R.; Kuntz, N.L.; Koenig, E.; East, L.; Upadhyay, S.; Han, B.; Shieh, P.B. Safety, tolerability, and pharmacokinetics of casimersen in patients with Duchenne muscular dystrophy amenable to exon 45 skipping: A randomized, double-blind, placebo-controlled, dose-titration trial. Muscle Nerve 2021, 64, 285–292. [Google Scholar] [CrossRef]
  27. Lehto, T.; Castillo Alvarez, A.; Gauck, S.; Gait, M.J.; Coursindel, T.; Wood, M.J.; Lebleu, B.; Boisguerin, P. Cellular trafficking determines the exon skipping activity of Pip6a-PMO in mdx skeletal and cardiac muscle cells. Nucleic Acids Res. 2014, 42, 3207–3217. [Google Scholar] [CrossRef]
  28. Lim, K.R.; Maruyama, R.; Yokota, T. Eteplirsen in the treatment of Duchenne muscular dystrophy. Drug Des. Dev. Ther. 2017, 11, 533–545. [Google Scholar] [CrossRef]
  29. Nelson, C.E.; Robinson-Hamm, J.N.; Gersbach, C.A. Genome engineering: A new approach to gene therapy for neuromuscular disorders. Nat. Rev. Neurol. 2017, 13, 647–661. [Google Scholar] [CrossRef] [PubMed]
  30. Olson, E.N. Toward the correction of muscular dystrophy by gene editing. Proc. Natl. Acad. Sci. USA 2021, 118, e2004840117. [Google Scholar] [CrossRef] [PubMed]
  31. Doudna, J.A. The promise and challenge of therapeutic genome editing. Nature 2020, 578, 229–236. [Google Scholar] [CrossRef] [PubMed]
  32. Anzalone, A.V.; Koblan, L.W.; Liu, D.R. Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nat. Biotechnol. 2020, 38, 824–844. [Google Scholar] [CrossRef] [PubMed]
  33. Yeh, C.D.; Richardson, C.D.; Corn, J.E. Advances in genome editing through control of DNA repair pathways. Nat. Cell Biol. 2019, 21, 1468–1478. [Google Scholar] [CrossRef] [PubMed]
  34. Long, C.; Amoasii, L.; Mireault, A.A.; McAnally, J.R.; Li, H.; Sanchez-Ortiz, E.; Bhattacharyya, S.; Shelton, J.M.; Bassel-Duby, R.; Olson, E.N. Postnatal genome editing partially restores dystrophin expression in a mouse model of muscular dystrophy. Science 2016, 351, 400–403. [Google Scholar] [CrossRef] [PubMed]
  35. Hakim, C.H.; Wasala, N.B.; Nelson, C.E.; Wasala, L.P.; Yue, Y.; Louderman, J.A.; Lessa, T.B.; Dai, A.; Zhang, K.; Jenkins, G.J.; et al. AAV CRISPR editing rescues cardiac and muscle function for 18 months in dystrophic mice. JCI Insight 2018, 3, e124297. [Google Scholar] [CrossRef]
  36. Nelson, C.E.; Hakim, C.H.; Ousterout, D.G.; Thakore, P.I.; Moreb, E.A.; Castellanos Rivera, R.M.; Madhavan, S.; Pan, X.; Ran, F.A.; Yan, W.X.; et al. In vivo genome editing improves muscle function in a mouse model of Duchenne muscular dystrophy. Science 2016, 351, 403–407. [Google Scholar] [CrossRef]
  37. Nelson, C.E.; Wu, Y.; Gemberling, M.P.; Oliver, M.L.; Waller, M.A.; Bohning, J.D.; Robinson-Hamm, J.N.; Bulaklak, K.; Castellanos Rivera, R.M.; Collier, J.H.; et al. Long-term evaluation of AAV-CRISPR genome editing for Duchenne muscular dystrophy. Nat. Med. 2019, 25, 427–432. [Google Scholar] [CrossRef]
  38. Young, C.S.; Hicks, M.R.; Ermolova, N.V.; Nakano, H.; Jan, M.; Younesi, S.; Karumbayaram, S.; Kumagai-Cresse, C.; Wang, D.; Zack, J.A.; et al. A Single CRISPR-Cas9 Deletion Strategy that Targets the Majority of DMD Patients Restores Dystrophin Function in hiPSC-Derived Muscle Cells. Cell Stem Cell 2016, 18, 533–540. [Google Scholar] [CrossRef]
  39. Moretti, A.; Fonteyne, L.; Giesert, F.; Hoppmann, P.; Meier, A.B.; Bozoglu, T.; Baehr, A.; Schneider, C.M.; Sinnecker, D.; Klett, K.; et al. Somatic gene editing ameliorates skeletal and cardiac muscle failure in pig and human models of Duchenne muscular dystrophy. Nat. Med. 2020, 26, 207–214. [Google Scholar] [CrossRef]
  40. Duchêne, B.L.; Cherif, K.; Iyombe-Engembe, J.P.; Guyon, A.; Rousseau, J.; Ouellet, D.L.; Barbeau, X.; Lague, P.; Tremblay, J.P. CRISPR-Induced Deletion with SaCas9 Restores Dystrophin Expression in Dystrophic Models In Vitro and In Vivo. Mol. Ther. 2018, 26, 2604–2616. [Google Scholar] [CrossRef]
  41. Chemello, F.; Bassel-Duby, R.; Olson, E.N. Correction of muscular dystrophies by CRISPR gene editing. J. Clin. Investig. 2020, 130, 2766–2776. [Google Scholar] [CrossRef]
  42. Choi, E.; Koo, T. CRISPR technologies for the treatment of Duchenne muscular dystrophy. Mol. Ther. 2021, 29, 3179–3191. [Google Scholar] [CrossRef]
  43. Xu, L.; Lau, Y.S.; Gao, Y.; Li, H.; Han, R. Life-Long AAV-Mediated CRISPR Genome Editing in Dystrophic Heart Improves Cardiomyopathy without Causing Serious Lesions in mdx Mice. Mol. Ther. 2019, 27, 1407–1414. [Google Scholar] [CrossRef]
  44. Karri, D.R.; Zhang, Y.; Chemello, F.; Min, Y.L.; Huang, J.; Kim, J.; Mammen, P.P.A.; Xu, L.; Liu, N.; Bassel-Duby, R.; et al. Long-term maintenance of dystrophin expression and resistance to injury of skeletal muscle in gene edited DMD mice. Mol. Ther. Nucleic Acids 2022, 28, 154–167. [Google Scholar] [CrossRef]
  45. Bergmann, O.; Zdunek, S.; Felker, A.; Salehpour, M.; Alkass, K.; Bernard, S.; Sjostrom, S.L.; Szewczykowska, M.; Jackowska, T.; Dos Remedios, C.; et al. Dynamics of Cell Generation and Turnover in the Human Heart. Cell 2015, 161, 1566–1575. [Google Scholar] [CrossRef]
  46. Takeda, S.; Clemens, P.R.; Hoffman, E.P. Exon-Skipping in Duchenne Muscular Dystrophy. J. Neuromuscul. Dis. 2021, 8, S343–S358. [Google Scholar] [CrossRef]
  47. Wasala, N.B.; Chen, S.J.; Duan, D. Duchenne muscular dystrophy animal models for high-throughput drug discovery and precision medicine. Expert Opin. Drug Discov. 2020, 15, 443–456. [Google Scholar] [CrossRef]
  48. Lionarons, J.M.; Hoogland, G.; Hendriksen, R.G.F.; Faber, C.G.; Hellebrekers, D.M.J.; Van Koeveringe, G.A.; Schipper, S.; Vles, J.S.H. Dystrophin is expressed in smooth muscle and afferent nerve fibers in the rat urinary bladder. Muscle Nerve 2019, 60, 202–210. [Google Scholar] [CrossRef]
  49. Elangkovan, N.; Dickson, G. Gene Therapy for Duchenne Muscular Dystrophy. J. Neuromuscul. Dis. 2021, 8, S303–S316. [Google Scholar] [CrossRef]
  50. Murphy, S.; Zweyer, M.; Henry, M.; Meleady, P.; Mundegar, R.R.; Swandulla, D.; Ohlendieck, K. Proteomic analysis of the sarcolemma-enriched fraction from dystrophic mdx-4cv skeletal muscle. J. Proteom. 2019, 191, 212–227. [Google Scholar] [CrossRef]
  51. Zabłocka, B.; Górecki, D.C.; Zabłocki, K. Disrupted Calcium Homeostasis in Duchenne Muscular Dystrophy: A Common Mechanism behind Diverse Consequences. Int. J. Mol. Sci. 2021, 22, 11040. [Google Scholar] [CrossRef] [PubMed]
  52. Fong, P.Y.; Turner, P.R.; Denetclaw, W.F.; Steinhardt, R.A. Increased activity of calcium leak channels in myotubes of Duchenne human and mdx mouse origin. Science 1990, 250, 673–676. [Google Scholar] [CrossRef] [PubMed]
  53. Turner, P.R.; Fong, P.Y.; Denetclaw, W.F.; Steinhardt, R.A. Increased calcium influx in dystrophic muscle. J. Cell Biol. 1991, 115, 1701–1712. [Google Scholar] [CrossRef] [PubMed]
  54. Rando, T.A.; Disatnik, M.H.; Yu, Y.; Franco, A. Muscle cells from mdx mice have an increased susceptibility to oxidative stress. Neuromuscul. Disord. 1998, 8, 14–21. [Google Scholar] [CrossRef]
  55. Kuznetsov, A.V.; Winkler, K.; Wiedemann, F.R.; von Bossanyi, P.; Dietzmann, K.; Kunz, W.S. Impaired mitochondrial oxidative phosphorylation in skeletal muscle of the dystrophin-deficient mdx mouse. Mol. Cell Biochem. 1998, 183, 87–96. [Google Scholar] [CrossRef]
  56. Timpani, C.A.; Hayes, A.; Rybalka, E. Revisiting the dystrophin-ATP connection: How half a century of research still implicates mitochondrial dysfunction in Duchenne Muscular Dystrophy aetiology. Med. Hypotheses 2015, 85, 1021–1033. [Google Scholar] [CrossRef]
  57. Even, P.C.; Decrouy, A.; Chinet, A. Defective regulation of energy metabolism in mdx-mouse skeletal muscles. Biochem. J. 1994, 304 Pt 2, 649–654. [Google Scholar] [CrossRef]
  58. De Palma, C.; Morisi, F.; Cheli, S.; Pambianco, S.; Cappello, V.; Vezzoli, M.; Rovere-Querini, P.; Moggio, M.; Ripolone, M.; Francolini, M.; et al. Autophagy as a new therapeutic target in Duchenne muscular dystrophy. Cell Death Dis. 2012, 3, e418. [Google Scholar] [CrossRef]
  59. Brenman, J.E.; Chao, D.S.; Xia, H.; Aldape, K.; Bredt, D.S. Nitric oxide synthase complexed with dystrophin and absent from skeletal muscle sarcolemma in Duchenne muscular dystrophy. Cell 1995, 82, 743–752. [Google Scholar] [CrossRef]
  60. Smith, M.A.; Schnellmann, R.G. Calpains, mitochondria, and apoptosis. Cardiovasc. Res. 2012, 96, 32–37. [Google Scholar] [CrossRef]
  61. Chen, Y.W.; Nagaraju, K.; Bakay, M.; McIntyre, O.; Rawat, R.; Shi, R.; Hoffman, E.P. Early onset of inflammation and later involvement of TGFbeta in Duchenne muscular dystrophy. Neurology 2005, 65, 826–834. [Google Scholar] [CrossRef]
  62. Szigyarto, C.A.; Spitali, P. Biomarkers of Duchenne muscular dystrophy: Current findings. Degener. Neurol. Neuromuscul. Dis. 2018, 8, 1–13. [Google Scholar]
  63. Yucel, N.; Chang, A.C.; Day, J.W.; Rosenthal, N.; Blau, H.M. Humanizing the mdx mouse model of DMD: The long and the short of it. NPJ Regen. Med. 2018, 3, 4. [Google Scholar] [CrossRef] [Green Version]
  64. Kornegay, J.N. The golden retriever model of Duchenne muscular dystrophy. Skelet Muscle 2017, 7, 9. [Google Scholar] [CrossRef]
  65. Brinkmeyer-Langford, C.; Chu, C.; Balog-Alvarez, C.; Yu, X.; Cai, J.J.; Nabity, M.; Kornegay, J.N. Expression profiling of disease progression in canine model of Duchenne muscular dystrophy. PLoS ONE 2018, 13, e0194485. [Google Scholar] [CrossRef]
  66. Min, Y.L.; Bassel-Duby, R.; Olson, E.N. CRISPR Correction of Duchenne Muscular Dystrophy. Annu Rev Med 2019, 70, 239–255. [Google Scholar] [CrossRef]
  67. Monaco, A.P.; Bertelson, C.J.; Liechti-Gallati, S.; Moser, H.; Kunkel, L.M. An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics 1988, 2, 90–95. [Google Scholar] [CrossRef]
  68. Andrews, J.G.; Wahl, R.A. Duchenne and Becker muscular dystrophy in adolescents: Current perspectives. Adolesc. Health Med. Ther 2018, 9, 53–63. [Google Scholar] [CrossRef]
  69. Flanigan, K.M. Duchenne and Becker muscular dystrophies. Neurol. Clin. 2014, 32, 671–688. [Google Scholar] [CrossRef]
  70. Ferreiro, V.; Giliberto, F.; Muñiz, G.M.; Francipane, L.; Marzese, D.M.; Mampel, A.; Roqué, M.; Frechtel, G.D.; Szijan, I. Asymptomatic Becker muscular dystrophy in a family with a multiexon deletion. Muscle Nerve 2009, 39, 239–243. [Google Scholar] [CrossRef]
  71. Wang, B.; Li, J.; Fu, F.H.; Xiao, X. Systemic human minidystrophin gene transfer improves functions and life span of dystrophin and dystrophin/utrophin-deficient mice. J. Orthop. Res. 2009, 27, 421–426. [Google Scholar] [CrossRef]
  72. Le Guiner, C.; Servais, L.; Montus, M.; Larcher, T.; Fraysse, B.; Moullec, S.; Allais, M.; François, V.; Dutilleul, M.; Malerba, A.; et al. Long-term microdystrophin gene therapy is effective in a canine model of Duchenne muscular dystrophy. Nat. Commun. 2017, 8, 16105. [Google Scholar] [CrossRef]
  73. Koppanati, B.M.; Li, J.; Reay, D.P.; Wang, B.; Daood, M.; Zheng, H.; Xiao, X.; Watchko, J.F.; Clemens, P.R. Improvement of the mdx mouse dystrophic phenotype by systemic in utero AAV8 delivery of a minidystrophin gene. Gene Ther. 2010, 17, 1355–1362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Wasala, N.B.; Shin, J.H.; Lai, Y.; Yue, Y.; Montanaro, F.; Duan, D. Cardiac-Specific Expression of ΔH2-R15 Mini-Dystrophin Normalized All Electrocardiogram Abnormalities and the End-Diastolic Volume in a 23-Month-Old Mouse Model of Duchenne Dilated Cardiomyopathy. Hum. Gene Ther. 2018, 29, 737–748. [Google Scholar] [CrossRef] [PubMed]
  75. Bowles, D.E.; McPhee, S.W.; Li, C.; Gray, S.J.; Samulski, J.J.; Camp, A.S.; Li, J.; Wang, B.; Monahan, P.E.; Rabinowitz, J.E.; et al. Phase 1 gene therapy for Duchenne muscular dystrophy using a translational optimized AAV vector. Mol. Ther. 2012, 20, 443–455. [Google Scholar] [CrossRef] [PubMed]
  76. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef] [PubMed]
  77. Mali, P.; Yang, L.; Esvelt, K.M.; Aach, J.; Guell, M.; DiCarlo, J.E.; Norville, J.E.; Church, G.M. RNA-guided human genome engineering via Cas9. Science 2013, 339, 823–826. [Google Scholar] [CrossRef] [PubMed]
  78. Cong, L.; Ran, F.A.; Cox, D.; Lin, S.; Barretto, R.; Habib, N.; Hsu, P.D.; Wu, X.; Jiang, W.; Marraffini, L.A.; et al. Multiplex genome engineering using CRISPR/Cas systems. Science 2013, 339, 819–823. [Google Scholar] [CrossRef]
  79. Makarova, K.S.; Wolf, Y.I.; Iranzo, J.; Shmakov, S.A.; Alkhnbashi, O.S.; Brouns, S.J.J.; Charpentier, E.; Cheng, D.; Haft, D.H.; Horvath, P.; et al. Evolutionary classification of CRISPR-Cas systems: A burst of class 2 and derived variants. Nat. Rev. Microbiol. 2020, 18, 67–83. [Google Scholar] [CrossRef]
  80. Gao, Y.; Gao, K.; Yang, H. CRISPR/Cas: A potential gene-editing tool in the nervous system. Cell Regen. 2020, 9, 12. [Google Scholar] [CrossRef]
  81. Zetsche, B.; Gootenberg, J.S.; Abudayyeh, O.O.; Slaymaker, I.M.; Makarova, K.S.; Essletzbichler, P.; Volz, S.E.; Joung, J.; van der Oost, J.; Regev, A.; et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell 2015, 163, 759–771. [Google Scholar] [CrossRef]
  82. Zuo, Z.; Liu, J. Cas9-catalyzed DNA Cleavage Generates Staggered Ends: Evidence from Molecular Dynamics Simulations. Sci. Rep. 2016, 5, 37584. [Google Scholar] [CrossRef]
  83. Lemos, B.R.; Kaplan, A.C.; Bae, J.E.; Ferrazzoli, A.E.; Kuo, J.; Anand, R.P.; Waterman, D.P.; Haber, J.E. CRISPR/Cas9 cleavages in budding yeast reveal templated insertions and strand-specific insertion/deletion profiles. Proc. Natl. Acad. Sci. USA 2018, 115, E2040–E2047. [Google Scholar] [CrossRef] [Green Version]
  84. Kakarougkas, A.; Jeggo, P.A. DNA DSB repair pathway choice: An orchestrated handover mechanism. Br. J. Radiol. 2014, 87, 20130685. [Google Scholar] [CrossRef]
  85. Moynahan, M.E.; Jasin, M. Mitotic homologous recombination maintains genomic stability and suppresses tumorigenesis. Nat. Rev. Mol. Cell Biol. 2010, 11, 196–207. [Google Scholar] [CrossRef]
  86. Lieber, M.R. The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annu. Rev. Biochem. 2010, 79, 181–211. [Google Scholar] [CrossRef]
  87. Sternberg, S.H.; Redding, S.; Jinek, M.; Greene, E.C.; Doudna, J.A. DNA interrogation by the CRISPR RNA-guided endonuclease Cas9. Nature 2014, 507, 62–67. [Google Scholar] [CrossRef]
  88. Xie, H.; Tang, L.; He, X.; Liu, X.; Zhou, C.; Liu, J.; Ge, X.; Li, J.; Liu, C.; Zhao, J.; et al. SaCas9 Requires 5′-NNGRRT-3′ PAM for Sufficient Cleavage and Possesses Higher Cleavage Activity than SpCas9 or FnCpf1 in Human Cells. Biotechnol. J. 2018, 13, e1700561. [Google Scholar] [CrossRef]
  89. Kim, E.; Koo, T.; Park, S.W.; Kim, D.; Kim, K.; Cho, H.Y.; Song, D.W.; Lee, K.J.; Jung, M.H.; Kim, S.; et al. In vivo genome editing with a small Cas9 orthologue derived from Campylobacter jejuni. Nat. Commun. 2017, 8, 14500. [Google Scholar]
  90. Wu, Z.; Yang, H.; Colosi, P. Effect of genome size on AAV vector packaging. Mol. Ther. 2010, 18, 80–86. [Google Scholar]
  91. Tran, M.H.; Park, H.; Nobles, C.L.; Karunadharma, P.; Pan, L.; Zhong, G.; Wang, H.; He, W.; Ou, T.; Crynen, G.; et al. A more efficient CRISPR-Cas12a variant derived from Lachnospiraceae bacterium MA2020. Mol. Ther. Nucleic Acids 2021, 24, 40–53. [Google Scholar] [CrossRef]
  92. Kim, D.; Kim, J.; Hur, J.K.; Been, K.W.; Yoon, S.H.; Kim, J.S. Genome-wide analysis reveals specificities of Cpf1 endonucleases in human cells. Nat. Biotechnol. 2016, 34, 863–868. [Google Scholar] [CrossRef]
  93. Esvelt, K.M.; Mali, P.; Braff, J.L.; Moosburner, M.; Yaung, S.J.; Church, G.M. Orthogonal Cas9 proteins for RNA-guided gene regulation and editing. Nat. Methods 2013, 10, 1116–1121. [Google Scholar] [CrossRef] [PubMed]
  94. Müller, M.; Lee, C.M.; Gasiunas, G.; Davis, T.H.; Cradick, T.J.; Siksnys, V.; Bao, G.; Cathomen, T.; Mussolino, C. Streptococcus thermophilus CRISPR-Cas9 Systems Enable Specific Editing of the Human Genome. Mol. Ther. 2016, 24, 636–644. [Google Scholar] [CrossRef] [PubMed]
  95. Hou, Z.; Zhang, Y.; Propson, N.E.; Howden, S.E.; Chu, L.F.; Sontheimer, E.J.; Thomson, J.A. Efficient genome engineering in human pluripotent stem cells using Cas9 from Neisseria meningitidis. Proc. Natl. Acad. Sci. USA 2013, 110, 15644–15649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Edraki, A.; Mir, A.; Ibraheim, R.; Gainetdinov, I.; Yoon, Y.; Song, C.Q.; Cao, Y.; Gallant, J.; Xue, W.; Rivera-Pérez, J.A.; et al. A Compact, High-Accuracy Cas9 with a Dinucleotide PAM for In Vivo Genome Editing. Mol. Cell 2019, 73, 714–726. [Google Scholar] [CrossRef]
  97. Shmakov, S.; Abudayyeh, O.O.; Makarova, K.S.; Wolf, Y.I.; Gootenberg, J.S.; Semenova, E.; Minakhin, L.; Joung, J.; Konermann, S.; Severinov, K.; et al. Discovery and Functional Characterization of Diverse Class 2 CRISPR-Cas Systems. Mol. Cell 2015, 60, 385–397. [Google Scholar] [CrossRef]
  98. Yan, W.X.; Hunnewell, P.; Alfonse, L.E.; Carte, J.M.; Keston-Smith, E.; Sothiselvam, S.; Garrity, A.J.; Chong, S.; Makarova, K.S.; Koonin, E.V.; et al. Functionally diverse type V CRISPR-Cas systems. Science 2019, 363, 88–91. [Google Scholar] [CrossRef]
  99. Strecker, J.; Jones, S.; Koopal, B.; Schmid-Burgk, J.; Zetsche, B.; Gao, L.; Makarova, K.S.; Koonin, E.V.; Zhang, F. Engineering of CRISPR-Cas12b for human genome editing. Nat. Commun. 2019, 10, 212. [Google Scholar] [CrossRef]
  100. Liu, J.J.; Orlova, N.; Oakes, B.L.; Ma, E.; Spinner, H.B.; Baney, K.L.M.; Chuck, J.; Tan, D.; Knott, G.J.; Harrington, L.B.; et al. CasX enzymes comprise a distinct family of RNA-guided genome editors. Nature 2019, 566, 218–223. [Google Scholar] [CrossRef]
  101. Hirano, H.; Gootenberg, J.S.; Horii, T.; Abudayyeh, O.O.; Kimura, M.; Hsu, P.D.; Nakane, T.; Ishitani, R.; Hatada, I.; Zhang, F.; et al. Structure and Engineering of Francisella novicida Cas9. Cell 2016, 164, 950–961. [Google Scholar] [CrossRef]
  102. Kleinstiver, B.P.; Prew, M.S.; Tsai, S.Q.; Topkar, V.V.; Nguyen, N.T.; Zheng, Z.; Gonzales, A.P.; Li, Z.; Peterson, R.T.; Yeh, J.R.; et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature 2015, 523, 481–485. [Google Scholar] [CrossRef]
  103. Slaymaker, I.M.; Gao, L.; Zetsche, B.; Scott, D.A.; Yan, W.X.; Zhang, F. Rationally engineered Cas9 nucleases with improved specificity. Science 2016, 351, 84–88. [Google Scholar] [CrossRef]
  104. Chen, J.S.; Dagdas, Y.S.; Kleinstiver, B.P.; Welch, M.M.; Sousa, A.A.; Harrington, L.B.; Sternberg, S.H.; Joung, J.K.; Yildiz, A.; Doudna, J.A. Enhanced proofreading governs CRISPR-Cas9 targeting accuracy. Nature 2017, 550, 407–410. [Google Scholar] [CrossRef]
  105. Kulcsár, P.I.; Tálas, A.; Huszár, K.; Ligeti, Z.; Tóth, E.; Weinhardt, N.; Fodor, E.; Welker, E. Crossing enhanced and high fidelity SpCas9 nucleases to optimize specificity and cleavage. Genome Biol. 2017, 18, 190. [Google Scholar] [CrossRef] [Green Version]
  106. Casini, A.; Olivieri, M.; Petris, G.; Montagna, C.; Reginato, G.; Maule, G.; Lorenzin, F.; Prandi, D.; Romanel, A.; Demichelis, F.; et al. A highly specific SpCas9 variant is identified by in vivo screening in yeast. Nat. Biotechnol. 2018, 36, 265–271. [Google Scholar] [CrossRef]
  107. Hu, J.H.; Miller, S.M.; Geurts, M.H.; Tang, W.; Chen, L.; Sun, N.; Zeina, C.M.; Gao, X.; Rees, H.A.; Lin, Z.; et al. Evolved Cas9 variants with broad PAM compatibility and high DNA specificity. Nature 2018, 556, 57–63. [Google Scholar] [CrossRef]
  108. Vakulskas, C.A.; Dever, D.P.; Rettig, G.R.; Turk, R.; Jacobi, A.M.; Collingwood, M.A.; Bode, N.M.; McNeill, M.S.; Yan, S.; Camarena, J.; et al. A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human hematopoietic stem and progenitor cells. Nat. Med. 2018, 24, 1216–1224. [Google Scholar] [CrossRef]
  109. Lee, J.K.; Jeong, E.; Lee, J.; Jung, M.; Shin, E.; Kim, Y.H.; Lee, K.; Jung, I.; Kim, D.; Kim, S.; et al. Directed evolution of CRISPR-Cas9 to increase its specificity. Nat. Commun. 2018, 9, 3048. [Google Scholar] [CrossRef]
  110. Nishimasu, H.; Shi, X.; Ishiguro, S.; Gao, L.; Hirano, S.; Okazaki, S.; Noda, T.; Abudayyeh, O.O.; Gootenberg, J.S.; Mori, H.; et al. Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science 2018, 361, 1259–1262. [Google Scholar] [CrossRef]
  111. Chatterjee, P.; Lee, J.; Nip, L.; Koseki, S.R.T.; Tysinger, E.; Sontheimer, E.J.; Jacobson, J.M.; Jakimo, N. A Cas9 with PAM recognition for adenine dinucleotides. Nat. Commun. 2020, 11, 2474. [Google Scholar] [CrossRef]
  112. Kleinstiver, B.P.; Prew, M.S.; Tsai, S.Q.; Nguyen, N.T.; Topkar, V.V.; Zheng, Z.; Joung, J.K. Broadening the targeting range of Staphylococcus aureus CRISPR-Cas9 by modifying PAM recognition. Nat. Biotechnol. 2015, 33, 1293–1298. [Google Scholar] [CrossRef]
  113. Gao, L.; Cox, D.B.T.; Yan, W.X.; Manteiga, J.C.; Schneider, M.W.; Yamano, T.; Nishimasu, H.; Nureki, O.; Crosetto, N.; Zhang, F. Engineered Cpf1 variants with altered PAM specificities. Nat. Biotechnol. 2017, 35, 789–792. [Google Scholar] [CrossRef]
  114. Kleinstiver, B.P.; Sousa, A.A.; Walton, R.T.; Tak, Y.E.; Hsu, J.Y.; Clement, K.; Welch, M.M.; Horng, J.E.; Malagon-Lopez, J.; Scarfò, I.; et al. Engineered CRISPR-Cas12a variants with increased activities and improved targeting ranges for gene, epigenetic and base editing. Nat. Biotechnol. 2019, 37, 276–282. [Google Scholar] [CrossRef]
  115. Pickar-Oliver, A.; Gersbach, C.A. The next generation of CRISPR-Cas technologies and applications. Nat. Rev. Mol. Cell Biol. 2019, 20, 490–507. [Google Scholar] [CrossRef]
  116. Porto, E.M.; Komor, A.C.; Slaymaker, I.M.; Yeo, G.W. Base editing: Advances and therapeutic opportunities. Nat. Rev. Drug Discov. 2020, 19, 839–859. [Google Scholar] [CrossRef]
  117. Rees, H.A.; Liu, D.R. Base editing: Precision chemistry on the genome and transcriptome of living cells. Nat. Rev. Genet. 2018, 19, 770–788. [Google Scholar] [CrossRef] [PubMed]
  118. Anzalone, A.V.; Randolph, P.B.; Davis, J.R.; Sousa, A.A.; Koblan, L.W.; Levy, J.M.; Chen, P.J.; Wilson, C.; Newby, G.A.; Raguram, A.; et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019, 576, 149–157. [Google Scholar] [CrossRef]
  119. Komor, A.C.; Kim, Y.B.; Packer, M.S.; Zuris, J.A.; Liu, D.R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 2016, 533, 420–424. [Google Scholar] [CrossRef]
  120. Gaudelli, N.M.; Komor, A.C.; Rees, H.A.; Packer, M.S.; Badran, A.H.; Bryson, D.I.; Liu, D.R. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 2017, 551, 464–471. [Google Scholar] [CrossRef] [PubMed]
  121. Nelson, J.W.; Randolph, P.B.; Shen, S.P.; Everette, K.A.; Chen, P.J.; Anzalone, A.V.; An, M.; Newby, G.A.; Chen, J.C.; Hsu, A.; et al. Engineered pegRNAs improve prime editing efficiency. Nat. Biotechnol. 2022, 40, 402–410. [Google Scholar] [CrossRef] [PubMed]
  122. Wojtal, D.; Kemaladewi, D.U.; Malam, Z.; Abdullah, S.; Wong, T.W.; Hyatt, E.; Baghestani, Z.; Pereira, S.; Stavropoulos, J.; Mouly, V.; et al. Spell Checking Nature: Versatility of CRISPR/Cas9 for Developing Treatments for Inherited Disorders. Am. J. Hum. Genet. 2016, 98, 90–101. [Google Scholar] [CrossRef] [PubMed]
  123. Cheng, A.W.; Wang, H.; Yang, H.; Shi, L.; Katz, Y.; Theunissen, T.W.; Rangarajan, S.; Shivalila, C.S.; Dadon, D.B.; Jaenisch, R. Multiplexed activation of endogenous genes by CRISPR-on, an RNA-guided transcriptional activator system. Cell Res. 2013, 23, 1163–1171. [Google Scholar] [CrossRef]
  124. Long, C.; McAnally, J.R.; Shelton, J.M.; Mireault, A.A.; Bassel-Duby, R.; Olson, E.N. Prevention of muscular dystrophy in mice by CRISPR/Cas9-mediated editing of germline DNA. Science 2014, 345, 1184–1188. [Google Scholar] [CrossRef]
  125. Maino, E.; Wojtal, D.; Evagelou, S.L.; Farheen, A.; Wong, T.W.Y.; Lindsay, K.; Scott, O.; Rizvi, S.Z.; Hyatt, E.; Rok, M.; et al. Targeted genome editing in vivo corrects a Dmd duplication restoring wild-type dystrophin expression. EMBO Mol. Med. 2021, 13, e13228. [Google Scholar] [CrossRef]
  126. Xu, L.; Park, K.H.; Zhao, L.; Xu, J.; El Refaey, M.; Gao, Y.; Zhu, H.; Ma, J.; Han, R. CRISPR-mediated Genome Editing Restores Dystrophin Expression and Function in mdx Mice. Mol. Ther. 2016, 24, 564–569. [Google Scholar] [CrossRef]
  127. Tabebordbar, M.; Zhu, K.; Cheng, J.K.W.; Chew, W.L.; Widrick, J.J.; Yan, W.X.; Maesner, C.; Wu, E.Y.; Xiao, R.; Ran, F.A.; et al. In vivo gene editing in dystrophic mouse muscle and muscle stem cells. Science 2016, 351, 407–411. [Google Scholar] [CrossRef]
  128. Bengtsson, N.E.; Hall, J.K.; Odom, G.L.; Phelps, M.P.; Andrus, C.R.; Hawkins, R.D.; Hauschka, S.D.; Chamberlain, J.R.; Chamberlain, J.S. Muscle-specific CRISPR/Cas9 dystrophin gene editing ameliorates pathophysiology in a mouse model for Duchenne muscular dystrophy. Nat. Commun. 2017, 8, 14454. [Google Scholar] [CrossRef]
  129. Bengtsson, N.E.; Tasfaout, H.; Hauschka, S.D.; Chamberlain, J.S. Dystrophin Gene-Editing Stability Is Dependent on Dystrophin Levels in Skeletal but Not Cardiac Muscles. Mol. Ther. 2021, 29, 1070–1085. [Google Scholar] [CrossRef]
  130. Young, C.S.; Mokhonova, E.; Quinonez, M.; Pyle, A.D.; Spencer, M.J. Creation of a Novel Humanized Dystrophic Mouse Model of Duchenne Muscular Dystrophy and Application of a CRISPR/Cas9 Gene Editing Therapy. J. Neuromuscul. Dis. 2017, 4, 139–145. [Google Scholar] [CrossRef]
  131. El Refaey, M.; Xu, L.; Gao, Y.; Canan, B.D.; Adesanya, T.M.A.; Warner, S.C.; Akagi, K.; Symer, D.E.; Mohler, P.J.; Ma, J.; et al. In Vivo Genome Editing Restores Dystrophin Expression and Cardiac Function in Dystrophic Mice. Circ. Res. 2017, 121, 923–929. [Google Scholar] [CrossRef] [PubMed]
  132. Min, Y.L.; Li, H.; Rodriguez-Caycedo, C.; Mireault, A.A.; Huang, J.; Shelton, J.M.; McAnally, J.R.; Amoasii, L.; Mammen, P.P.A.; Bassel-Duby, R.; et al. CRISPR-Cas9 corrects Duchenne muscular dystrophy exon 44 deletion mutations in mice and human cells. Sci. Adv. 2019, 5, eaav4324. [Google Scholar] [CrossRef] [PubMed]
  133. Zhang, Y.; Nishiyama, T.; Li, H.; Huang, J.; Atmanli, A.; Sanchez-Ortiz, E.; Wang, Z.; Mireault, A.A.; Mammen, P.P.A.; Bassel-Duby, R.; et al. A consolidated AAV system for single-cut CRISPR correction of a common Duchenne muscular dystrophy mutation. Mol. Ther. Methods Clin. Dev. 2021, 22, 122–132. [Google Scholar] [CrossRef] [PubMed]
  134. Min, Y.L.; Chemello, F.; Li, H.; Rodriguez-Caycedo, C.; Sanchez-Ortiz, E.; Mireault, A.A.; McAnally, J.R.; Shelton, J.M.; Zhang, Y.; Bassel-Duby, R.; et al. Correction of Three Prominent Mutations in Mouse and Human Models of Duchenne Muscular Dystrophy by Single-Cut Genome Editing. Mol. Ther. 2020, 28, 2044–2055. [Google Scholar] [CrossRef]
  135. Zhang, Y.; Li, H.; Nishiyama, T.; McAnally, J.R.; Sanchez-Ortiz, E.; Huang, J.; Mammen, P.P.A.; Bassel-Duby, R.; Olson, E.N. A humanized knockin mouse model of Duchenne muscular dystrophy and its correction by CRISPR-Cas9 therapeutic gene editing. Mol. Ther. Nucleic Acids 2022, 29, 525–537. [Google Scholar] [CrossRef]
  136. Zhang, Y.; Li, H.; Min, Y.L.; Sanchez-Ortiz, E.; Huang, J.; Mireault, A.A.; Shelton, J.M.; Kim, J.; Mammen, P.P.A.; Bassel-Duby, R.; et al. Enhanced CRISPR-Cas9 correction of Duchenne muscular dystrophy in mice by a self-complementary AAV delivery system. Sci. Adv. 2020, 6, eaay6812. [Google Scholar] [CrossRef]
  137. Koo, T.; Lu-Nguyen, N.B.; Malerba, A.; Kim, E.; Kim, D.; Cappellari, O.; Cho, H.Y.; Dickson, G.; Popplewell, L.; Kim, J.S. Functional Rescue of Dystrophin Deficiency in Mice Caused by Frameshift Mutations Using Campylobacter jejuni Cas9. Mol. Ther. 2018, 26, 1529–1538. [Google Scholar] [CrossRef]
  138. Amoasii, L.; Long, C.; Li, H.; Mireault, A.A.; Shelton, J.M.; Sanchez-Ortiz, E.; McAnally, J.R.; Bhattacharyya, S.; Schmidt, F.; Grimm, D.; et al. Single-cut genome editing restores dystrophin expression in a new mouse model of muscular dystrophy. Sci. Transl. Med. 2017, 9, eaan8081. [Google Scholar] [CrossRef]
  139. Amoasii, L.; Li, H.; Zhang, Y.; Min, Y.L.; Sanchez-Ortiz, E.; Shelton, J.M.; Long, C.; Mireault, A.A.; Bhattacharyya, S.; McAnally, J.R.; et al. In vivo non-invasive monitoring of dystrophin correction in a new Duchenne muscular dystrophy reporter mouse. Nat. Commun. 2019, 10, 4537. [Google Scholar] [CrossRef]
  140. Amoasii, L.; Hildyard, J.C.W.; Li, H.; Sanchez-Ortiz, E.; Mireault, A.; Caballero, D.; Harron, R.; Stathopoulou, T.R.; Massey, C.; Shelton, J.M.; et al. Gene editing restores dystrophin expression in a canine model of Duchenne muscular dystrophy. Science 2018, 362, 86–91. [Google Scholar] [CrossRef]
  141. Hakim, C.H.; Kumar, S.R.P.; Pérez-López, D.O.; Wasala, N.B.; Zhang, D.; Yue, Y.; Teixeira, J.; Pan, X.; Zhang, K.; Million, E.D.; et al. Cas9-specific immune responses compromise local and systemic AAV CRISPR therapy in multiple dystrophic canine models. Nat. Commun. 2021, 12, 6769. [Google Scholar] [CrossRef]
  142. Zhang, Y.; Long, C.; Li, H.; McAnally, J.R.; Baskin, K.K.; Shelton, J.M.; Bassel-Duby, R.; Olson, E.N. CRISPR-Cpf1 correction of muscular dystrophy mutations in human cardiomyocytes and mice. Sci. Adv. 2017, 3, e1602814. [Google Scholar] [CrossRef]
  143. Mata López, S.; Balog-Alvarez, C.; Vitha, S.; Bettis, A.K.; Canessa, E.H.; Kornegay, J.N.; Nghiem, P.P. Challenges associated with homologous directed repair using CRISPR-Cas9 and TALEN to edit the DMD genetic mutation in canine Duchenne muscular dystrophy. PLoS ONE 2020, 15, e0228072. [Google Scholar]
  144. Lee, K.; Conboy, M.; Park, H.M.; Jiang, F.; Kim, H.J.; Dewitt, M.A.; Mackley, V.A.; Chang, K.; Rao, A.; Skinner, C.; et al. Nanoparticle delivery of Cas9 ribonucleoprotein and donor DNA in vivo induces homology-directed DNA repair. Nat. Biomed. Eng. 2017, 1, 889–901. [Google Scholar] [CrossRef]
  145. Pickar-Oliver, A.; Gough, V.; Bohning, J.D.; Liu, S.; Robinson-Hamm, J.N.; Daniels, H.; Majoros, W.H.; Devlin, G.; Asokan, A.; Gersbach, C.A. Full-length dystrophin restoration via targeted exon integration by AAV-CRISPR in a humanized mouse model of Duchenne muscular dystrophy. Mol. Ther. 2021, 29, 3243–3257. [Google Scholar] [CrossRef]
  146. Chemello, F.; Chai, A.C.; Li, H.; Rodriguez-Caycedo, C.; Sanchez-Ortiz, E.; Atmanli, A.; Mireault, A.A.; Liu, N.; Bassel-Duby, R.; Olson, E.N. Precise correction of Duchenne muscular dystrophy exon deletion mutations by base and prime editing. Sci. Adv. 2021, 7, eabg4910. [Google Scholar] [CrossRef]
  147. Ryu, S.M.; Koo, T.; Kim, K.; Lim, K.; Baek, G.; Kim, S.T.; Kim, H.S.; Kim, D.E.; Lee, H.; Chung, E.; et al. Adenine base editing in mouse embryos and an adult mouse model of Duchenne muscular dystrophy. Nat. Biotechnol. 2018, 36, 536–539. [Google Scholar] [CrossRef]
  148. Xu, L.; Zhang, C.; Li, H.; Wang, P.; Gao, Y.; Mokadam, N.A.; Ma, J.; Arnold, W.D.; Han, R. Efficient precise in vivo base editing in adult dystrophic mice. Nat. Commun. 2021, 12, 3719. [Google Scholar] [CrossRef]
  149. Li, J.; Wang, K.; Zhang, Y.; Qi, T.; Yuan, J.; Zhang, L.; Qiu, H.; Wang, J.; Yang, H.T.; Dai, Y.; et al. Therapeutic Exon Skipping Through a CRISPR-Guided Cytidine Deaminase Rescues Dystrophic Cardiomyopathy in Vivo. Circulation 2021, 144, 1760–1776. [Google Scholar] [CrossRef]
  150. Long, C.; Li, H.; Tiburcy, M.; Rodriguez-Caycedo, C.; Kyrychenko, V.; Zhou, H.; Zhang, Y.; Min, Y.L.; Shelton, J.M.; Mammen, P.P.A.; et al. Correction of diverse muscular dystrophy mutations in human engineered heart muscle by single-site genome editing. Sci. Adv. 2018, 4, eaap9004. [Google Scholar] [CrossRef]
  151. Lattanzi, A.; Duguez, S.; Moiani, A.; Izmiryan, A.; Barbon, E.; Martin, S.; Mamchaoui, K.; Mouly, V.; Bernardi, F.; Mavilio, F.; et al. Correction of the Exon 2 Duplication in DMD Myoblasts by a Single CRISPR/Cas9 System. Mol. Ther. Nucleic Acids 2017, 7, 11–19. [Google Scholar] [CrossRef] [PubMed]
  152. Pini, V.; Mariot, V.; Dumonceaux, J.; Counsell, J.; O’Neill, H.C.; Farmer, S.; Conti, F.; Muntoni, F. Transiently expressed CRISPR/Cas9 induces wild-type dystrophin in vitro in DMD patient myoblasts carrying duplications. Sci. Rep. 2022, 12, 3756. [Google Scholar] [CrossRef]
  153. Wang, D.N.; Wang, Z.Q.; Jin, M.; Lin, M.T.; Wang, N. CRISPR/Cas9-based genome editing for the modification of multiple duplications that cause Duchenne muscular dystrophy. Gene Ther. 2022. online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  154. Ousterout, D.G.; Kabadi, A.M.; Thakore, P.I.; Majoros, W.H.; Reddy, T.E. Multiplex CRISPR/Cas9-based genome editing for correction of dystrophin mutations that cause Duchenne muscular dystrophy. Nat. Commun. 2015, 6, 6244. [Google Scholar] [CrossRef] [PubMed]
  155. Chen, M.; Shi, H.; Gou, S.; Wang, X.; Li, L.; Jin, Q.; Wu, H.; Zhang, H.; Li, Y.; Wang, L.; et al. In vivo genome editing in mouse restores dystrophin expression in Duchenne muscular dystrophy patient muscle fibers. Genome Med. 2021, 13, 57. [Google Scholar] [CrossRef] [PubMed]
  156. Maggio, I.; Liu, J.; Janssen, J.M.; Chen, X.; Gonçalves, M.A. Adenoviral vectors encoding CRISPR/Cas9 multiplexes rescue dystrophin synthesis in unselected populations of DMD muscle cells. Sci. Rep. 2016, 6, 37051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Maggio, I.; Stefanucci, L.; Janssen, J.M.; Liu, J.; Chen, X.; Mouly, V.; Gonçalves, M.A. Selection-free gene repair after adenoviral vector transduction of designer nucleases: Rescue of dystrophin synthesis in DMD muscle cell populations. Nucleic Acids Res. 2016, 44, 1449–1470. [Google Scholar] [CrossRef] [PubMed]
  158. Brescia, M.; Janssen, J.M.; Liu, J.; Gonçalves, M. High-Capacity Adenoviral Vectors Permit Robust and Versatile Testing of DMD Gene Repair Tools and Strategies in Human Cells. Cells 2020, 9, 869. [Google Scholar] [CrossRef]
  159. Kyrychenko, V.; Kyrychenko, S.; Tiburcy, M.; Shelton, J.M.; Long, C.; Schneider, J.W.; Zimmermann, W.H.; Bassel-Duby, R.; Olson, E.N. Functional correction of dystrophin actin binding domain mutations by genome editing. JCI Insight 2017, 2, e95918. [Google Scholar] [CrossRef]
  160. Matre, P.R.; Mu, X.; Wu, J.; Danila, D.; Hall, M.A.; Kolonin, M.G.; Darabi, R.; Huard, J. CRISPR/Cas9-Based Dystrophin Restoration Reveals a Novel Role for Dystrophin in Bioenergetics and Stress Resistance of Muscle Progenitors. Stem Cells 2019, 37, 1615–1628. [Google Scholar] [CrossRef]
  161. Li, H.L.; Fujimoto, N.; Sasakawa, N.; Shirai, S.; Ohkame, T.; Sakuma, T.; Tanaka, M.; Amano, N.; Watanabe, A.; Sakurai, H.; et al. Precise correction of the dystrophin gene in duchenne muscular dystrophy patient induced pluripotent stem cells by TALEN and CRISPR-Cas9. Stem Cell Reports 2015, 4, 143–154. [Google Scholar] [CrossRef]
  162. Yuan, J.; Ma, Y.; Huang, T.; Chen, Y.; Peng, Y.; Li, B.; Li, J.; Zhang, Y.; Song, B.; Sun, X.; et al. Genetic Modulation of RNA Splicing with a CRISPR-Guided Cytidine Deaminase. Mol. Cell 2018, 72, 380–394. [Google Scholar] [CrossRef]
  163. Happi Mbakam, C.; Rousseau, J.; Tremblay, G.; Yameogo, P.; Tremblay, J.P. Prime Editing Permits the Introduction of Specific Mutations in the Gene Responsible for Duchenne Muscular Dystrophy. Int. J. Mol. Sci. 2022, 23, 6160. [Google Scholar] [CrossRef]
  164. Tuffery-Giraud, S.; Béroud, C.; Leturcq, F.; Yaou, R.B.; Hamroun, D.; Michel-Calemard, L.; Moizard, M.P.; Bernard, R.; Cossée, M.; Boisseau, P.; et al. Genotype-phenotype analysis in 2405 patients with a dystrophinopathy using the UMD-DMD database: A model of nationwide knowledgebase. Hum. Mutat. 2009, 30, 934–945. [Google Scholar] [CrossRef]
  165. Taylor, P.J.; Maroulis, S.; Mullan, G.L.; Pedersen, R.L.; Baumli, A.; Elakis, G.; Piras, S.; Walsh, C.; Prósper-Gutiérrez, B.; De La Puente-Alonso, F.; et al. Measurement of the clinical utility of a combined mutation detection protocol in carriers of Duchenne and Becker muscular dystrophy. J. Med. Genet. 2007, 44, 368–372. [Google Scholar] [CrossRef]
  166. Van Putten, M.; Hulsker, M.; Nadarajah, V.D.; van Heiningen, S.H.; van Huizen, E.; van Iterson, M.; Admiraal, P.; Messemaker, T.; den Dunnen, J.T.; t Hoen, P.A.; et al. The effects of low levels of dystrophin on mouse muscle function and pathology. PLoS ONE 2012, 7, e31937. [Google Scholar] [CrossRef] [Green Version]
  167. Van Putten, M.; van der Pijl, E.M.; Hulsker, M.; Verhaart, I.E.; Nadarajah, V.D.; van der Weerd, L.; Aartsma-Rus, A. Low dystrophin levels in heart can delay heart failure in mdx mice. J. Mol. Cell. Cardiol. 2014, 69, 17–23. [Google Scholar] [CrossRef]
  168. Jin, Y.; Shen, Y.; Su, X.; Weintraub, N.L.; Tang, Y. Effective restoration of dystrophin expression in iPSC (Mdx)-derived muscle progenitor cells using the CRISPR/Cas9 system and homology-directed repair technology. Comput. Struct. Biotechnol. J. 2020, 18, 765–773. [Google Scholar] [CrossRef]
  169. Zhang, Y.; Long, C.; Bassel-Duby, R.; Olson, E.N. Myoediting: Toward Prevention of Muscular Dystrophy by Therapeutic Genome Editing. Physiol. Rev. 2018, 98, 1205–1240. [Google Scholar] [CrossRef]
  170. Xie, R.; Wang, X.; Wang, Y.; Ye, M.; Zhao, Y.; Yandell, B.S.; Gong, S. pH-Responsive Polymer Nanoparticles for Efficient Delivery of Cas9 Ribonucleoprotein with or without Donor DNA. Adv. Mater. 2022, 34, e2110618. [Google Scholar] [CrossRef]
  171. Suzuki, K.; Tsunekawa, Y.; Hernandez-Benitez, R.; Wu, J.; Zhu, J.; Kim, E.J.; Hatanaka, F.; Yamamoto, M.; Araoka, T.; Li, Z.; et al. In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration. Nature 2016, 540, 144–149. [Google Scholar] [CrossRef]
  172. Suzuki, K.; Izpisua Belmonte, J.C. In vivo genome editing via the HITI method as a tool for gene therapy. J. Hum. Genet. 2018, 63, 157–164. [Google Scholar] [CrossRef]
  173. Newby, G.A.; Liu, D.R. In vivo somatic cell base editing and prime editing. Mol. Ther. 2021, 29, 3107–3124. [Google Scholar] [CrossRef]
  174. Zhou, C.; Sun, Y.; Yan, R.; Liu, Y.; Zuo, E.; Gu, C.; Han, L.; Wei, Y.; Hu, X.; Zeng, R.; et al. Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis. Nature 2019, 571, 275–278. [Google Scholar] [CrossRef]
  175. Grünewald, J.; Zhou, R.; Garcia, S.P.; Iyer, S.; Lareau, C.A.; Aryee, M.J.; Joung, J.K. Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature 2019, 569, 433–437. [Google Scholar] [CrossRef]
  176. Jin, S.; Zong, Y.; Gao, Q.; Zhu, Z.; Wang, Y.; Qin, P.; Liang, C.; Wang, D.; Qiu, J.L.; Zhang, F.; et al. Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice. Science 2019, 364, 292–295. [Google Scholar] [CrossRef]
  177. Lee, H.K.; Willi, M.; Miller, S.M.; Kim, S.; Liu, C.; Liu, D.R.; Hennighausen, L. Targeting fidelity of adenine and cytosine base editors in mouse embryos. Nat. Commun. 2018, 9, 4804. [Google Scholar] [CrossRef]
  178. Chen, P.J.; Hussmann, J.A.; Yan, J.; Knipping, F.; Ravisankar, P.; Chen, P.F.; Chen, C.; Nelson, J.W.; Newby, G.A.; Sahin, M.; et al. Enhanced prime editing systems by manipulating cellular determinants of editing outcomes. Cell 2021, 184, 5635–5652. [Google Scholar] [CrossRef]
  179. Chamberlain, J.S.; Metzger, J.; Reyes, M.; Townsend, D.; Faulkner, J.A. Dystrophin-deficient mdx mice display a reduced life span and are susceptible to spontaneous rhabdomyosarcoma. FASEB J. 2007, 21, 2195–2204. [Google Scholar] [CrossRef]
  180. Pastoret, C.; Sebille, A. mdx mice show progressive weakness and muscle deterioration with age. J. Neurol. Sci. 1995, 129, 97–105. [Google Scholar] [CrossRef]
  181. Lefaucheur, J.P.; Pastoret, C.; Sebille, A. Phenotype of dystrophinopathy in old mdx mice. Anat. Rec. 1995, 242, 70–76. [Google Scholar] [CrossRef] [PubMed]
  182. Lim, K.R.Q.; Nguyen, Q.; Dzierlega, K.; Huang, Y.; Yokota, T. CRISPR-Generated Animal Models of Duchenne Muscular Dystrophy. Genes 2020, 11, 342. [Google Scholar] [CrossRef] [PubMed]
  183. Xie, J.; Ge, W.; Li, N.; Liu, Q.; Chen, F.; Yang, X.; Huang, X.; Ouyang, Z.; Zhang, Q.; Zhao, Y.; et al. Efficient base editing for multiple genes and loci in pigs using base editors. Nat. Commun. 2019, 10, 2852. [Google Scholar] [CrossRef] [PubMed]
  184. Klymiuk, N.; Blutke, A.; Graf, A.; Krause, S.; Burkhardt, K.; Wuensch, A.; Krebs, S.; Kessler, B.; Zakhartchenko, V.; Kurome, M.; et al. Dystrophin-deficient pigs provide new insights into the hierarchy of physiological derangements of dystrophic muscle. Hum. Mol. Genet. 2013, 22, 4368–4382. [Google Scholar] [CrossRef]
  185. Valentine, B.A.; Cooper, B.J.; de Lahunta, A.; O’Quinn, R.; Blue, J.T. Canine X-linked muscular dystrophy. An animal model of Duchenne muscular dystrophy: Clinical studies. J. Neurol. Sci. 1988, 88, 69–81. [Google Scholar] [CrossRef]
  186. Sui, T.; Lau, Y.S.; Liu, D.; Liu, T.; Xu, L.; Gao, Y.; Lai, L.; Li, Z.; Han, R. A novel rabbit model of Duchenne muscular dystrophy generated by CRISPR/Cas9. Dis. Models Mech. 2018, 11, dmm032201. [Google Scholar] [CrossRef] [Green Version]
  187. Chen, Y.; Zheng, Y.; Kang, Y.; Yang, W.; Niu, Y.; Guo, X.; Tu, Z.; Si, C.; Wang, H.; Xing, R.; et al. Functional disruption of the dystrophin gene in rhesus monkey using CRISPR/Cas9. Hum. Mol. Genet. 2015, 24, 3764–3774. [Google Scholar] [CrossRef]
  188. McGreevy, J.W.; Hakim, C.H.; McIntosh, M.A.; Duan, D. Animal models of Duchenne muscular dystrophy: From basic mechanisms to gene therapy. Dis. Models Mech. 2015, 8, 195–213. [Google Scholar] [CrossRef]
  189. Oh, H.J.; Chung, E.; Kim, J.; Kim, M.J.; Kim, G.A.; Lee, S.H.; Ra, K.; Eom, K.; Park, S.; Chae, J.H.; et al. Generation of a Dystrophin Mutant in Dog by Nuclear Transfer Using CRISPR/Cas9-Mediated Somatic Cells: A Preliminary Study. Int. J. Mol. Sci. 2022, 23, 2898. [Google Scholar] [CrossRef]
  190. Egorova, T.V.; Zotova, E.D. CRISPR/Cas9-generated mouse model of Duchenne muscular dystrophy recapitulating a newly identified large 430 kb deletion in the human DMD gene. Dis. Models Mech. 2019, 12, dmm037655. [Google Scholar] [CrossRef]
  191. Deconinck, A.E.; Rafael, J.A.; Skinner, J.A.; Brown, S.C.; Potter, A.C.; Metzinger, L.; Watt, D.J.; Dickson, J.G.; Tinsley, J.M.; Davies, K.E. Utrophin-dystrophin-deficient mice as a model for Duchenne muscular dystrophy. Cell 1997, 90, 717–727. [Google Scholar] [CrossRef]
  192. Perkins, K.J.; Davies, K.E. The role of utrophin in the potential therapy of Duchenne muscular dystrophy. Neuromuscul. Disord. 2002, 12, S78–S89. [Google Scholar] [CrossRef]
  193. Smith, B.F.; Yue, Y.; Woods, P.R.; Kornegay, J.N.; Shin, J.H.; Williams, R.R.; Duan, D. An intronic LINE-1 element insertion in the dystrophin gene aborts dystrophin expression and results in Duchenne-like muscular dystrophy in the corgi breed. Lab. Investig. 2011, 91, 216–231. [Google Scholar] [CrossRef]
  194. Cozzi, F.; Cerletti, M.; Luvoni, G.C.; Lombardo, R.; Brambilla, P.G.; Faverzani, S.; Blasevich, F.; Cornelio, F.; Pozza, O.; Mora, M. Development of muscle pathology in canine X-linked muscular dystrophy. II. Quantitative characterization of histopathological progression during postnatal skeletal muscle development. Acta Neuropathol. 2001, 101, 469–478. [Google Scholar] [CrossRef]
  195. Wells, D.J. Tracking progress: An update on animal models for Duchenne muscular dystrophy. Dis. Models Mech. 2018, 11, dmm035774. [Google Scholar] [CrossRef]
  196. Samulski, R.J.; Muzyczka, N. AAV-Mediated Gene Therapy for Research and Therapeutic Purposes. Annu. Rev. Virol. 2014, 1, 427–451. [Google Scholar] [CrossRef]
  197. Crudele, J.M.; Chamberlain, J.S. AAV-based gene therapies for the muscular dystrophies. Hum. Mol. Genet. 2019, 28, R102–R107. [Google Scholar] [CrossRef]
  198. Frontera, W.R.; Ochala, J. Skeletal muscle: A brief review of structure and function. Calcif. Tissue Int. 2015, 96, 183–195. [Google Scholar] [CrossRef]
  199. Kornegay, J.N.; Li, J.; Bogan, J.R.; Bogan, D.J.; Chen, C.; Zheng, H.; Wang, B.; Qiao, C.; Howard, J.F., Jr.; Xiao, X. Widespread muscle expression of an AAV9 human mini-dystrophin vector after intravenous injection in neonatal dystrophin-deficient dogs. Mol. Ther. 2010, 18, 1501–1508. [Google Scholar] [CrossRef]
  200. Hinderer, C.; Katz, N.; Buza, E.L.; Dyer, C.; Goode, T.; Bell, P.; Richman, L.K.; Wilson, J.M. 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. [Google Scholar] [CrossRef]
  201. Buscara, L.; Gross, D.A.; Daniele, N. Of rAAV and Men: From Genetic Neuromuscular Disorder Efficacy and Toxicity Preclinical Studies to Clinical Trials and Back. J. Pers. Med. 2020, 10, 258. [Google Scholar] [CrossRef]
  202. Ren, C.; Kumar, S.; Shaw, D.R.; Ponnazhagan, S. Genomic stability of self-complementary adeno-associated virus 2 during early stages of transduction in mouse muscle in vivo. Hum. Gene Ther. 2005, 16, 1047–1057. [Google Scholar] [CrossRef]
  203. McCarty, D.M. Self-complementary AAV vectors; advances and applications. Mol. Ther. 2008, 16, 1648–1656. [Google Scholar] [CrossRef]
  204. Boutin, S.; Monteilhet, V.; Veron, P.; Leborgne, C.; Benveniste, O.; Montus, M.F.; 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. [Google Scholar] [CrossRef]
  205. Louis Jeune, V.; Joergensen, J.A.; Hajjar, R.J.; Weber, T. Pre-existing anti-adeno-associated virus antibodies as a challenge in AAV gene therapy. Hum. Gene Ther. Methods 2013, 24, 59–67. [Google Scholar] [CrossRef]
  206. Moreno, A.M.; Palmer, N.; Alemán, F.; Chen, G.; Pla, A.; Jiang, N.; Leong Chew, W.; Law, M.; Mali, P. Immune-orthogonal orthologues of AAV capsids and of Cas9 circumvent the immune response to the administration of gene therapy. Nat. Biomed. Eng. 2019, 3, 806–816. [Google Scholar] [CrossRef]
  207. Chew, W.L.; Tabebordbar, M.; Cheng, J.K.; Mali, P.; Wu, E.Y.; Ng, A.H.; Zhu, K.; Wagers, A.J.; Church, G.M. A multifunctional AAV-CRISPR-Cas9 and its host response. Nat. Methods 2016, 13, 868–874. [Google Scholar]
  208. Charlesworth, C.T.; Deshpande, P.S.; Dever, D.P.; Camarena, J.; Lemgart, V.T.; Cromer, M.K.; Vakulskas, C.A.; Collingwood, M.A.; Zhang, L.; Bode, N.M.; et al. Identification of preexisting adaptive immunity to Cas9 proteins in humans. Nat. Med. 2019, 25, 249–254. [Google Scholar] [CrossRef]
  209. Wagner, D.L.; Amini, L.; Wendering, D.J.; Burkhardt, L.M.; Akyüz, L.; Reinke, P.; Volk, H.D.; Schmueck-Henneresse, M. High prevalence of Streptococcus pyogenes Cas9-reactive T cells within the adult human population. Nat. Med. 2019, 25, 242–248. [Google Scholar] [CrossRef]
  210. Wienert, B.; Shin, J.; Zelin, E.; Pestal, K.; Corn, J.E. In vitro-transcribed guide RNAs trigger an innate immune response via the RIG-I pathway. PLoS Biol. 2018, 16, e2005840. [Google Scholar] [CrossRef]
  211. Kim, S.; Koo, T.; Jee, H.G.; Cho, H.Y.; Lee, G.; Lim, D.G.; Shin, H.S.; Kim, J.S. CRISPR RNAs trigger innate immune responses in human cells. Genome Res. 2018, 28, 367–373. [Google Scholar] [CrossRef] [PubMed]
  212. Ferdosi, S.R.; Ewaisha, R.; Moghadam, F.; Krishna, S.; Park, J.G.; Ebrahimkhani, M.R.; Kiani, S.; Anderson, K.S. Multifunctional CRISPR-Cas9 with engineered immunosilenced human T cell epitopes. Nat. Commun. 2019, 10, 1842. [Google Scholar] [CrossRef] [PubMed]
  213. Fu, Y.; Foden, J.A.; Khayter, C.; Maeder, M.L.; Reyon, D.; Joung, J.K.; Sander, J.D. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat. Biotechnol. 2013, 31, 822–826. [Google Scholar] [CrossRef] [PubMed]
  214. Lin, Y.; Cradick, T.J.; Brown, M.T.; Deshmukh, H.; Ranjan, P.; Sarode, N.; Wile, B.M.; Vertino, P.M.; Stewart, F.J.; Bao, G. CRISPR/Cas9 systems have off-target activity with insertions or deletions between target DNA and guide RNA sequences. Nucleic Acids Res. 2014, 42, 7473–7485. [Google Scholar] [CrossRef] [PubMed]
  215. Murugan, K.; Suresh, S.K.; Seetharam, A.S.; Severin, A.J.; Sashital, D.G. Systematic in vitro specificity profiling reveals nicking defects in natural and engineered CRISPR-Cas9 variants. Nucleic Acids Res. 2021, 49, 4037–4053. [Google Scholar] [CrossRef] [PubMed]
  216. Kleinstiver, B.P.; Pattanayak, V.; Prew, M.S.; Tsai, S.Q.; Nguyen, N.T.; Zheng, Z.; Joung, J.K. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature 2016, 529, 490–495. [Google Scholar] [CrossRef]
  217. Kleinstiver, B.P.; Tsai, S.Q.; Prew, M.S.; Nguyen, N.T.; Welch, M.M.; Lopez, J.M.; McCaw, Z.R.; Aryee, M.J.; Joung, J.K. Genome-wide specificities of CRISPR-Cas Cpf1 nucleases in human cells. Nat. Biotechnol. 2016, 34, 869–874. [Google Scholar] [CrossRef] [Green Version]
  218. Fu, Y.; Sander, J.D.; Reyon, D.; Cascio, V.M.; Joung, J.K. Improving CRISPR-Cas nuclease specificity using truncated guide RNAs. Nat. Biotechnol. 2014, 32, 279–284. [Google Scholar] [CrossRef]
  219. Kocak, D.D.; Josephs, E.A.; Bhandarkar, V.; Adkar, S.S.; Kwon, J.B.; Gersbach, C.A. Increasing the specificity of CRISPR systems with engineered RNA secondary structures. Nat. Biotechnol. 2019, 37, 657–666. [Google Scholar] [CrossRef]
  220. Lau, C.H.; Suh, Y. In vivo genome editing in animals using AAV-CRISPR system: Applications to translational research of human disease. F1000Res. 2017, 6, 2153. [Google Scholar] [CrossRef]
  221. Duddy, W.; Duguez, S.; Johnston, H.; Cohen, T.V.; Phadke, A.; Gordish-Dressman, H.; Nagaraju, K.; Gnocchi, V.; Low, S.; Partridge, T. Muscular dystrophy in the mdx mouse is a severe myopathy compounded by hypotrophy, hypertrophy and hyperplasia. Skelet Muscle 2015, 5, 16. [Google Scholar] [CrossRef]
  222. Wong, T.W.Y.; Ahmed, A.; Yang, G.; Maino, E.; Steiman, S.; Hyatt, E.; Chan, P.; Lindsay, K.; Wong, N.; Golebiowski, D.; et al. A novel mouse model of Duchenne muscular dystrophy carrying a multi-exonic Dmd deletion exhibits progressive muscular dystrophy and early-onset cardiomyopathy. Dis. Models Mech. 2020, 13, dmm045369. [Google Scholar] [CrossRef]
  223. Ancel, S.; Stuelsatz, P.; Feige, J.N. Muscle Stem Cell Quiescence: Controlling Stemness by Staying Asleep. Trends Cell Biol. 2021, 31, 556–568. [Google Scholar] [CrossRef]
  224. Yin, H.; Price, F.; Rudnicki, M.A. Satellite cells and the muscle stem cell niche. Physiol. Rev. 2013, 93, 23–67. [Google Scholar] [CrossRef]
  225. Nance, M.E.; Shi, R.; Hakim, C.H.; Wasala, N.B.; Yue, Y.; Pan, X.; Zhang, T.; Robinson, C.A.; Duan, S.X.; Yao, G.; et al. AAV9 Edits Muscle Stem Cells in Normal and Dystrophic Adult Mice. Mol. Ther. 2019, 27, 1568–1585. [Google Scholar] [CrossRef]
  226. Kwon, J.B.; Ettyreddy, A.R.; Vankara, A.; Bohning, J.D.; Devlin, G.; Hauschka, S.D.; Asokan, A.; Gersbach, C.A. In Vivo Gene Editing of Muscle Stem Cells with Adeno-Associated Viral Vectors in a Mouse Model of Duchenne Muscular Dystrophy. Mol. Ther. Methods Clin. Dev. 2020, 19, 320–329. [Google Scholar] [CrossRef]
  227. Goldstein, J.M.; Tabebordbar, M.; Zhu, K.; Wang, L.D.; Messemer, K.A.; Peacker, B.; Ashrafi Kakhki, S.; Gonzalez-Celeiro, M.; Shwartz, Y.; Cheng, J.K.W.; et al. In Situ Modification of Tissue Stem and Progenitor Cell Genomes. Cell Rep. 2019, 27, 1254–1264. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Therapeutic strategies for CRISPR-based genome editing. (A) Double-cut exon excision using two sgRNAs. In the end, exon 51 is removed from the genome by NHEJ machinery. (B) Single-cut exon skipping by disruption of the splice donor site. The exon 44 skipping induces the splicing of exon 42 to exon 45 at the mRNA level. (C) Single-cut exon reframing. Small insertions or deletions occurring in the exon 51 region can restore the reading frame with at least one-third probability. (D) Accurate mutation correction in the exon 23 by HDR pathway in the existence of a donor template. (E) HITI-mediated exon knockin. Exon 52 is precisely incorporated back into the genome by NHEJ machinery. (F) Base editing-driven correction. The ABE is used to treat a point mutation in the exon 53 via inducing A•T to G•C transition, whereas the CBE is deployed to mutate the GT at the splice donor site of exon 4, causing exon 4 skipping in the transcripts. (G) Prime editing-induced exon reframing. Prime editing can introduce all genomic modifications, such as single-nucleotide transitions, small insertions, and short deletions.
Figure 1. Therapeutic strategies for CRISPR-based genome editing. (A) Double-cut exon excision using two sgRNAs. In the end, exon 51 is removed from the genome by NHEJ machinery. (B) Single-cut exon skipping by disruption of the splice donor site. The exon 44 skipping induces the splicing of exon 42 to exon 45 at the mRNA level. (C) Single-cut exon reframing. Small insertions or deletions occurring in the exon 51 region can restore the reading frame with at least one-third probability. (D) Accurate mutation correction in the exon 23 by HDR pathway in the existence of a donor template. (E) HITI-mediated exon knockin. Exon 52 is precisely incorporated back into the genome by NHEJ machinery. (F) Base editing-driven correction. The ABE is used to treat a point mutation in the exon 53 via inducing A•T to G•C transition, whereas the CBE is deployed to mutate the GT at the splice donor site of exon 4, causing exon 4 skipping in the transcripts. (G) Prime editing-induced exon reframing. Prime editing can introduce all genomic modifications, such as single-nucleotide transitions, small insertions, and short deletions.
Cells 11 02964 g001aCells 11 02964 g001b
Table 1. CRISPR-mediated therapeutic strategies in preclinical DMD animals.
Table 1. CRISPR-mediated therapeutic strategies in preclinical DMD animals.
StrategyMutationNucleaseTarget RegionDMD ModelDeliveryInfusionReference
Double-cut
exon exicision
Ex23 mutSpCas9i22, i23mdx miceAAV9IM, IV, IP, RO[34,35]
Ex23 mutSaCas9i22, i23mdx miceAAV8 IM, IV, IP[36,37]
ΔEx52SpCas9i50, i51DMDΔ52 miceAAV9IM, IV[39]
ΔEx52SaCas9Ex47, Ex58hDMDΔ52/mdx miceAAV9IV[40]
Ex23 mutSaCas9i20, i23mdx miceAAVrh74IV[43]
Dup Ex18-30SaCas9i21Dup 18-30 miceAAV9IV[125]
Ex23 mutSpCas9i20, i23mdx miceAAVIM[126]
Ex23 mutSaCas9i22, i23mdx miceAAV9IM, IV, IP[127]
Ex53 mutSaCas9,
SpCas9
i51, i53mdx4Cv miceAAV6IM, RO[128,129]
ΔEx45SpCas9i44, i55hDMDΔ45/mdxD2 micePlasmidIM[130]
Ex23 mutSaCas9i20, i23mdx/Utr+/− miceAAVIV[131]
Single-cut
exon skipping
and reframing
ΔEx44SpCas9Ex45DMDΔ44 miceAAV9IM, IP[132]
ΔEx50SaCas9-KKHEx51DMDΔ50 miceAAV9IP[133]
ΔEx43SpCas9Ex44DMDΔ43 miceAAV9IM[134]
ΔEx45SpCas9Ex44DMDΔ45 miceAAV9IM[134]
ΔEx52SpCas9Ex53DMDΔ52 miceAAV9IM[134]
ΔEx50SpCas9-VRQREx51DMDΔ50;h51KI miceAAV9IP[135]
ΔEx44SpCas9Ex45DMDΔ44 miceAAV9IP[44,136]
Ex23 mutCjCas9Ex23DMD/Ex23 mut miceAAV9IM[137]
ΔEx50SpCas9Ex51DMDΔ50 miceAAV9IM, IP[138]
ΔEx50SpCas9Ex51ΔEx50-Dmd-Luc miceAAV9IM, IP[139]
ΔEx50SpCas9Ex51DMDΔ50 dogsAAV9IM, IV[140]
Pseudo Ex13SpCas9i13WCMD dogsAAV8IM, IV[141]
Pseudo Ex19SpCas9i19LRMD dogsAAV8IM, IV[141]
HDR-based
correction
Ex23 mutSpCas9Ex23mdx miceInjectionZygote[124]
Ex53 mutSpCas9Ex53mdx4Cv miceAAV6IM[128]
Ex23 mutLbCas12aEx23mdx miceInjectionZygote[142]
i6 mutSpCas9i6GRMD dogsPlasmidIM[143]
Ex23 mutSpCas9Ex23mdx miceNanoparticleIM[144]
Exon knockinEx51 mutSaCas9Ex52hDMDΔ52/mdx miceAAV9IM, IV[145]
Base editingEx51 mutABEmax-nSpCas9Ex50DMDΔ51 miceAAV9IM[146]
Ex20 mutABE-nSpCas9Ex20DMD/Ex20* miceAAV9IM[147]
Ex53 mutABE-nSpCas9-iNGEx53mdx4Cv miceAAV9IV[148]
Ex4 mutCBE-nSaCas9Ex4DmdE4* miceAAV9IP[149]
Ex, exon; i, intron; mut, mutation; AAV, adeno-associated viral vector; Δ, deletion; Dup, duplication; IM, intramuscular; IV, intravenous; IP, intraperitoneal; RO, Retro-orbital; KI, Knockin.
Table 2. CRISPR-mediated therapeutic strategies in human DMD cells.
Table 2. CRISPR-mediated therapeutic strategies in human DMD cells.
StrategyMutationNucleaseTarget RegionDMD ModelDeliveryReference
Double-cut
exon exicision
ΔEx46-51SpCas9i44, i55human iPSCselectroporation[38]
ΔEx52SpCas9i50, i51human iPSCsAAV6 [39]
ΔEx49-50,
ΔEx50-52,
ΔEx51-53,
ΔEx51-56
SaCas9Ex47, Ex58human myoblastslentivirus[40]
Dup Ex18-30SpCas9i27human myoblastslentivirus[122]
Dup Ex55-59SpCas9i54human iPSCsnucleofection[150]
Dup Ex2SpCas9i2human myoblastslentivirus[151]
Dup Ex3-16SpCas9i9human myoblastslentivirus[152]
Dup Ex18-25SpCas9i25human myoblastslentivirus[153]
ΔEx48-50SpCas9i50, i51human myoblastselectroporation[154]
ΔEx48-50SpCas9i44, i55human myoblastselectroporation[154]
ΔEx51SpCas9,
AsCas12a
i44, i55;
i45, i54
human myoblastselectroporation[155]
ΔEx45-52SpCas9i52, i53human myoblastsadenovirus[156,157]
ΔEx45-52,
ΔEx48-50
SpCas9,
eSpCas9(1.1)
i43, i54human myoblastsadenovirus[156,157,158]
ΔEx8-9SpCas9i2, i7;
i5, i7;
i6, i11
human iPSCsnucleofection[159]
ΔEx3-7SpCas9i7, i9human iPSCsnucleofection[159]
Ex23 mutSpCas9i22, i23mouse myoblastslipotransfection[160]
Single-cut
exon skipping
and reframing
ΔEx44SpCas9Ex45human iPSCsnucleofection[132]
ΔEx48-50SaCas9-KKHEx51human iPSCsnucleofection[133]
ΔEx43,
ΔEx45
SpCas9Ex44human iPSCsnucleofection[134]
ΔEx52SpCas9Ex51human iPSCsnucleofection[134]
ΔEx48-50SpCas9-VRQREx51human iPSCsnucleofection[135]
ΔEx48-50LbCas12a,
AsCas12a
Ex51human iPSCsnucleofection[142]
ΔEx48-50SpCas9Ex51human iPSCsnucleofection[150]
Pseudo Ex47SpCas9i47human iPSCsnucleofection[150]
ΔEx48-50eSpCas9(1.1)Ex51human myoblastsadenovirus[158]
HDR-based
knockin
i6 mutSpCas9i6canine myoblastsnucleofection[143]
ΔEx44SpCas9Ex44human iPSCselectroporation[161]
Base editingΔEx51ABEmax-nSpCas9Ex50human iPSCsnucleofection[146]
ΔEx51CBE-nSaCas9Ex50human iPSCslipotransfection[162]
Prime editingΔEx51PE2-dSpCas9Ex52human iPSCsnucleofection[146]
Ex6 mutPE2-nSpCas9Ex6human myoblastselectroporation[163]
Δ, deletion; Ex, exon; i, intron; iPSC, induced pluripotent stem cell; AAV, adeno-associated viral vector; Dup, duplication; mut, mutation.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chen, G.; Wei, T.; Yang, H.; Li, G.; Li, H. CRISPR-Based Therapeutic Gene Editing for Duchenne Muscular Dystrophy: Advances, Challenges and Perspectives. Cells 2022, 11, 2964. https://doi.org/10.3390/cells11192964

AMA Style

Chen G, Wei T, Yang H, Li G, Li H. CRISPR-Based Therapeutic Gene Editing for Duchenne Muscular Dystrophy: Advances, Challenges and Perspectives. Cells. 2022; 11(19):2964. https://doi.org/10.3390/cells11192964

Chicago/Turabian Style

Chen, Guofang, Tingyi Wei, Hui Yang, Guoling Li, and Haisen Li. 2022. "CRISPR-Based Therapeutic Gene Editing for Duchenne Muscular Dystrophy: Advances, Challenges and Perspectives" Cells 11, no. 19: 2964. https://doi.org/10.3390/cells11192964

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop