Abstract:Duchenne muscular dystrophy (DMD) is an X-linked recessive hereditary disease caused by mutations in the DMD gene that encodes dystrophin. It is characterized by progressive muscle weakness and degeneration of skeletal muscle and myocardium due to the absence of dystrophin. The disease often occurs at the age of 2-5 years, and most children may die of heart failure or respiratory insufficiency at the age of around 20 years. At present, supportive therapy is often used in clinical practice to improve symptoms, but this cannot improve the outcome of this disease. The development of gene therapy brings new hope to the cure of this disease. This article summarizes gene replacement therapy for DMD, including the research advances in DMD gene transduction technology mediated by adeno-associated virus, utrophin protein upregulation technology, and clustered regularly interspaced short palindromic repeat gene editing technology, and reviews the recommendations to solve the issues of adeno-associated viral load, long-term effective expression of transgenic products, and utrophin protein expression, in order to provide a reference for further research.
Boutin S, Monteilhet V, Veron P, et al. 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[J]. Hum Gene Ther, 2010, 21(6):704-712.
[8]
Wu Z, Sun J, Zhang T, et al. Optimization of self-complementary AAV vectors for liver-directed expression results in sustained correction of hemophilia B at low vector dose[J]. Mol Ther, 2008, 16(2):280-289.
[9]
Koo T, Popplewell L, Athanasopoulos T, et al. Triple trans-splicing adeno-associated virus vectors capable of transferring the coding sequence for full-length dystrophin protein into dystrophic mice[J]. Hum Gene Ther, 2014, 25(2):98-108.
[10]
Ferreira V, Petry H, Salmon F. Immune responses to AAV-vectors, the glybera example from bench to bedside[J]. Front Immunol, 2014, 5:82.
Dupont JB. Restriction factors against recombinant adeno-associated virus vectormediated gene transfer in dystrophin-deficient muscles[J]. Curr Gene Ther, 2016, 16(3):168-183.
[13]
Kodippili K, Hakim CH, Pan X, et al. Dual AAV gene therapy for duchenne muscular dystrophy with a 7-kb mini-dystrophin gene in the canine model[J]. Hum Gene Ther, 2018, 29(3):299-311.
[14]
Nance ME, Duan D. Perspective on adeno-associated virus capsid modification for duchenne muscular dystrophy gene therapy[J]. Hum Gene Ther, 2015, 26(12):786-800.
[15]
Wang Z, Tapscott SJ, Chamberlain JS, et al. Immunity and AAV-mediated gene therapy for muscular dystrophies in large animal models and human trials[J]. Front Microbiol, 2011, 2:201.
[16]
Hayashita-Kinoh H, Yugeta N, Okada H, et al. Intra-amniotic rAAV-mediated microdystrophin gene transfer improves canine X-linked muscular dystrophy and may induce immune tolerance[J]. Mol Ther, 2015, 23(4):627-637.
[17]
George LA, Sullivan SK, Giermasz A, et al. Hemophilia B gene therapy with a high-specific-activity factor IX variant[J]. N Engl J Med, 2017, 377(23):2215-2227.
[18]
Le Guiner C, Servais L, Montus M, et al. Long-term microdystrophin gene therapy is effective in a canine model of Duchenne muscular dystrophy[J]. Nat Commun, 2017, 8:16105.
[19]
Dupont JB, Tournaire B, Georger C, et al. Short-lived recombinant adeno-associated virus transgene expression in dystrophic muscle is associated with oxidative damage to transgene mRNA[J]. Mol Ther Methods Clin Dev, 2015, 2:15010.
[20]
Guiraud S, Chen H, Burns DT, et al. Advances in genetic therapeutic strategies for Duchenne muscular dystrophy[J]. Exp Physiol, 2015, 100(12):1458-1467.
[21]
Weir AP, Burton EA, Harrod G, et al. A-and B-utrophin have different expression patterns and are differentially up-regulated in mdx muscle[J]. J Biol Chem, 2002, 277(47):45285-45290.
[22]
van Westering TL, Betts CA, Wood MJ. Current understanding of molecular pathology and treatment of cardiomyopathy in duchenne muscular dystrophy[J]. Molecules, 2015, 20(5):8823-8855.
[23]
Tinsley J, Robinson N, Davies KE. Safety, tolerability, and pharmacokinetics of SMT C1100, a 2-arylbenzoxazole utrophin modulator, following single-and multiple-dose administration to healthy male adult volunteers[J]. J Clin Pharmacol, 2015, 55(6):698-707.
[24]
Ricotti V, Spinty S, Roper H, et al. Safety, tolerability, and pharmacokinetics of SMT C1100, a 2-arylbenzoxazole utrophin modulator, following single-and multiple-dose administration to pediatric patients with duchenne muscular dystrophy[J]. PLoS One, 2016, 11(4):e0152840.
[25]
Guiraud S, Squire SE, Edwards B, et al. Second-generation compound for the modulation of utrophin in the therapy of DMD[J]. Hum Mol Genet, 2015, 24(15):4212-4224.
[26]
Strimpakos G, Corbi N, Pisani C, et al. Novel adeno-associated viral vector delivering the utrophin gene regulator jazz counteracts dystrophic pathology in mdx mice[J]. J Cell Physiol, 2014, 229(9):1283-1291.
[27]
Sonnemann KJ, Heun-Johnson H, Turner AJ, et al. Functional substitution by TAT-utrophin in dystrophin-deficient mice[J]. PLoS Med, 2009, 6(5):e1000083.
[28]
Call JA, Ervasti JM, Lowe DA. TAT-μUtrophin mitigates the pathophysiology of dystrophin and utrophin double-knockout mice[J]. J Appl Physiol (1985), 2011, 111(1):200-205.
[29]
Peladeau C, Ahmed A, Amirouche A, et al. Combinatorial therapeutic activation with heparin and AICAR stimulates additive effects on utrophin A expression in dystrophic muscles[J]. Hum Mol Genet, 2016, 25(1):24-43.
[30]
Ljubicic V, Jasmin BJ. Metformin increases peroxisome proliferator-activated receptor γ Co-activator-1α and utrophin a expression in dystrophic skeletal muscle[J]. Muscle Nerve, 2015, 52(1):139-142.
[31]
Wojtal D, Kemaladewi DU, Malam Z, et al. Spell checking nature:versatility of CRISPR/Cas9 for developing treatments for inherited disorders[J]. Am J Hum Genet, 2016, 98(1):90-101.
[32]
Juretić N, Diaz J, Romero F, et al. Interleukin-6 and neuregulin-1 as regulators of utrophin expression via the activation of NRG-1/ErbB signaling pathway in mdx cells[J]. Biochim Biophys Acta, 2017, 1863(3):770-780.
[33]
Duan D. A new kid on the playground of CRISPR DMD therapy[J]. Hum Gene Ther Clin Dev, 2017, 28(2):62-64.
[34]
Iyombe-Engembe JP, Ouellet DL, Barbeau X, et al. Efficient restoration of the dystrophin gene reading frame and protein structure in DMD myoblasts using the CinDel method[J]. Mol Ther Nucleic Acids, 2016, 5:e283.
[35]
Gee P, Xu H, Hotta A. Cellular reprogramming, genome editing, and alternative CRISPR Cas9 technologies for precise gene therapy of duchenne muscular dystrophy[J]. Stem Cells Int, 2017, 2017:8765154.
[36]
Ousterout DG, Kabadi AM, Thakore PI, et al. Multiplex CRISPR/Cas9-based genome editing for correction of dystrophin mutations that cause Duchenne muscular dystrophy[J]. Nat Commun, 2015, 6:6244.
[37]
Young CS, Hicks MR, Ermolova NV, et al. A single CRISPR-Cas9 deletion strategy that targets the majority of DMD patients restores dystrophin function in hiPSC-derived muscle cells[J]. Cell Stem Cell, 2016, 18(4):533-540.
[38]
Young CS, Mokhonova E, Quinonez M, et al. Creation of a novel humanized dystrophic mouse model of duchenne muscular dystrophy and application of a CRISPR/Cas9 gene editing therapy[J]. J Neuromuscul Dis, 2017, 4(2):139-145.
[39]
Lattanzi A, Duguez S, Moiani A, et al. Correction of the Exon 2 duplication in DMD myoblasts by a single CRISPR/Cas9 system[J]. Mol Ther Nucleic Acids, 2017, 7:11-19.
Zetsche B, Gootenberg JS, Abudayyeh OO, et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system[J]. Cell, 2015, 163(3):759-771.
[42]
Zaidi SS, Mahfouz MM, Mansoor S. CRISPR-Cpf1:a new tool for plant genome editing[J]. Trends Plant Sci, 2017, 22(7):550-553.
[43]
Zhang Y, Long C, Li H, et al. CRISPR-Cpf1 correction of muscular dystrophy mutations in human cardiomyocytes and mice[J]. Sci Adv, 2017, 3(4):e1602814.
[44]
Perrin A, Rousseau J, Tremblay JP. Increased expression of laminin subunit alpha 1 chain by dCas9-VP160[J]. Mol Ther Nucleic Acids, 2017, 6:68-79.
[45]
Komor AC, Kim YB, Packer MS, et al. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage[J]. Nature, 2016, 533(7603):420-424.
[46]
Fu Y, Sander JD, Reyon D, et al. Improving CRISPR-Cas nuclease specificity using truncated guide RNAs[J]. Nat Biotechnol, 2014, 32(3):279-284.
[47]
Wang JZ, Wu P, Shi ZM, et al. The AAV-mediated and RNA-guided CRISPR/Cas9 system for gene therapy of DMD and BMD[J]. Brain Dev, 2017, 39(7):547-556.
[48]
Kim S, Koo T, Jee HG, et al. CRISPR RNAs trigger innate immune responses in human cells[J]. Genome Res, 2018.[Epub ahead of print].