Skip to main content

Gene Therapy for Central Nervous System in Duchenne Muscular Dystrophy

  • Chapter
  • First Online:
Book cover Muscle Gene Therapy

Abstract

The development of molecular therapies enabling compensation of brain alterations in Duchenne muscular dystrophy is a major objective given the high level of functional impairment associated with intellectual disability and neuropsychiatric disorders in this syndrome. Functional and preclinical studies in mice lacking distinct brain dystrophins identified an accurate set of phenotypes, from the molecular to neurophysiological and behavioral levels, which can be used as markers of efficacy for brain gene therapy. Pioneer studies in this past decade provided encouraging results, demonstrating that both dmd-gene splice-switching correction and replacement strategies hold realistic prospects to rescue expression and function of the brain full-length (Dp427) or short C-terminal (Dp71) dystrophins responsible for variable degrees of cognitive impairment in DMD. Strategies that could correct or alleviate both muscle and brain dysfunctions entail selection of molecular tools able to cross the blood-brain barrier following systemic delivery, to largely spread in neural tissues, and to selectively target the neural cell types (neurons, astrocytes) that require rescued expression of distinct brain dystrophins. Recent breakthroughs show that this can be achieved by engineering naked antisense oligonucleotides with specific chemistries and/or adeno-associated virus vectors with selective capsid properties, thus raising new hopes to bring gene therapy closer to whole-body delivery and full treatment of DMD symptoms.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 259.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 329.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Cotton S, Voudouris NJ, Greenwood KM (2001) Intelligence and Duchenne muscular dystrophy: full-scale, verbal, and performance intelligence quotients. Dev Med Child Neurol 43(7):497–501

    Article  CAS  Google Scholar 

  2. Poysky J (2007) Behavior patterns in Duchenne muscular dystrophy: report on the Parent Project Muscular Dystrophy behavior workshop 8-9 of December 2006, Philadelphia, USA. Neuromuscul Disord 17(11–12):986–994. https://doi.org/10.1016/j.nmd.2007.06.465

    Article  PubMed  Google Scholar 

  3. Hendriksen JG, Vles JS (2008) Neuropsychiatric disorders in males with duchenne muscular dystrophy: frequency rate of attention-deficit hyperactivity disorder (ADHD), autism spectrum disorder, and obsessive--compulsive disorder. J Child Neurol 23(5):477–481. https://doi.org/10.1177/0883073807309775

    Article  PubMed  Google Scholar 

  4. Cyrulnik SE, Fee RJ, Batchelder A, Kiefel J, Goldstein E, Hinton VJ (2008) Cognitive and adaptive deficits in young children with Duchenne muscular dystrophy (DMD). J Int Neuropsychol Soc 14(5):853–861. https://doi.org/10.1017/S135561770808106X

    Article  PubMed  Google Scholar 

  5. Lidov HG, Byers TJ, Watkins SC, Kunkel LM (1990) Localization of dystrophin to postsynaptic regions of central nervous system cortical neurons. Nature 348(6303):725–728. https://doi.org/10.1038/348725a0

    Article  CAS  PubMed  Google Scholar 

  6. Ricotti V, Mandy WP, Scoto M, Pane M, Deconinck N, Messina S, Mercuri E, Skuse DH, Muntoni F (2016) Neurodevelopmental, emotional, and behavioural problems in Duchenne muscular dystrophy in relation to underlying dystrophin gene mutations. Dev Med Child Neurol 58(1):77–84. https://doi.org/10.1111/dmcn.12922

    Article  PubMed  Google Scholar 

  7. Perronnet C, Vaillend C (2010) Dystrophins, utrophins, and associated scaffolding complexes: role in mammalian brain and implications for therapeutic strategies. J Biomed Biotechnol 2010:849426. https://doi.org/10.1155/2010/849426

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Cyrulnik SE, Hinton VJ (2008) Duchenne muscular dystrophy: a cerebellar disorder? Neurosci Biobehav Rev 32(3):486–496. https://doi.org/10.1016/j.neubiorev.2007.09.001

    Article  CAS  PubMed  Google Scholar 

  9. Snow WM, Anderson JE, Jakobson LS (2013) Neuropsychological and neurobehavioral functioning in Duchenne muscular dystrophy: a review. Neurosci Biobehav Rev 37(5):743–752. https://doi.org/10.1016/j.neubiorev.2013.03.016

    Article  PubMed  Google Scholar 

  10. Hinton VJ, De Vivo DC, Nereo NE, Goldstein E, Stern Y (2000) Poor verbal working memory across intellectual level in boys with Duchenne dystrophy. Neurology 54(11):2127–2132

    Article  CAS  Google Scholar 

  11. Wicksell RK, Kihlgren M, Melin L, Eeg-Olofsson O (2004) Specific cognitive deficits are common in children with Duchenne muscular dystrophy. Dev Med Child Neurol 46(3):154–159

    Article  Google Scholar 

  12. Cotton SM, Voudouris NJ, Greenwood KM (2005) Association between intellectual functioning and age in children and young adults with Duchenne muscular dystrophy: further results from a meta-analysis. Dev Med Child Neurol 47(4):257–265

    Article  Google Scholar 

  13. Mento G, Tarantino V, Bisiacchi PS (2011) The neuropsychological profile of infantile Duchenne muscular dystrophy. Clin Neuropsychol 25(8):1359–1377. https://doi.org/10.1080/13854046.2011.617782

    Article  PubMed  Google Scholar 

  14. Rapaport D, Passos-Bueno MR, Brandao L, Love D, Vainzof M, Zatz M (1991) Apparent association of mental retardation and specific patterns of deletions screened with probes cf56a and cf23a in Duchenne muscular dystrophy. Am J Med Genet 39(4):437–441. https://doi.org/10.1002/ajmg.1320390414

    Article  CAS  PubMed  Google Scholar 

  15. Lenk U, Hanke R, Thiele H, Speer A (1993) Point mutations at the carboxy terminus of the human dystrophin gene: implications for an association with mental retardation in DMD patients. Hum Mol Genet 2(11):1877–1881

    Article  CAS  Google Scholar 

  16. Desguerre I, Christov C, Mayer M, Zeller R, Becane HM, Bastuji-Garin S, Leturcq F, Chiron C, Chelly J, Gherardi RK (2009) Clinical heterogeneity of duchenne muscular dystrophy (DMD): definition of sub-phenotypes and predictive criteria by long-term follow-up. PLoS One 4(2):e4347. https://doi.org/10.1371/journal.pone.0004347

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Taylor PJ, Betts GA, Maroulis S, Gilissen C, Pedersen RL, Mowat DR, Johnston HM, Buckley MF (2010) Dystrophin gene mutation location and the risk of cognitive impairment in Duchenne muscular dystrophy. PLoS One 5(1):e8803. https://doi.org/10.1371/journal.pone.0008803

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Daoud F, Angeard N, Demerre B, Martie I, Benyaou R, Leturcq F, Cossee M, Deburgrave N, Saillour Y, Tuffery S, Urtizberea A, Toutain A, Echenne B, Frischman M, Mayer M, Desguerre I, Estournet B, Reveillere C, Penisson B, Cuisset JM, Kaplan JC, Heron D, Rivier F, Chelly J (2009) Analysis of Dp71 contribution in the severity of mental retardation through comparison of Duchenne and Becker patients differing by mutation consequences on Dp71 expression. Hum Mol Genet 18(20):3779–3794. https://doi.org/10.1093/hmg/ddp320

    Article  CAS  PubMed  Google Scholar 

  19. D’Angelo MG, Lorusso ML, Civati F, Comi GP, Magri F, Del Bo R, Guglieri M, Molteni M, Turconi AC, Bresolin N (2011) Neurocognitive profiles in Duchenne muscular dystrophy and gene mutation site. Pediatr Neurol 45(5):292–299. https://doi.org/10.1016/j.pediatrneurol.2011.08.003

    Article  PubMed  PubMed Central  Google Scholar 

  20. Miranda R, Nagapin F, Bozon B, Laroche S, Aubin T, Vaillend C (2015) Altered social behavior and ultrasonic communication in the dystrophin-deficient mdx mouse model of Duchenne muscular dystrophy. Mol Autism 6:60. https://doi.org/10.1186/s13229-015-0053-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Chaussenot R, Edeline JM, Le Bec B, El Massioui N, Laroche S, Vaillend C (2015) Cognitive dysfunction in the dystrophin-deficient mouse model of Duchenne muscular dystrophy: a reappraisal from sensory to executive processes. Neurobiol Learn Mem 124:111–122. https://doi.org/10.1016/j.nlm.2015.07.006

    Article  CAS  PubMed  Google Scholar 

  22. Pastoret C, Sebille A (1995) mdx mice show progressive weakness and muscle deterioration with age. J Neurol Sci 129(2):97–105

    Article  CAS  Google Scholar 

  23. Rafael JA, Nitta Y, Peters J, Davies KE (2000) Testing of SHIRPA, a mouse phenotypic assessment protocol, on Dmd(mdx) and Dmd(mdx3cv) dystrophin-deficient mice. Mamm Genome 11(9):725–728

    Article  CAS  Google Scholar 

  24. Grady RM, Wozniak DF, Ohlemiller KK, Sanes JR (2006) Cerebellar synaptic defects and abnormal motor behavior in mice lacking alpha- and beta-dystrobrevin. J Neurosci 26(11):2841–2851. https://doi.org/10.1523/JNEUROSCI.4823-05.2006

    Article  CAS  PubMed  Google Scholar 

  25. Perronnet C, Chagneau C, Le Blanc P, Samson-Desvignes N, Mornet D, Laroche S, De La Porte S, Vaillend C (2012) Upregulation of brain utrophin does not rescue behavioral alterations in dystrophin-deficient mice. Hum Mol Genet 21(10):2263–2276. https://doi.org/10.1093/hmg/dds047

    Article  CAS  PubMed  Google Scholar 

  26. Vianello S, Yu H, Voisin V, Haddad H, He X, Foutz AS, Sebrie C, Gillet B, Roulot M, Fougerousse F, Perronnet C, Vaillend C, Matecki S, Escolar D, Bossi L, Israel M, de la Porte S (2013) Arginine butyrate: a therapeutic candidate for Duchenne muscular dystrophy. FASEB J 27(6):2256–2269. https://doi.org/10.1096/fj.12-215723

    Article  CAS  PubMed  Google Scholar 

  27. Goyenvalle A, Griffith G, Babbs A, El Andaloussi S, Ezzat K, Avril A, Dugovic B, Chaussenot R, Ferry A, Voit T, Amthor H, Buhr C, Schurch S, Wood MJ, Davies KE, Vaillend C, Leumann C, Garcia L (2015) Functional correction in mouse models of muscular dystrophy using exon-skipping tricyclo-DNA oligomers. Nat Med 21(3):270–275. https://doi.org/10.1038/nm.3765

    Article  CAS  PubMed  Google Scholar 

  28. Vaillend C, Chaussenot R (2017) Relationships linking emotional, motor, cognitive and GABAergic dysfunctions in dystrophin-deficient mdx mice. Hum Mol Genet 26(6):1041–1055. https://doi.org/10.1093/hmg/ddx013

    Article  CAS  PubMed  Google Scholar 

  29. Vaillend C, Rendon A, Misslin R, Ungerer A (1995) Influence of dystrophin-gene mutation on mdx mouse behavior. I. Retention deficits at long delays in spontaneous alternation and bar-pressing tasks. Behav Genet 25(6):569–579

    Article  CAS  Google Scholar 

  30. Manning J, Kulbida R, Rai P, Jensen L, Bouma J, Singh SP, O’Malley D, Yilmazer-Hanke D (2014) Amitriptyline is efficacious in ameliorating muscle inflammation and depressive symptoms in the mdx mouse model of Duchenne muscular dystrophy. Exp Physiol 99(10):1370–1386. https://doi.org/10.1113/expphysiol.2014.079475

    Article  CAS  PubMed  Google Scholar 

  31. Sekiguchi M, Zushida K, Yoshida M, Maekawa M, Kamichi S, Yoshida M, Sahara Y, Yuasa S, Takeda S, Wada K (2009) A deficit of brain dystrophin impairs specific amygdala GABAergic transmission and enhances defensive behaviour in mice. Brain 132(Pt 1):124–135. https://doi.org/10.1093/brain/awn253

    Article  PubMed  Google Scholar 

  32. Muntoni F, Mateddu A, Serra G (1991) Passive avoidance behaviour deficit in the mdx mouse. Neuromuscul Disord 1(2):121–123

    Article  CAS  Google Scholar 

  33. Vaillend C, Ungerer A (1999) Behavioral characterization of mdx3cv mice deficient in C-terminal dystrophins. Neuromuscul Disord 9(5):296–304

    Article  CAS  Google Scholar 

  34. Remmelink E, Aartsma-Rus A, Smit AB, Verhage M, Loos M, van Putten M (2016) Cognitive flexibility deficits in a mouse model for the absence of full-length dystrophin. Genes Brain Behav 15(6):558–567. https://doi.org/10.1111/gbb.12301

    Article  CAS  PubMed  Google Scholar 

  35. Vaillend C, Billard JM, Laroche S (2004) Impaired long-term spatial and recognition memory and enhanced CA1 hippocampal LTP in the dystrophin-deficient Dmd(mdx) mouse. Neurobiol Dis 17(1):10–20. https://doi.org/10.1016/j.nbd.2004.05.004

    Article  CAS  PubMed  Google Scholar 

  36. Miranda R, Sebrie C, Degrouard J, Gillet B, Jaillard D, Laroche S, Vaillend C (2009) Reorganization of inhibitory synapses and increased PSD length of perforated excitatory synapses in hippocampal area CA1 of dystrophin-deficient mdx mice. Cereb Cortex 19(4):876–888. https://doi.org/10.1093/cercor/bhn135

    Article  CAS  PubMed  Google Scholar 

  37. Miranda R, Nudel U, Laroche S, Vaillend C (2011) Altered presynaptic ultrastructure in excitatory hippocampal synapses of mice lacking dystrophins Dp427 or Dp71. Neurobiol Dis 43(1):134–141. https://doi.org/10.1016/j.nbd.2011.02.017

    Article  CAS  PubMed  Google Scholar 

  38. Del Tongo C, Carretta D, Fulgenzi G, Catini C, Minciacchi D (2009) Parvalbumin-positive GABAergic interneurons are increased in the dorsal hippocampus of the dystrophic mdx mouse. Acta Neuropathol 118(6):803–812. https://doi.org/10.1007/s00401-009-0567-3

    Article  CAS  PubMed  Google Scholar 

  39. Knuesel I, Mastrocola M, Zuellig RA, Bornhauser B, Schaub MC, Fritschy JM (1999) Short communication: altered synaptic clustering of GABAA receptors in mice lacking dystrophin (mdx mice). Eur J Neurosci 11(12):4457–4462

    Article  CAS  Google Scholar 

  40. Vaillend C, Perronnet C, Ros C, Gruszczynski C, Goyenvalle A, Laroche S, Danos O, Garcia L, Peltekian E (2010) Rescue of a dystrophin-like protein by exon skipping in vivo restores GABAA-receptor clustering in the hippocampus of the mdx mouse. Mol Ther 18(9):1683–1688. https://doi.org/10.1038/mt.2010.134

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Krasowska E, Zablocki K, Gorecki DC, Swinny JD (2014) Aberrant location of inhibitory synaptic marker proteins in the hippocampus of dystrophin-deficient mice: implications for cognitive impairment in duchenne muscular dystrophy. PLoS One 9(9):e108364. https://doi.org/10.1371/journal.pone.0108364

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Fuenzalida M, Espinoza C, Perez MA, Tapia-Rojas C, Cuitino L, Brandan E, Inestrosa NC (2016) Wnt signaling pathway improves central inhibitory synaptic transmission in a mouse model of Duchenne muscular dystrophy. Neurobiol Dis 86:109–120. https://doi.org/10.1016/j.nbd.2015.11.018

    Article  CAS  PubMed  Google Scholar 

  43. Fritschy JM, Panzanelli P, Tyagarajan SK (2012) Molecular and functional heterogeneity of GABAergic synapses. Cell Mol Life Sci 69(15):2485–2499. https://doi.org/10.1007/s00018-012-0926-4

    Article  CAS  PubMed  Google Scholar 

  44. Vaillend C, Billard JM (2002) Facilitated CA1 hippocampal synaptic plasticity in dystrophin-deficient mice: role for GABAA receptors? Hippocampus 12(6):713–717. https://doi.org/10.1002/hipo.10068

    Article  CAS  PubMed  Google Scholar 

  45. Kueh SL, Dempster J, Head SI, Morley JW (2011) Reduced postsynaptic GABAA receptor number and enhanced gaboxadol induced change in holding currents in Purkinje cells of the dystrophin-deficient mdx mouse. Neurobiol Dis 43(3):558–564. https://doi.org/10.1016/j.nbd.2011.05.002

    Article  CAS  PubMed  Google Scholar 

  46. de Brouwer AP, Nabuurs SB, Verhaart IE, Oudakker AR, Hordijk R, Yntema HG, Hordijk-Hos JM, Voesenek K, de Vries BB, van Essen T, Chen W, Hu H, Chelly J, den Dunnen JT, Kalscheuer VM, Aartsma-Rus AM, Hamel BC, van Bokhoven H, Kleefstra T (2014) A 3-base pair deletion, c.9711_9713del, in DMD results in intellectual disability without muscular dystrophy. Eur J Hum Genet 22(4):480–485. https://doi.org/10.1038/ejhg.2013.169

    Article  CAS  PubMed  Google Scholar 

  47. Daoud F, Candelario-Martinez A, Billard JM, Avital A, Khelfaoui M, Rozenvald Y, Guegan M, Mornet D, Jaillard D, Nudel U, Chelly J, Martinez-Rojas D, Laroche S, Yaffe D, Vaillend C (2009) Role of mental retardation-associated dystrophin-gene product Dp71 in excitatory synapse organization, synaptic plasticity and behavioral functions. PLoS One 4(8):e6574. https://doi.org/10.1371/journal.pone.0006574

    Article  CAS  PubMed Central  Google Scholar 

  48. Tadayoni R, Rendon A, Soria-Jasso LE, Cisneros B (2012) Dystrophin Dp71: the smallest but multifunctional product of the Duchenne muscular dystrophy gene. Mol Neurobiol 45(1):43–60. https://doi.org/10.1007/s12035-011-8218-9

    Article  CAS  PubMed  Google Scholar 

  49. Hendriksen RG, Hoogland G, Schipper S, Hendriksen JG, Vles JS, Aalbers MW (2015) A possible role of dystrophin in neuronal excitability: a review of the current literature. Neurosci Biobehav Rev 51:255–262. https://doi.org/10.1016/j.neubiorev.2015.01.023

    Article  CAS  PubMed  Google Scholar 

  50. Connors NC, Adams ME, Froehner SC, Kofuji P (2004) The potassium channel Kir4.1 associates with the dystrophin-glycoprotein complex via alpha-syntrophin in glia. J Biol Chem 279(27):28387–28392. https://doi.org/10.1074/jbc.M402604200

    Article  CAS  PubMed  Google Scholar 

  51. Haenggi T, Fritschy JM (2006) Role of dystrophin and utrophin for assembly and function of the dystrophin glycoprotein complex in non-muscle tissue. Cell Mol Life Sci 63(14):1614–1631. https://doi.org/10.1007/s00018-005-5461-0

    Article  CAS  PubMed  Google Scholar 

  52. Nicchia GP, Rossi A, Nudel U, Svelto M, Frigeri A (2008) Dystrophin-dependent and -independent AQP4 pools are expressed in the mouse brain. Glia 56(8):869–876. https://doi.org/10.1002/glia.20661

    Article  PubMed  Google Scholar 

  53. Enger R, Gundersen GA, Haj-Yasein NN, Eilert-Olsen M, Thoren AE, Vindedal GF, Petersen PH, Skare O, Nedergaard M, Ottersen OP, Nagelhus EA (2012) Molecular scaffolds underpinning macroglial polarization: an analysis of retinal Muller cells and brain astrocytes in mouse. Glia 60(12):2018–2026. https://doi.org/10.1002/glia.22416

    Article  PubMed  PubMed Central  Google Scholar 

  54. Fort PE, Sene A, Pannicke T, Roux MJ, Forster V, Mornet D, Nudel U, Yaffe D, Reichenbach A, Sahel JA, Rendon A (2008) Kir4.1 and AQP4 associate with Dp71- and utrophin-DAPs complexes in specific and defined microdomains of Muller retinal glial cell membrane. Glia 56(6):597–610. https://doi.org/10.1002/glia.20633

    Article  PubMed  Google Scholar 

  55. Giocanti-Auregan A, Vacca O, Benard R, Cao S, Siqueiros L, Montanez C, Paques M, Sahel JA, Sennlaub F, Guillonneau X, Rendon A, Tadayoni R (2016) Altered astrocyte morphology and vascular development in dystrophin-Dp71-null mice. Glia 64(5):716–729. https://doi.org/10.1002/glia.22956

    Article  PubMed  Google Scholar 

  56. Sene A, Tadayoni R, Pannicke T, Wurm A, El Mathari B, Benard R, Roux MJ, Yaffe D, Mornet D, Reichenbach A, Sahel JA, Rendon A (2009) Functional implication of Dp71 in osmoregulation and vascular permeability of the retina. PLoS One 4(10):e7329. https://doi.org/10.1371/journal.pone.0007329

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Louie K, Apte RS, Mori K, Gehlbach P (2004) Severe proliferative retinopathy in a patient with advanced muscular dystrophy. Br J Ophthalmol 88(12):1604–1605. https://doi.org/10.1136/bjo.2004.046615

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Fagan XJ, Levy J, Al-Qureshi S, Harper CA (2012) Proliferative retinopathy in Duchenne muscular dystrophy and its response to bevacizumab. Clin Exp Ophthalmol 40(9):906–907. https://doi.org/10.1111/j.1442-9071.2012.02822.x

    Article  PubMed  Google Scholar 

  59. Hahn P, Lin P, Fekrat S (2013) Ultra-widefield imaging of Duchenne muscular dystrophy-associated proliferative retinal vasculopathy improved with panretinal laser photocoagulation alone. Ophthalmic Surg Lasers Imaging Retina 44(3):293–295. https://doi.org/10.3928/23258160-20130503-17

    Article  PubMed  Google Scholar 

  60. El Mathari B, Sene A, Charles-Messance H, Vacca O, Guillonneau X, Grepin C, Sennlaub F, Sahel JA, Rendon A, Tadayoni R (2015) Dystrophin Dp71 gene deletion induces retinal vascular inflammation and capillary degeneration. Hum Mol Genet 24(14):3939–3947. https://doi.org/10.1093/hmg/ddv132

    Article  CAS  PubMed  Google Scholar 

  61. Cia D, Simonutti M, Fort PE, Doly M, Rendon A (2014) Slight alteration of the electroretinogram in mice lacking dystrophin dp71. Ophthalmic Res 51(4):196–203. https://doi.org/10.1159/000357272

    Article  CAS  PubMed  Google Scholar 

  62. Goyenvalle A, Vulin A, Fougerousse F, Leturcq F, Kaplan JC, Garcia L, Danos O (2004) Rescue of dystrophic muscle through U7 snRNA-mediated exon skipping. Science 306(5702):1796–1799. https://doi.org/10.1126/science.1104297

    Article  CAS  PubMed  Google Scholar 

  63. Dallerac G, Perronnet C, Chagneau C, Leblanc-Veyrac P, Samson-Desvignes N, Peltekian E, Danos O, Garcia L, Laroche S, Billard JM, Vaillend C (2011) Rescue of a dystrophin-like protein by exon skipping normalizes synaptic plasticity in the hippocampus of the mdx mouse. Neurobiol Dis 43(3):635–641. https://doi.org/10.1016/j.nbd.2011.05.012

    Article  CAS  PubMed  Google Scholar 

  64. Casaca-Carreira J, Temel Y, Larrakoetxea I, Jahanshahi A (2017) Distribution and penetration of intracerebroventricularly administered 2′OMePS oligonucleotide in the mouse brain. Nucleic Acid Ther 27(1):4–10. https://doi.org/10.1089/nat.2016.0642

    Article  CAS  PubMed  Google Scholar 

  65. Klimczak RR, Koerber JT, Dalkara D, Flannery JG, Schaffer DV (2009) A novel adeno-associated viral variant for efficient and selective intravitreal transduction of rat Muller cells. PLoS One 4(10):e7467. https://doi.org/10.1371/journal.pone.0007467

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Vacca O, Darche M, Schaffer DV, Flannery JG, Sahel JA, Rendon A, Dalkara D (2014) AAV-mediated gene delivery in Dp71-null mouse model with compromised barriers. Glia 62(3):468–476. https://doi.org/10.1002/glia.22617

    Article  PubMed  Google Scholar 

  67. Vacca O, Charles-Messance H, El Mathari B, Sene A, Barbe P, Fouquet S, Aragon J, Darche M, Giocanti-Auregan A, Paques M, Sahel JA, Tadayoni R, Montanez C, Dalkara D, Rendon A (2016) AAV-mediated gene therapy in Dystrophin-Dp71 deficient mouse leads to blood-retinal barrier restoration and oedema reabsorption. Hum Mol Genet 25(14):3070–3079. https://doi.org/10.1093/hmg/ddw159

    Article  CAS  PubMed  Google Scholar 

  68. Ehninger D, Li W, Fox K, Stryker MP, Silva AJ (2008) Reversing neurodevelopmental disorders in adults. Neuron 60(6):950–960. https://doi.org/10.1016/j.neuron.2008.12.007

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Vaillend C, Poirier R, Laroche S (2008) Genes, plasticity and mental retardation. Behav Brain Res 192(1):88–105. https://doi.org/10.1016/j.bbr.2008.01.009

    Article  CAS  PubMed  Google Scholar 

  70. Septien L, Gras P, Borsotti JP, Giroud M, Nivelon JL, Dumas R (1991) Mental development in Duchenne muscular dystrophy. Correlation of data of the brain scanner. Pediatrie 46(12):817–819

    CAS  PubMed  Google Scholar 

  71. Anderson JL, Head SI, Rae C, Morley JW (2002) Brain function in Duchenne muscular dystrophy. Brain 125(Pt 1):4–13

    Article  CAS  Google Scholar 

  72. Dashkoff J, Lerner EP, Truong N, Klickstein JA, Fan Z, Mu D, Maguire CA, Hyman BT, Hudry E (2016) Tailored transgene expression to specific cell types in the central nervous system after peripheral injection with AAV9. Mol Ther Methods Clin Dev 3:16081. https://doi.org/10.1038/mtm.2016.81

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Nico B, Frigeri A, Nicchia GP, Corsi P, Ribatti D, Quondamatteo F, Herken R, Girolamo F, Marzullo A, Svelto M, Roncali L (2003) Severe alterations of endothelial and glial cells in the blood-brain barrier of dystrophic mdx mice. Glia 42(3):235–251. https://doi.org/10.1002/glia.10216

    Article  PubMed  Google Scholar 

  74. Nico B, Paola Nicchia G, Frigeri A, Corsi P, Mangieri D, Ribatti D, Svelto M, Roncali L (2004) Altered blood-brain barrier development in dystrophic MDX mice. Neuroscience 125(4):921–935. https://doi.org/10.1016/j.neuroscience.2004.02.008

    Article  CAS  PubMed  Google Scholar 

  75. Lien CF, Mohanta SK, Frontczak-Baniewicz M, Swinny JD, Zablocka B, Gorecki DC (2012) Absence of glial alpha-dystrobrevin causes abnormalities of the blood-brain barrier and progressive brain edema. J Biol Chem 287(49):41374–41385. https://doi.org/10.1074/jbc.M112.400044

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Menezes MJ, McClenahan FK, Leiton CV, Aranmolate A, Shan X, Colognato H (2014) The extracellular matrix protein laminin alpha2 regulates the maturation and function of the blood-brain barrier. J Neurosci 34(46):15260–15280. https://doi.org/10.1523/JNEUROSCI.3678-13.2014

    Article  CAS  PubMed  Google Scholar 

  77. Dalkara D, Kolstad KD, Caporale N, Visel M, Klimczak RR, Schaffer DV, Flannery JG (2009) Inner limiting membrane barriers to AAV-mediated retinal transduction from the vitreous. Mol Ther 17(12):2096–2102. https://doi.org/10.1038/mt.2009.181

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Dalkara D, Sahel JA (2014) Gene therapy for inherited retinal degenerations. C R Biol 337(3):185–192. https://doi.org/10.1016/j.crvi.2014.01.002

    Article  PubMed  Google Scholar 

  79. Aschauer DF, Kreuz S, Rumpel S (2013) Analysis of transduction efficiency, tropism and axonal transport of AAV serotypes 1, 2, 5, 6, 8 and 9 in the mouse brain. PLoS One 8(9):e76310. https://doi.org/10.1371/journal.pone.0076310

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Jackson KL, Dayton RD, Deverman BE, Klein RL (2016) Better targeting, better efficiency for wide-scale neuronal transduction with the synapsin promoter and AAV-PHP.B. Front Mol Neurosci 9:116. https://doi.org/10.3389/fnmol.2016.00116

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Liu Y, Siriwon N, Rohrs JA, Wang P (2015) Generation of targeted adeno-associated virus (AAV) vectors for human gene therapy. Curr Pharm Des 21(22):3248–3256

    Article  CAS  Google Scholar 

  82. Buning H, Huber A, Zhang L, Meumann N, Hacker U (2015) Engineering the AAV capsid to optimize vector-host-interactions. Curr Opin Pharmacol 24:94–104. https://doi.org/10.1016/j.coph.2015.08.002

    Article  CAS  PubMed  Google Scholar 

  83. Xia H, Mao Q, Eliason SL, Harper SQ, Martins IH, Orr HT, Paulson HL, Yang L, Kotin RM, Davidson BL (2004) RNAi suppresses polyglutamine-induced neurodegeneration in a model of spinocerebellar ataxia. Nat Med 10(8):816–820. https://doi.org/10.1038/nm1076

    Article  PubMed  Google Scholar 

  84. van Putten M, Hulsker M, Nadarajah VD, van Heiningen SH, van Huizen E, van Iterson M, Admiraal P, Messemaker T, den Dunnen JT, t Hoen PA, Aartsma-Rus A (2012) The effects of low levels of dystrophin on mouse muscle function and pathology. PLoS One 7(2):e31937. https://doi.org/10.1371/journal.pone.0031937

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Wu B, Lu P, Cloer C, Shaban M, Grewal S, Milazi S, Shah SN, Moulton HM, Lu QL (2012) Long-term rescue of dystrophin expression and improvement in muscle pathology and function in dystrophic mdx mice by peptide-conjugated morpholino. Am J Pathol 181(2):392–400. https://doi.org/10.1016/j.ajpath.2012.04.006

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Le Guiner C, Montus M, Servais L, Cherel Y, Francois V, Thibaud JL, Wary C, Matot B, Larcher T, Guigand L, Dutilleul M, Domenger C, Allais M, Beuvin M, Moraux A, Le Duff J, Devaux M, Jaulin N, Guilbaud M, Latournerie V, Veron P, Boutin S, Leborgne C, Desgue D, Deschamps JY, Moullec S, Fromes Y, Vulin A, Smith RH, Laroudie N, Barnay-Toutain F, Riviere C, Bucher S, Le TH, Delaunay N, Gasmi M, Kotin RM, Bonne G, Adjali O, Masurier C, Hogrel JY, Carlier P, Moullier P, Voit T (2014) Forelimb treatment in a large cohort of dystrophic dogs supports delivery of a recombinant AAV for exon skipping in Duchenne patients. Mol Ther 22(11):1923–1935. https://doi.org/10.1038/mt.2014.151

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Kuzmanovic M, Dudley VJ, Sarthy VP (2003) GFAP promoter drives Muller cell-specific expression in transgenic mice. Invest Ophthalmol Vis Sci 44(8):3606–3613

    Article  Google Scholar 

Download references

Acknowledgments

This review was written jointly by the three authors. AFM-Telethon (Association Française contre les Myopathies, France) and Agence Nationale de la Recherche (ANR, France) supported the recent research referred from the Vaillend laboratory. Faouzi Zarrouki was a recipient of a PhD fellowship from the Ministère de l’Enseignement Supérieur et de la Recherche (France). Ophélie Vacca received a postdoctoral fellowship from the AFM.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Cyrille Vaillend .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Vaillend, C., Zarrouki, F., Vacca, O. (2019). Gene Therapy for Central Nervous System in Duchenne Muscular Dystrophy. In: Duan, D., Mendell, J. (eds) Muscle Gene Therapy. Springer, Cham. https://doi.org/10.1007/978-3-030-03095-7_24

Download citation

Publish with us

Policies and ethics