4,785
Views
30
CrossRef citations to date
0
Altmetric
Review

The potential of utrophin modulators for the treatment of Duchenne muscular dystrophy

, &
Pages 179-192 | Received 15 Dec 2017, Accepted 06 Feb 2018, Published online: 15 Feb 2018

References

  • Guiraud S, Aartsma-Rus A, Vieira NM, et al. The pathogenesis and therapy of muscular dystrophies. Annu Rev Genomics Hum Genet. 2015;16:281–308.
  • Bladen CL, Salgado D, Monges S, et al. The TREAT-NMD DMD Global Database: analysis of more than 7,000 Duchenne muscular dystrophy mutations. Hum Mutat. 2015 Apr;36(4):395–402.
  • Monaco AP, Neve RL, Colletti-Feener C, et al. Isolation of candidate cDNAs for portions of the Duchenne muscular dystrophy gene. Nature. 1986 Oct 16–22;323(6089):646–650.
  • Bach JR, O’Brien J, Krotenberg R, et al. Management of end stage respiratory failure in Duchenne muscular dystrophy. Muscle Nerve. 1987 Feb;10(2):177–182.
  • Hoffman EP, Brown RH Jr., Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell. 1987 Dec 24;51(6):919–928.
  • Blake DJ, Weir A, Newey SE, et al. Function and genetics of dystrophin and dystrophin-related proteins in muscle. Physiol Rev. 2002 Apr;82(2):291–329.
  • England SB, Nicholson LVB, Johnson MA, et al. Very mild muscular dystrophy associated with the deletion of 46% of dystrophin. Nature. 1990;343(6254):180–182.
  • Emery AE. Dystrophin function. Lancet. 1990 May 26;335(8700):1289.
  • Guiraud S, Davies KE. Pharmacological advances for treatment in Duchenne muscular dystrophy. Curr Opin Pharmacol. 2017 May;6(34):36–48.
  • Guiraud S, Chen H, Burns DT, et al. Advances in genetic therapeutic strategies for Duchenne muscular dystrophy. Exp Physiol. 2015 Dec;100(12):1458–1467.
  • Khurana TS, Davies KE. Pharmacological strategies for muscular dystrophy. Nat Rev Drug Discov. 2003 May;2(5):379–390.
  • Bushby K, Finkel R, Wong B, et al. Ataluren treatment of patients with nonsense mutation dystrophinopathy. Muscle Nerve. 2014 Oct;50(4):477–487.
  • Niks EH, Aartsma-Rus A. Exon skipping: a first in class strategy for Duchenne muscular dystrophy. Expert Opin Biol Ther. 2017 Feb;17(2):225–236.
  • Robinson-Hamm JN, Gersbach CA. Gene therapies that restore dystrophin expression for the treatment of Duchenne muscular dystrophy. Hum Genet. 2016 Sep;135(9):1029–1040.
  • Love DR, Hill DF, Dickson G, et al. An autosomal transcript in skeletal muscle with homology to dystrophin. Nature. 1989 May 4;339(6219):55–58.
  • Pearce M, Blake DJ, Tinsley JM, et al. The utrophin and dystrophin genes share similarities in genomic structure. Hum Mol Genet. 1993 Nov;2(11):1765–1772.
  • Tome FM, Matsumura K, Chevallay M, et al. Expression of dystrophin-associated glycoproteins during human fetal muscle development: a preliminary immunocytochemical study. Neuromuscular Disorders. 1994 Jul;4(4):343–348.
  • Muntoni F, Torelli S, Ferlini A. Dystrophin and mutations: one gene, several proteins, multiple phenotypes. Lancet Neurol. 2003 Dec;2(12):731–740.
  • Clerk A, Morris GE, Dubowitz V, et al. Dystrophin-related protein, utrophin, in normal and dystrophic human fetal skeletal muscle. Histochem J. 1993 Aug;25(8):554–561.
  • 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 Biol Chem. 2002 Nov 22;277(47):45285–45290.
  • Burton EA, Tinsley JM, Holzfeind PJ, et al. A second promoter provides an alternative target for therapeutic up-regulation of utrophin in Duchenne muscular dystrophy. Proc Natl Acad Sci USA. 1999 Nov 23;96(24):14025–14030.
  • Dennis CL, Tinsley JM, Deconinck AE, et al. Molecular and functional analysis of the utrophin promoter. Nucleic Acids Res. 1996 May 1;24(9):1646–1652.
  • Wilson J, Putt W, Jimenez C, et al. Up71 and up140, two novel transcripts of utrophin that are homologues of short forms of dystrophin. Hum Mol Genet. 1999 Jul;8(7):1271–1278.
  • Sadoulet-Puccio HM, Kunkel LM. Dystrophin and its isoforms. Brain Pathology. 1996 Jan;6(1):25–35.
  • Blake DJ, Schofield JN, Zuellig RA, et al. G-utrophin, the autosomal homologue of dystrophin Dp116, is expressed in sensory ganglia and brain. Proc Natl Acad Sci USA. 1995 Apr 25;92(9):3697–3701.
  • Tinsley JM, Blake DJ, Roche A, et al. Primary structure of dystrophin-related protein. Nature. 1992 Dec 10;360(6404):591–593.
  • Ervasti JM. Dystrophin, its interactions with other proteins, and implications for muscular dystrophy. Biochim Biophys Acta. 2007 Feb;1772(2):108–117.
  • Rybakova IN, Humston JL, Sonnemann KJ, et al. Dystrophin and utrophin bind actin through distinct modes of contact. J Biol Chem. 2006 Apr 14;281(15):9996–10001.
  • Belanto JJ, Mader TL, Eckhoff MD, et al. Microtubule binding distinguishes dystrophin from utrophin. Proc Natl Acad Sci USA. 2014 Apr 15;111(15):5723–5728.
  • Li D, Bareja A, Judge L, et al. Sarcolemmal nNOS anchoring reveals a qualitative difference between dystrophin and utrophin. J Cell Sci. 2010 Jun 15;123(Pt 12):2008–2013.
  • van den Bergen JC, Wokke BH, Hulsker MA, et al. Studying the role of dystrophin-associated proteins in influencing Becker muscular dystrophy disease severity. Neuromuscular Disorders. 2015 Mar;25(3):231–237.
  • Bulfield G, Siller WG, Wight PA, et al. X chromosome-linked muscular dystrophy (mdx) in the mouse. Proc Natl Acad Sci USA. 1984 Feb;81(4):1189–1192.
  • Schofield JN, Gorecki DC, Blake DJ, et al. Dystroglycan mRNA expression during normal and mdx mouse embryogenesis: a comparison with utrophin and the apo-dystrophins. Dev Dyn. 1995 Oct;204(2):178–185.
  • Helliwell TR, Man NT, Morris GE, et al. The dystrophin-related protein, utrophin, is expressed on the sarcolemma of regenerating human skeletal muscle fibres in dystrophies and inflammatory myopathies. Neuromuscular Disorders. 1992;2(3):177–184.
  • Arechavala-Gomeza V, Kinali M, Feng L, et al. Immunohistological intensity measurements as a tool to assess sarcolemma-associated protein expression. Neuropathol Appl Neurobiol. 2010 Jun;36(4):265–274.
  • Kleopa KA, Drousiotou A, Mavrikiou E, et al. Naturally occurring utrophin correlates with disease severity in Duchenne muscular dystrophy. Hum Mol Genet. 2006 May 15;15(10):1623–1628.
  • Vainzof M, Passos-Bueno MR, Man N, et al. Absence of correlation between utrophin localization and quantity and the clinical severity in Duchenne/Becker dystrophies. Am J Med Genet. 1995 Sep 25;58(4):305–309.
  • van den Bergen JC. Schade van Westrum SM, Dekker L, et al. Clinical characterisation of Becker muscular dystrophy patients predicts favourable outcome in exon-skipping therapy. J Neurol Neurosurg Psychiatry. 2014 Jan;85(1):92–98.
  • Janghra N, Morgan JE, Sewry CA, et al. Correlation of utrophin levels with the dystrophin protein complex and muscle fibre regeneration in duchenne and becker muscular dystrophy muscle biopsies. PloS One. 2016;11(3):e0150818.
  • Wilson K, Faelan C, Patterson-Kane JC, et al. Duchenne and becker muscular dystrophies: a review of animal models, clinical end points, and biomarker quantification. Toxicol Pathol. 2017 Oct;45(7):961–976.
  • Deconinck AE, Rafael JA, Skinner JA, et al. Utrophin-dystrophin-deficient mice as a model for Duchenne muscular dystrophy. Cell. 1997 Aug 22;90(4):717–727.
  • Chevron MP, Echenne B, Demaille J. Absence of dystrophin and utrophin in a boy with severe muscular dystrophy. N Engl J Med. 1994 Oct 27;331(17):1162–1163.
  • Deconinck N, Tinsley J, De Backer F, et al. Expression of truncated utrophin leads to major functional improvements in dystrophin-deficient muscles of mice. Nat Med. 1997 Nov;3(11):1216–1221.
  • Tinsley J, Deconinck N, Fisher R, et al. Expression of full-length utrophin prevents muscular dystrophy in mdx mice. Nat Med. 1998 Dec;4(12):1441–1444.
  • Guiraud S, Edwards B, Squire SE, et al. Identification of serum protein biomarkers for utrophin based DMD therapy. Sci Rep. 2017 Mar;2(7):43697.
  • Kennedy TL, Moir L, Hemming S, et al. Utrophin influences mitochondrial pathology and oxidative stress in dystrophic muscle. Skeletal Muscle. 2017 Oct 24;7(1):22.
  • Fisher R, Tinsley JM, Phelps SR, et al. Non-toxic ubiquitous over-expression of utrophin in the mdx mouse. Neuromuscul Disord. 2001;11(8):713–721.
  • Squire S, Raymackers JM, Vandebrouck C, et al. Prevention of pathology in mdx mice by expression of utrophin: analysis using an inducible transgenic expression system. Hum Mol Genet. 2002 Dec 15;11(26):3333–3344.
  • Chancellor DR, Davies KE, De Moor O, et al. Discovery of 2-arylbenzoxazoles as upregulators of utrophin production for the treatment of Duchenne muscular dystrophy. J Med Chem. 2011 May 12;54(9):3241–3250.
  • Tinsley JM, Fairclough RJ, Storer R, et al. Daily treatment with SMTC1100, a novel small molecule utrophin upregulator, dramatically reduces the dystrophic symptoms in the mdx mouse. PloS One. 2011;6(5):e19189.
  • Guiraud S, Squire SE, Edwards B, et al. Second-generation compound for the modulation of utrophin in the therapy of DMD. Hum Mol Genet. 2015 Aug 1; 24(15):4212–4224.
  • Moorwood C, Lozynska O, Suri N, et al. Drug discovery for Duchenne muscular dystrophy via utrophin promoter activation screening. PloS One. 2011;6(10):e26169.
  • Wynne G, Uorinen A, Emer E, et al. Discovery of small molecule utrophin modulators for the therapy of Duchenne muscular dystrophy. World Muscle Society Conference, Saint Malo France. 2017.
  • Gramolini AO, Angus LM, Schaeffer L, et al. Induction of utrophin gene expression by heregulin in skeletal muscle cells: role of the N-box motif and GA binding protein. Proc Natl Acad Sci USA. 1999 Mar 16;96(6):3223–3227.
  • Basu U, Gyrd-Hansen M, Baby SM, et al. Heregulin-induced epigenetic regulation of the utrophin-A promoter. FEBS Lett. 2007 Sep 4;581(22):4153–4158.
  • Krag TO, Bogdanovich S, Jensen CJ, et al. Heregulin ameliorates the dystrophic phenotype in mdx mice. Proc Natl Acad Sci USA. 2004 Sep 21;101(38):13856–13860.
  • Fujimori K, Itoh Y, Yamamoto K, et al. Interleukin 6 induces overexpression of the sarcolemmal utrophin in neonatal mdx skeletal muscle. Hum Gene Ther. 2002 Mar 1;13(4):509–518.
  • Gauthier-Rouviere C, Bonet-Kerrache A. RhoA leads to up-regulation and relocalization of utrophin in muscle fibers. Biochem Biophys Res Commun. 2009 Jul 3;384(3):322–328.
  • Chaubourt E, Fossier P, Baux G, et al. Nitric oxide and l-arginine cause an accumulation of utrophin at the sarcolemma: a possible compensation for dystrophin loss in Duchenne muscular dystrophy. Neurobiol Dis. 1999 Dec;6(6):499–507.
  • Banks GB, Combs AC, Odom GL, et al. Muscle structure influences utrophin expression in mdx mice. PLoS Genet. 2014 Jun;10(6):e1004431.
  • Weir AP, Morgan JE, Davies KE. A-utrophin up-regulation in mdx skeletal muscle is independent of regeneration. Neuromuscular Disorders. 2004 Jan;14(1):19–23.
  • Chakkalakal JV, Stocksley MA, Harrison MA, et al. Expression of utrophin A mRNA correlates with the oxidative capacity of skeletal muscle fiber types and is regulated by calcineurin/NFAT signaling. Proc Natl Acad Sci USA. 2003 Jun 24;100(13):7791–7796.
  • Consolino CM, Brooks SV. Susceptibility to sarcomere injury induced by single stretches of maximally activated muscles of mdx mice. J Appl Physiol. 2004 Feb;96(2):633–638.
  • Miura P, Chakkalakal JV, Boudreault L, et al. Pharmacological activation of PPARbeta/delta stimulates utrophin A expression in skeletal muscle fibers and restores sarcolemmal integrity in mature mdx mice. Hum Mol Genet. 2009 Dec 1;18(23):4640–4649.
  • Ljubicic V, Miura P, Burt M, et al. Chronic AMPK activation evokes the slow, oxidative myogenic program and triggers beneficial adaptations in mdx mouse skeletal muscle. Hum Mol Genet. 2011 Sep 01;20(17):3478–3493.
  • Ljubicic V, Jasmin BJ. Metformin increases peroxisome proliferator-activated receptor gamma Co-activator-1alpha and utrophin a expression in dystrophic skeletal muscle. Muscle Nerve. 2015 Jul;52(1):139–142.
  • Ballmann C, Hollinger K, Selsby JT, et al. Histological and biochemical outcomes of cardiac pathology in mdx mice with dietary quercetin enrichment. Exp Physiol. 2015 Jan;100(1):12–22.
  • Gordon BS, Delgado Diaz DC, Kostek MC. Resveratrol decreases inflammation and increases utrophin gene expression in the mdx mouse model of Duchenne muscular dystrophy. Clin Nutrition. 2013 Feb;32(1):104–111.
  • 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. Hum Mol Genet. 2016 Jan 01;25(1):24–43.
  • D’Arcy CE, Feeney SJ, McLean CA, et al. Identification of FHL1 as a therapeutic target for Duchenne muscular dystrophy. Hum Mol Genet. 2014 Feb 1;23(3):618–636.
  • Koo JH, Kim TH, Park SY, et al. Galpha13 ablation reprograms myofibers to oxidative phenotype and enhances whole-body metabolism. J Clin Invest. 2017 Oct 2;127(10):3845–3860.
  • Stark DA, Coffey NJ, Pancoast HR, et al. Ephrin-A3 promotes and maintains slow muscle fiber identity during postnatal development and reinnervation. J Cell Biol. 2015 Dec 7;211(5):1077–1091.
  • Sonnemann KJ, Heun-Johnson H, Turner AJ, et al. Functional substitution by TAT-utrophin in dystrophin-deficient mice. PLoS Med. 2009 May 26;6(5):e1000083.
  • Call JA, Ervasti JM, Lowe DA. TAT-muUtrophin mitigates the pathophysiology of dystrophin and utrophin double-knockout mice. J Appl Physiol. 2011 Jul;111(1):200–205.
  • Courdier-Fruh I, Briguet A. Utrophin is a calpain substrate in muscle cells. Muscle Nerve. 2006 Jun;33(6):753–759.
  • Badalamente MA, Stracher A. Delay of muscle degeneration and necrosis in mdx mice by calpain inhibition. Muscle Nerve. 2000 Jan;23(1):106–111.
  • Childers MK, Bogan JR, Bogan DJ, et al. Chronic administration of a leupeptin-derived calpain inhibitor fails to ameliorate severe muscle pathology in a canine model of duchenne muscular dystrophy. Front Pharmacol. 2011;2:89.
  • Amenta AR, Yilmaz A, Bogdanovich S, et al. Biglycan recruits utrophin to the sarcolemma and counters dystrophic pathology in mdx mice. Proc Natl Acad Sci USA. 2011 Jan 11;108(2):762–767.
  • Chamberlain JR, Chamberlain JS. Progress toward gene therapy for duchenne muscular dystrophy. Mol Therapy. 2017 May 3;25(5):1125–1131.
  • Gilbert R, Nalbantoglu J, Petrof BJ, et al. Adenovirus-mediated utrophin gene transfer mitigates the dystrophic phenotype of mdx mouse muscles. Hum Gene Ther. 1999 May 20;10(8):1299–1310.
  • Odom GL, Gregorevic P, Allen JM, et al. Microutrophin delivery through rAAV6 increases lifespan and improves muscle function in dystrophic dystrophin/utrophin-deficient mice. Mol Therapy. 2008 Sep;16(9):1539–1545.
  • Cerletti M, Negri T, Cozzi F, et al. Dystrophic phenotype of canine X-linked muscular dystrophy is mitigated by adenovirus-mediated utrophin gene transfer. Gene Ther. 2003 May;10(9):750–757.
  • Ebihara S, Guibinga GH, Gilbert R, et al. Differential effects of dystrophin and utrophin gene transfer in immunocompetent muscular dystrophy (mdx) mice. Physiol Genomics. 2000 Sep 8;3(3):133–144.
  • Zhao J, Kodippili K, Yue Y, et al. Dystrophin contains multiple independent membrane-binding domains. Hum Mol Genet. 2016 Sep 1;25(17):3647–3653.
  • Biosciences S. Letter to duchenne community about solid’s first clinical trial. 2017. Available from: https://solidbiocom/content/letter-duchennecommunity-about-solid-s-first-clinical-trial
  • Pfizer. A study to evaluate the safety and tolerability of PF-06939926 gene therapy in duchenne muscular dystrophy. 2017. Available from: https://clinicaltrialsgov/ct2/show/NCT03362502
  • Lai Y, Zhao J, Yue Y, et al. A novel rationally designed AAV micro-utrophin vector recruits nNOS to the sarcolemma. 15th Annual Meeting of the American-Society-of-Gene-and-Cell-Therapy 2012;20.
  • Onori A, Pisani C, Strimpakos G, et al. UtroUp is a novel six zinc finger artificial transcription factor that recognises 18 base pairs of the utrophin promoter and efficiently drives utrophin upregulation. BMC Mol Biol. 2013 Jan 30;14:3.
  • 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 Cell Physiol. 2014 Sep;229(9):1283–1291.
  • Durko M, Allen C, Nalbantoglu J, et al. CT-GalNAc transferase overexpression in adult mice is associated with extrasynaptic utrophin in skeletal muscle fibres. J Muscle Res Cell Motil. 2010 Sep;31(3):181–193.
  • Gibbs EM, Marshall JL, Ma E, et al. High levels of sarcospan are well tolerated and act as a sarcolemmal stabilizer to address skeletal muscle and pulmonary dysfunction in DMD. Hum Mol Genet. 2016 Oct 24.
  • Xu R, Camboni M, Martin PT. Postnatal overexpression of the CT GalNAc transferase inhibits muscular dystrophy in mdx mice without altering muscle growth or neuromuscular development: evidence for a utrophin-independent mechanism. Neuromuscular Disord. 2007 Mar;17(3):209–220.
  • Basu U, Lozynska O, Moorwood C, et al. Translational regulation of utrophin by miRNAs. PloS One. 2011;6(12):e29376.
  • Mishra MK, Loro E, Sengupta K, et al. Functional improvement of dystrophic muscle by repression of utrophin: let-7c interaction. PloS One. 2017;12(10):e0182676.
  • Amirouche A, Jahnke VE, Lunde JA, et al. Muscle-specific microRNA-206 targets multiple components in dystrophic skeletal muscle representing beneficial adaptations. Am J Physiol Cell Physiol. 2017 Mar 1;312(3):C209–C21.
  • Tjondrokoesoemo A, Schips T, Kanisicak O, et al. Genetic overexpression of Serpina3n attenuates muscular dystrophy in mice. Hum Mol Genet. 2016 Mar 15;25(6):1192–1202.
  • Vanhoutte D, Schips TG, Kwong JQ, et al. Thrombospondin expression in myofibers stabilizes muscle membranes. eLife. 2016 Sep 26;5.
  • 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 Clin Pharmacol. 2015 Jun;55(6):698–707.
  • 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. PloS One. 2016;11(4):e0152840.
  • Summit therapeutic plc. Ezutromid significantly reduced muscle damage in DMD patients ini 24-week interim data from Summit’s PhaseOut DMD clinical trial. 2018. Available from: http://otpinvestiscom/clients/uk/summit_corporation_plc/rns/regulatory-storyaspx?cid=1575&newsid=970514.
  • Forbes SC, Walter GA, Rooney WD, et al. Skeletal muscles of ambulant children with Duchenne muscular dystrophy: validation of multicenter study of evaluation with MR imaging and MR spectroscopy. Radiology. 2013 Oct;269(1):198–207.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.