2,839
Views
24
CrossRef citations to date
0
Altmetric
Review

RNA processing in skeletal muscle biology and disease

ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 1-20 | Received 17 Oct 2018, Accepted 06 Dec 2018, Published online: 15 Jan 2019

References

  • Frontera WR, Ochala J. Skeletal muscle: a brief review of structure and function. Calcif Tissue Int. 2015;96:183–195.
  • Janssen I, Heymsfield SB, Wang Z, et al. Skeletal muscle mass and distribution in 468 men and women aged 18–88 yr. J Appl Physiol. 2000;89:81–88.
  • Schiaffino S, Reggiani C. Fiber types in mammalian skeletal muscles. Physiol Rev. 2011;91:1447–1531.
  • Zierath JR, Hawley JA. Skeletal muscle fiber type: influence on contractile and metabolic properties. PLoS Biol. 2004;2:e348.
  • Trovato F, Imbesi R, Conway N, et al. Morphological and functional aspects of human skeletal muscle. J Funct Morphol Kinesiol. 2016;1:289–302.
  • Takekura H, Flucher BE, Franzini-Armstrong C. Sequential docking, molecular differentiation, and positioning of T-tubule/SR junctions in developing mouse skeletal muscle. Dev Biol. 2001;239:204–214.
  • Franzini-Armstrong C. Simultaneous maturation of transverse tubules and sarcoplasmic reticulum during muscle differentiation in the mouse. Dev Biol. 1991;146:353–363.
  • Greising SM, Gransee HM, Mantilla CB, et al. Systems biology of skeletal muscle: fiber type as an organizing principle. Wiley Interdiscip Rev Syst Biol Med. 2012;4:457–473.
  • Bassel-Duby R, Olson EN. Signaling pathways in skeletal muscle remodeling. Annu Rev Biochem. 2006;75:19–37.
  • Black AJ, Ravi S, Jefferson LS, et al. Dietary fat quantity and type induce transcriptome-wide effects on alternative splicing of pre-mRNA in rat skeletal muscle. J Nutr. 2017;147:jn254482.
  • Clarke K, Ricciardi S, Pearson T, et al. The role of Eif6 in skeletal muscle homeostasis revealed by endurance training co-expression networks. Cell Rep. 2017;21:1507–1520.
  • Phillips SM, Glover EI, Rennie MJ. Alterations of protein turnover underlying disuse atrophy in human skeletal muscle. J Appl Physiol. 2009;107:645–654.
  • Sylow L, Kleinert M, Richter EA, et al. Exercise-stimulated glucose uptake — regulation and implications for glycaemic control. Nat Rev Endocrinol. 2017;13:133–148.
  • Lenasi T, Peterlin BM, Barboric M. Cap-binding protein complex links pre-mRNA capping to transcription elongation and alternative splicing through positive transcription elongation factor b (P-TEFb). J Biol Chem. 2011;286:22758–22768.
  • Ramanathan A, Robb GB, Chan SH. mRNA capping: biological functions and applications. Nucleic Acids Res. 2016;44:7511–7526.
  • Moteki S, Price D. Functional coupling of capping and transcription of mRNA. Mol Cell. 2002;10:599–609.
  • Shatkin AJ, Manley JL. The ends of the affair: capping and polyadenylation. Nat Struct Biol. 2000;7:838–842.
  • Gonatopoulos-Pournatzis T, Cowling VH. Cap-binding complex (CBC). Biochem J. 2014;457:231–242.
  • Pabis M, Neufeld N, Steiner MC, et al. The nuclear cap-binding complex interacts with the U4/U6·U5 tri-snRNP and promotes spliceosome assembly in mammalian cells. Rna. 2013;19:1054–1063.
  • Flaherty SM, Fortes P, Izaurralde E, et al. Participation of the nuclear cap binding complex in pre-mRNA 3ʹ processing. Proc Natl Acad Sci USA. 1997;94:11893–11898.
  • Andersen PR, Domanski M, Kristiansen MS, et al. The human cap-binding complex is functionally connected to the nuclear RNA exosome. Nat Struct Mol Biol. 2013;20:1367–1376.
  • Izaurralde E, Adam S. Transport of macromolecules between the nucleus and the cytoplasm. Rna. 1998;4:351–364.
  • Nojima T, Hirose T, Kimura H, et al. The interaction between cap-binding complex and RNA export factor is required for intronless mRNA export. J Biol Chem. 2007;282:15645–15651.
  • Daneholt B. Assembly and transport of a premessenger RNP particle. Proc Natl Acad Sci USA. 2001;98:7012–7017.
  • Grüner S, Peter D, Weber R, et al. The structures of eIF4E-eIF4G complexes reveal an extended interface to regulate translation initiation. Mol Cell. 2016;64:467–479.
  • Jackson RJ, Hellen CUT, Pestova TV. The mechanism of eukaryotic translation initiation and principles of its regulation. Nat Rev Mol Cell Biol. 2010;11:113–127.
  • Pestova TV, Kolupaeva VG, Lomakin IB, et al. Molecular mechanisms of translation initiation in eukaryotes. Proc Natl Acad Sci USA. 2001;98:7029–7036.
  • Frey JW, Jacobs BL, Goodman CA, et al. A role for Raptor phosphorylation in the mechanical activation of mTOR signaling. Cell Signal. 2014;26:313–322.
  • Gordon BS, Williamson DL, Lang CH, et al. Nutrient-induced stimulation of protein synthesis in mouse skeletal muscle is limited by the mTORC1 repressor REDD1. J Nutr. 2015;145:708–713.
  • Gordon BS, Steiner JL, Lang CH, et al. Reduced REDD1 expression contributes to activation of mTORC1 following electrically induced muscle contraction. Am J Physiol Endocrinol Metab. 2014;307:E703–11.
  • Rossetti ML, Fukuda DH, Gordon BS. Androgens induce growth of the limb skeletal muscles in a rapamycin-insensitive manner. Am J Physiol Integr Comp Physiol. 2018; 315:R721-R729.
  • Kelleher AR, Kimball SR, Dennis MD, et al. The mTORC1 signaling repressors REDD1/2 are rapidly induced and activation of p70S6K1 by leucine is defective in skeletal muscle of an immobilized rat hindlimb. AJP Endocrinol Metab. 2013;304:E229–36.
  • Kelleher AR, Gordon BS, Kimball SR, et al. Changes in REDD1, REDD2, and atrogene mRNA expression are prevented in skeletal muscle fixed in a stretched position during hindlimb immobilization. Physiol Rep. 2014;2:246.
  • Kelleher AR, Pereira SL, Jefferson LS, et al. REDD2 expression in rat skeletal muscle correlates with nutrient-induced activation of mTORC1: responses to aging, immobilization, and remobilization. Am J Physiol Endocrinol Metab. 2015;308:122–129.
  • Gingras AC, Kennedy SG, O’Leary MA, et al. 4E-BP1, a repressor of mRNA translation, is phosphorylated and inactivated by the Akt(PKB) signaling pathway. Genes Dev. 1998;12:502–513.
  • Haghighat A, Mader S, Pause A, et al. Repression of cap-dependent translation by 4E-binding protein 1: competition with p220 for binding to eukaryotic initiation factor-4E. Embo J. 1995;14:5701–5709.
  • Suzuki C, Garces RG, Edmonds KA, et al. PDCD4 inhibits translation initiation by binding to eIF4A using both its MA3 domains. Proc Natl Acad Sci U S A. 2008;105:3274–3279.
  • Dennis MD, Shenberger JS, Stanley BA, et al. Hyperglycemia mediates a shift from cap-dependent to cap-independent translation via a 4E-BP1-dependent mechanism. Diabetes. 2013;62:2204–2214.
  • Rommel C, Bodine SC, Clarke BA, et al. Mediation of IGF-1-induced skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways. Nat Cell Biol. 2001;3:1009–1013.
  • Binsch C, Jelenik T, Pfitzer A, et al. Absence of the kinase S6k1 mimics the effect of chronic endurance exercise on glucose tolerance and muscle oxidative stress. Mol Metab. 2017;6:1443–1453.
  • Ohanna M, Sobering AK, Lapointe T, et al. Atrophy of S6K1 −/− skeletal muscle cells reveals distinct mTOR effectors for cell cycle and size control. Nat Cell Biol. 2005;7:286–294.
  • Marabita M, Baraldo M, Solagna F, et al. S6K1 is required for increasing skeletal muscle force during hypertrophy. Cell Rep. 2016;17:501–513.
  • Steiner JL, Pruznak AM, Deiter G, et al. Disruption of genes encoding eIF4E binding proteins-1 and −2 does not alter basal or sepsis-induced changes in skeletal muscle protein synthesis in male or female mice. PLoS One. 2014;9:99582.
  • Merkin J, Russell C, Chen P, et al. Evolutionary dynamics of gene and isoform regulation in mammalian tissues. Science. 2012;338(6114):1593–1599.
  • Apponi LH, Corbett AH, Pavlath GK. RNA-binding proteins and gene regulation in myogenesis. Trends Pharmacol Sci. 2011;32:652–658.
  • Bland CS, Wang ET, Vu A, et al. Global regulation of alternative splicing during myogenic differentiation. Nucleic Acids Res. 2010;38:7651–7664.
  • Brinegar AE, Xia Z, Loehr JA, et al. Extensive alternative splicing transitions during postnatal skeletal muscle development are required for Ca 2+ handling. Elife. 2017;6:e27192.
  • Prokic I, Cowling BS, Laporte J. Amphiphysin 2 (BIN1) in physiology and diseases. J Mol Med. 2014;92:453–463.
  • Fugier C, Klein AF, Hammer C, et al. Misregulated alternative splicing of BIN1 is associated with T tubule alterations and muscle weakness in myotonic dystrophy. Nat Med. 2011;17:720–725.
  • Tjondrokoesoemo A, Park KH, Ferrante C, et al. Disrupted membrane structure and intracellular Ca 2+ signaling in adult skeletal muscle with acute knockdown of Bin1. PLoS One. 2011;6.
  • Lee E, Marcucci M, Daniell L, et al. Amphiphysin 2 (Bin1) and T-tubule biogenesis in muscle. Science. 2002;297:1193–1196.
  • Pineda-Lucena A, Ho CSW, Mao DYL, et al. A structure-based model of the c-Myc/Bin1 protein interaction shows alternative splicing of Bin1 and c-Myc phosphorylation are key binding determinants. J Mol Biol. 2005;351:182–194.
  • Ribeiro I, Yuan L, Tanentzapf G, et al. Phosphoinositide regulation of integrin trafficking required for muscle attachment and maintenance. PLoS Genet. 2011;7:e1001295.
  • Di Paolo G, De Camilli P. Phosphoinositides in cell regulation and membrane dynamics. Nature. 2006;443:651–657.
  • Böhm J, Vasli N, Maurer M, et al. Altered splicing of the BIN1 muscle-specific exon in humans and dogs with highly progressive centronuclear myopathy. PLoS Genet. 2013;9:e1003430.
  • Tuluc P, Molenda N, Schlick B, et al. A Cav1.1 Ca2+ channel splice variant with high conductance and voltage-sensitivity alters EC coupling in developing skeletal muscle. Biophys J. 2009;96:35–44.
  • Sultana N, Dienes B, Benedetti A, et al. Restricting calcium currents is required for correct fiber type specification in skeletal muscle. Development. 2016;143:1547–1559.
  • Tang ZZ, Yarotskyy V, Wei L, et al. Muscle weakness in myotonic dystrophy associated with misregulated splicing and altered gating of Cav1.1 calcium channel. Hum Mol Genet. 2012;21:1312–1324.
  • Santoro M, Masciullo M, Bonvissuto D, et al. Alternative splicing of human insulin receptor gene (INSR) in type i and type II skeletal muscle fibers of patients with myotonic dystrophy type 1 and type 2. Mol Cell Biochem. 2013;380:259–265.
  • Mosthaf L, Grako K, Dull TJ, et al. Functionally distinct insulin receptors generated by tissue-specific alternative splicing. Embo J. 1990;9:2409–2413.
  • Seino S, Bell G. Alternative splicing of human insulin receptor messenger RNA. Biochem Biophys Res Commun. 1989;159:312–316.
  • Belfiore A, Frasca F, Pandini G, et al. Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocr Rev. 2009;30:586–623.
  • Talukdar I, Sen S, Urbano R, et al. HnRNP A1 and hnRNP F modulate the alternative splicing of exon 11 of the insulin receptor gene. PLoS One. 2011;6.
  • Echeverria GV, Cooper TA. Muscleblind-like 1 activates insulin receptor exon 11 inclusion by enhancing U2AF65 binding and splicing of the upstream intron. Nucleic Acids Res. 2014;42:1893–1903.
  • Sen S, Talukdar I, Webster NJG. SRp20 and CUG-BP1 modulate insulin receptor exon 11 alternative splicing. Mol Cell Biol. 2009;29:871–880.
  • Bodine SC, Stitt TN, Gonzalez M, et al. Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol. 2001;3:1014–1019.
  • Savkur RS, Philips AV, Cooper TA, et al. Insulin receptor splicing alteration in myotonic dystrophy type 2. Am J Hum Genet. 2004;74:1309–1313.
  • Michel RN, Dunn SE, Chin ER. Calcineurin and skeletal muscle growth. Proc Nutr Soc. 2004;63:341–349.
  • Kimura T, Pace SM, Wei L, et al. A variably spliced region in the type 1 ryanodine receptor may participate in an inter-domain interaction. Biochem J. 2007;401:317–324.
  • Delling U, Tureckova J, Lim HW, et al. A calcineurin-NFATc3-dependent pathway regulates skeletal muscle differentiation and slow myosin heavy-chain expression. Mol Cell Biol. 2000;20:6600–6611.
  • Liu Y, Cseresnyés Z, Randall WR, et al. Activity-dependent nuclear translocation and intranuclear distribution of NFATc in adult skeletal muscle fibers. J Cell Biol. 2001;155:27–39.
  • Chin ER, Olson EN, Richardson JA, et al. A calcineurin-dependent transcriptional pathway controls skeletal muscle fiber type. Genes Dev. 1998;12:2499–2509.
  • Giudice J, Loehr JA, Rodney GG, et al. Alternative splicing of four trafficking genes regulates myofiber structure and skeletal muscle physiology. Cell Rep. 2016;17:1923–1933.
  • Lee KM, Tarn WY. Coupling pre-mRNA processing to transcription on the RNA factory assembly line. RNA Biol. 2013;10:380–390.
  • Tian B, Manley JL. Alternative polyadenylation of mRNA precursors. Nat Rev Mol Cell Biol. 2016;18:18–30.
  • Chen W, Jia Q, Song Y, et al. Alternative polyadenylation: methods, findings, and impacts. Genomics Proteomics Bioinformatics. 2017;15:287–300.
  • Blazie SM, Babb C, Wilky H, et al. Comparative RNA-Seq analysis reveals pervasive tissue-specific alternative polyadenylation in Caenorhabditis elegans intestine and muscles. BMC Biol. 2015;13:4. DOI:10.1186/s12915-015-0116-6.
  • Boutet SC, Cheung TH, Quach NL, et al. Alternative polyadenylation mediates microRNA regulation of muscle stem cell function. Cell Stem Cell. 2012;10:327–336.
  • Hirai H, Verma M, Watanabe S, et al. MyoD regulates apoptosis of myoblasts through microRNA-mediated down-regulation of Pax3. J Cell Biol. 2010;10:327–336.
  • Crist CG, Montarras D, Pallafacchina G, et al. Muscle stem cell behavior is modified by microRNA-27 regulation of Pax3 expression. Proc Natl Acad Sci U S A. 2009;11:13383–13387.
  • Fredericks AM, Cygan KJ, Brown BA, et al. RNA-binding proteins: splicing factors and disease. Biomolecules. 2015;5:893–909.
  • Hasan A, Cotobal C, Duncan CDS, et al. Systematic analysis of the role of RNA-binding proteins in the regulation of RNA stability. PLoS Genet. 2014;10:e1004684.
  • Gerstberger S, Hafner M, Tuschl T. A census of human RNA-binding proteins. Nat Rev Genet. 2014;15:829–845.
  • Batra R, Charizanis K, Manchanda M, et al. Loss of MBNL leads to disruption of developmentally regulated alternative polyadenylation in RNA-mediated disease. Mol Cell. 2014;56:311–322.
  • Martinez-Contreras R, Cloutier P, Shkreta L, et al. Alternative splicing in the postgenomic era. New York (NY): Springer Verlag New York; 2007.
  • Martinez-Contreras R, Fisette JF, Nasim FUH, et al. Intronic binding sites for hnRNP A/B and hnRNP F/H proteins stimulate pre-mRNA splicing. PLoS Biol. 2006;4:172–185.
  • Sun S, Zhang ZUO, Fregoso O, et al. Mechanisms of activation and repression by the alternative splicing factors RBFOX1/2. Rna. 2012;18:274–283.
  • Paul S, Dansithong W, Kim D, et al. Interaction of musleblind, CUG-BP1 and hnRNP H proteins in DM1-associated aberrant IR splicing. Embo J. 2006;25:4271–4283.
  • Li Q, Zheng S, Han A, et al. The splicing regulator PTBP2 controls a program of embryonic splicing required for neuronal maturation. Elife. 2014;3:e01201.
  • Wagner EJ, Garcia-Blanco MA. RNAi-mediated PTB depletion leads to enhanced exon definition. Mol Cell. 2002;10:943–949.
  • Xue Y, Zhou Y, Wu T, et al. Genome-wide analysis of PTB-RNA interactions reveals a strategy used by the general splicing repressor to modulate exon inclusion or skipping. Mol Cell. 2009;36:996–1006.
  • Llorian M, Schwartz S, Clark TA, et al. Position-dependent alternative splicing activity revealed by global profiling of alternative splicing events regulated by PTB. Nat Struct Mol Biol. 2010;17:1114–1123.
  • Keppetipola N, Sharma S, Li Q, et al. Neuronal regulation of pre-mRNA splicing by polypyrimidine tract binding proteins, PTBP1 and PTBP2. Crit Rev Biochem Mol Biol. 2012;47:360–378.
  • Tang ZZ, Sharma S, Zheng S, et al. Regulation of the mutually exclusive exons 8a and 8 in the CaV1.2 calcium channel transcript by polypyrimidine tract-binding protein. J Biol Chem. 2011;286:10007–10016.
  • Hall MP, Nagel RJ, Fagg WS, et al. Quaking and PTB control overlapping splicing regulatory networks during muscle cell differentiation quaking and PTB control overlapping splicing regulatory networks during muscle cell differentiation. Rna. 2013;19:627–638.
  • Galarneau A, Richard S. Target RNA motif and target mRNAs of the quaking STAR protein. Nat Struct Mol Biol. 2005;12:691–698.
  • Dasgupta T, Ladd A. The importance of CELF control: molecular and biological roles of the CUG-BP, Elav-like family of RNA binding proteins. Wiley Interdiscip Rev RNA. 2012;3:104–121.
  • Vlasova IA, Tahoe NM, Fan D, et al. Conserved GU-rich elements mediate mRNA decay by binding to CUG-binding protein 1. Mol Cell. 2008;29:263–270.
  • Kress C, Gautier-Courteille C, Osborne HB, et al. Inactivation of CUG-BP1/CELF1 causes growth, viability, and spermatogenesis defects in mice. Mol Cell Biol. 2007;27:1146–1157.
  • Giudice J, Xia Z, Li W, et al. Neonatal cardiac dysfunction and transcriptome changes caused by the absence of Celf1. Sci Rep. 2016;6:35550.
  • Wang ET, Ward AJ, Cherone JM, et al. Antagonistic regulation of mRNA expression and splicing by CELF and MBNL proteins. Genome Res. 2015;25:858–871.
  • Wang ET, Cody NAL, Jog S, et al. Transcriptome-wide regulation of pre-mRNA splicing and mRNA localization by muscleblind proteins. Cell. 2012;150:710–724.
  • Kühn U, Gündel M, Knoth A, et al. Poly(A) tail length is controlled by the nuclear Poly(A)-binding protein regulating the interaction between Poly(A) polymerase and the cleavage and polyadenylation specificity factor. J Biol Chem. 2009;284:22803–22814.
  • Jenal M, Elkon R, Loayza-Puch F, et al. The poly(A)-binding protein nuclear 1 suppresses alternative cleavage and polyadenylation sites. Cell. 2012;149:538–553.
  • Abbassi-Daloii T, Yousefi S, de Klerk E, et al. An alanine expanded PABPN1 causes increased utilization of intronic polyadenylation sites. Aging Mech Dis. 2017;3:6.
  • Goers ES, Purcell J, Voelker RB, et al. MBNL1 binds GC motifs embedded in pyrimidines to regulate alternative splicing. Nucleic Acids Res. 2010;38:2467–2484.
  • Kanadia RN, Johnstone KA, Mankodi A, et al. A muscleblind knockout model for myotonic dystrophy. Science. 2003;302:1978–1980.
  • Suenaga K, Lee KY, Nakamori M, et al. Muscleblind-like 1 knockout mice reveal novel splicing defects in the myotonic dystrophy brain. PLoS One. 2012;7:e33218.
  • Singh RK, Kolonin AM, Fiorotto ML, et al. Rbfox-splicing factors maintain skeletal muscle mass by regulating calpain3 and proteostasis. Cell Rep. 2018;24:197–208.
  • Pedrotti S, Giudice J, Dagnino-Acosta A, et al. The RNA-binding protein Rbfox1 regulates splicing required for skeletal muscle structure and function. Hum Mol Genet. 2015;24:2360–2374.
  • Singh RK, Xia Z, Bland CS, et al. Rbfox2-coordinated alternative splicing of Mef2d and Rock2 controls myoblast fusion during myogenesis. Mol Cell. 2014;55:592–603.
  • Lovci MT, Ghanem D, Marr H, et al. Rbfox proteins regulate alternative mRNA splicing through evolutionarily conserved RNA bridges. Nat Struct Mol Biol. 2013;20:1434–1442.
  • Sellier C, Cerro-Herreros E, Blatter M, et al. RbFOX1/MBNL1 competition for CCUG RNA repeats binding contributes to myotonic dystrophy type 1/type 2 differences. Nat Commun. 2018;9:1–15.
  • Runfola V, Sebastian S, Dilworth FJ, et al. Rbfox proteins regulate tissue-specific alternative splicing of Mef2D required for muscle differentiation. J Cell Sci. 2015;128:631–637.
  • Penn BH, Bergstrom DA, Dilworth FJ, et al. A MyoD -generated feed-forward circuit temporally patterns gene expression dining skeletal muscle differentiation. Genes Dev. 2004;18:2348–2353.
  • Mal A, Sturniolo M, Schiltz RL, et al. A role for histone deacetylase HDAC1 in modulating the transcriptional activity of MyoD: inhibition of the myogenic program. Embo J. 2001;20:1739–1753.
  • Mal A, Harter ML. MyoD is functionally linked to the silencing of a muscle-specific regulatory gene prior to skeletal myogenesis. Proc Natl Acad Sci USA. 2003;100:1735–1739.
  • Puri PL, Sartorelli V, Yang XJ, et al. Differential roles of p300 and PCAF acetyltransferases in muscle differentiation. Mol Cell. 1997;1:35–45.
  • Asp P, Blum R, Vethantham V, et al. Genome-wide remodeling of the epigenetic landscape during myogenic differentiation. Proc Natl Acad Sci USA. 2011;108:E149–E158.
  • Gómez-Del Arco P, Perdiguero E, Yunes-Leites PS, et al. The chromatin remodeling complex Chd4/NuRD controls striated muscle identity and metabolic homeostasis. Cell Metab. 2016;23:881–892.
  • Schor IE, Rascovan N, Pelisch F, et al. Neuronal cell depolarization induces intragenic chromatin modifications affecting NCAM alternative splicing. Proc Natl Acad Sci USA. 2009;106:43225–43230.
  • Schor IE, Fiszbein A, Petrillo E, et al. Intragenic epigenetic changes modulate NCAM alternative splicing in neuronal differentiation. Embo J. 2013;32:2264–2274.
  • Sims RJ, Millhouse S, Chen CF, et al. Recognition of trimethylated histone H3 Lysine 4 facilitates the recruitment of transcription postinitiation factors and pre-mRNA splicing. Mol Cell. 2007;28:665–676.
  • Saint-André V, Batsché E, Rachez C, et al. Histone H3 lysine 9 trimethylation and HP1γ favor inclusion of alternative exons. Nat Struct Mol Biol. 2011;18:337–344.
  • Luco RF, Pan Q, Tominaga K, et al. Regulation of alternative splicing by histone modifications. Science. 2010;327:996–1000.
  • Yong-Eun K, Chungoo P, Kyoon Eon K, et al. Histone and RNA-binding protein interaction creates crosstalk network for regulation of alternative splicing. Biochem Biophys Res Commun. 2018;499:30–36.
  • Jiang H, Mankodi A, Swanson MS, et al. Myotonic dystrophy type 1 is associated with nuclear foci of mutant RNA, sequestration of muscleblind proteins and deregulated alternative splicing in neurons. Hum Mol Genet. 2004;13:3079–3088.
  • Jauvin D, Chrétien J, Pandey SK, et al. Targeting DMPK with antisense oligonucleotide improves muscle strength in myotonic dystrophy type 1 mice. Mol Ther Nucleic Acids. 2017;7:465–474.
  • Nakamori M, Sobczak K, Puwanant A, et al. Splicing biomarkers of disease severity in myotonic dystrophy. Ann Neurol. 2014;74:862–872.
  • Thomas JD, Sznajder ŁJ, Bardhi O, et al. Disrupted prenatal RNA processing and myogenesis in congenital myotonic dystrophy. Genes Dev. 2017;31:1122–1133.
  • Fu YH, Pizzuti A, Fenwick RG Jr., et al. An unstable triplet repeat in a gene related to myotonic muscular dystrophy. Science. 1992;255:1256–1258.
  • Mahadevan M, Tsilfidis C, Sabourin L, et al. Myotonic dystrophy mutation: an unstable CTG repeat in the 3 ′ untranslated region of the gene. Science. 1992;255:1253–1255.
  • Liquori CL, Ricker K, Moseley ML, et al. Myotonic dystrophy type 2 caused by a CCTG expansion in intron I of ZNF9. Science. 2001;293:864–867.
  • Yuan Y, Compton SA, Sobczak K, et al. Muscleblind-like 1 interacts with RNA hairpins in splicing target and pathogenic RNAs. Nucleic Acids Res. 2007;35:5474–5486.
  • Thomas JD, Oliveira R, Sznajder ŁJ, et al. Myotonic dystrophy and developmental regulation of RNA processing. Compr Physiol. 2018;8:509–553.
  • Savkur RS, Philips AV, Cooper TA. Aberrant regulation of insulin receptor alternative splicing is associated with insulin resistance in myotonic dystrophy. Nat Genet. 2001;29:40–47.
  • Kuyumcu-Martinez NM, Wang G-S, Cooper T. Increased steady state levels of CUGBP1 in myotonic dystrophy 1 are due to PKC-mediated hyper-phosphorylation. Mol Cell. 2007;28:68–78.
  • Wheeler TM, Leger AJ, Pandey SK, et al. Targeting nuclear RNA for in vivo correction of myotonic dystrophy. Nature. 2014;488:111–115.
  • Scotti MM, Swanson MS. RNA mis-splicing in disease. Nat Rev Genet. 2016;17:19–32.
  • Nakamori M, Hamanaka K, Thomas JD, et al. Aberrant myokine signaling in congenital myotonic dystrophy. Cell Rep. 2017;21:1240–1252.
  • Pelosi M, De Rossi M, Barberi L, et al. IL-6 impairs myogenic differentiation by downmodulation of p90RSK/eEF2 and mTOR/p70S6K axes, without affecting AKT activity. Biomed Res Int. 2014;2014:206026.
  • Tuffery-Giraud S, Miro J, Koenig M, et al. Normal and altered pre-mRNA processing in the DMD gene. Hum Genet. 2017;136:1155–1172.
  • Lim KRQ, Maruyama R, Yokota T. Eteplirsen in the treatment of Duchenne muscular dystrophy. Drug Des Devel Ther. 2017;11:533–545.
  • Echigoya Y, Lim KRQ, Trieu N, et al. Quantitative antisense screening and optimization for exon 51 skipping in duchenne muscular dystrophy. Mol Ther. 2017;25:2561–2572.
  • Echevarría L, Aupy P, Goyenvalle A. Exon-skipping advances for Duchenne muscular dystrophy. Hum Mol Genet. 2018;27:163–172.
  • Kendall GC, Mokhonova EI, Moran M, et al. Dantrolene enhances antisense-mediated exon skipping in human and mouse models of Duchenne muscular dystrophy. Sci Transl Med. 2012;4:164ra160.
  • Kapeli K, Martinez FJ, Yeo GW. Genetic mutations in RNA-binding proteins and their roles in ALS. Hum Genet. 2017;136:1193–1214.
  • Polymenidou M, Lagier-Tourenne C, Hutt KR, et al. Misregulated RNA processing in amyotrophic lateral sclerosis. Brain Res. 2012;1462:3–15.
  • Taylor JP, Brown RH, Cleveland DW. Decoding ALS: from genes to mechanism. Nature. 2016;539:197–206.
  • Majounie E, Renton AE, Mok K, et al. Frequency of the C9orf72 hexanucleotide repeat expansion in patients with amyotrophic lateral sclerosis and frontotemporal dementia: A cross-sectional study. Lancet Neurol. 2012;11:323–330.
  • Barker HV, Niblock M, Lee Y-B, et al. RNA misprocessing in C9orf72-linked neurodegeneration. Front Cell Neurosci. 2017;11:195.
  • Mori K, Weng SM, Arzberger T, et al. The C9orf72 GGGGCC repeat is translated into aggregating dipeptide-repeat proteins in FTLD/ALS. Science. 2013;339:1335–1338.
  • Lee YB, Chen HJ, Peres JN, et al. Hexanucleotide repeats in ALS/FTD form length-dependent RNA Foci, sequester RNA binding proteins, and are neurotoxic. Cell Rep. 2013;5:1178–1186.
  • Arai T, Hasegawa M, Akiyama H, et al. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun. 2006;351:602–611.
  • Neumann M, Sampathu DM, Kwong LK, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314:130–133.
  • Tollervey JR, Curk T, Rogelj B, et al. Characterizing the RNA targets and position-dependent splicing regulation by TDP-43. Nat Neurosci. 2011;14:452–458.
  • Xiao S, Sanelli T, Dib S, et al. RNA targets of TDP-43 identified by UV-CLIP are deregulated in ALS. Mol Cell Neurosci. 2011;47:167–180.
  • Tollervey JR, Curk T, Rogelj B, et al. Characterizing the RNA targets and position-dependent splicing regulation by TDP-43. Nat Neurosci. 2011;14:452–458.
  • Tank EM, Figueroa-Romero C, Hinder LM, et al. Abnormal RNA stability in amyotrophic lateral sclerosis. Nat Commun. 2018;9:2845.
  • Tada M, Doi H, Koyano S, et al. Matrin 3 is a component of neuronal cytoplasmic inclusions of motor neurons in sporadic amyotrophic lateral sclerosis. Am J Pathol. 2018;188:507–514.
  • Farrawell NE, Lambert-Smith IA, Warraich ST, et al. Distinct partitioning of ALS associated TDP-43, FUS and SOD1 mutants into cellular inclusions. Sci Rep. 2015;5:13416.
  • Johnson JO, Pioro EP, Boehringer A, et al. Mutations in the Matrin 3 gene cause familial amyotrophic lateral sclerosis. Nat Neurosci. 2014;17:664–666.
  • Uemura Y, Oshima T, Yamamoto M, et al. Matrin3 binds directly to intronic pyrimidine-rich sequences and controls alternative splicing. Genes Cells. 2017;22:785–798.
  • Banerjee A, Vest KE, Pavlath GK, et al. Nuclear poly(A) binding protein 1 (PABPN1) and matrin3 interact in muscle cells and regulate RNA processing. Nucleic Acids Res. 2017;45:10706–10725.
  • Zhang ZC, Chook YM. Structural and energetic basis of ALS-causing mutations in the atypical proline–tyrosine nuclear localization signal of the Fused in Sarcoma protein (FUS). Proc Natl Acad Sci U S A. 2012;109:12017–12021.
  • Deng HX, Zhai H, Bigio EH, et al. FUS-immunoreactive inclusions are a common feature in sporadic and non-SOD1 familial amyotrophic lateral sclerosis. Ann Neurol. 2010;67:739–748.
  • Mailman MD, Heinz JW, Papp AC, et al. Molecular analysis of spinal muscular atrophy and modification of the phenotype by SMN2. Genet Med. 2002;4:20–26.
  • Hua Y, Sahashi K, Hung G, et al. Antisense correction of SMN2 splicing in the CNS rescues necrosis in a type III SMA mouse model. Genes Dev. 2010;24:1634–1644.
  • Mendell JR, Al-Zaidy S, Shell R, et al. Single-dose gene-replacement therapy for spinal muscular atrophy. N Engl J Med. 2017;377:1713–1722.
  • Passini MA, Bu J, Richards AM, et al. Antisense oligonucleotides ameliorate symptoms of severe spinal muscular atrophy. Sci Transl Med. 2011;3:72ra18.
  • Azzouz M, Le T, Ralph G. Lentivector-mediated SMN replacement in a mouse model of spinal muscular atrophy. J Clin Invest. 2004;114:1726–1731.
  • Naryshkin NA, Weetall M, Dakka A, et al. SMN2 splicing modifiers improve motor function and longevity in mice with spinal muscular atrophy. Science. 2014;345:688–693.

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.