Muscle-specific microRNAs as biomarkers of Duchenne Muscular Dystrophy progression and response to therapies

L Giordani, M Sandoná, A Rotini, P L Puri, S Consalvi, V Saccone, L Giordani, M Sandoná, A Rotini, P L Puri, S Consalvi, V Saccone

Abstract

Recent studies have revealed the contribution of fibro-adipogenic progenitors (FAPs) to the pathogenesis and progression of Duchenne Muscular Dystrophy (DMD). While FAPs direct compensatory regeneration at early stages of disease, as the disease progresses they contribute to the progressive replacement of contractile myofibers with fibrotic scars and fatty infiltration. Using the mouse model of DMD - the mdx mice - we have recently reported that FAPs mediate the ability of HDAC inhibitors (HDACi) to promote muscle regeneration and prevent fibro-adipogenic degeneration at early stages of disease. This effect is mediated by the induction of myomiRs that, in turn, target the SWI/SNF components BAF60A and B, thereby favoring the formation of BAF60C-based SWI/SNF complex, which directs the switch from the fibro-adipogenic to the myogenic lineage. Here we show direct evidence of induction of miR-206 and BAF60C, and reduction of BAF60A, in FAPs isolated from mdx muscles exposed to the HDACi Trichostatin A (TSA). We also discuss how increased expression of myomiRs in dystrophic muscles can be integrated with circulating myomiRs to provide accurate biomarkers of disease progression and response to treatment.

Keywords: BAF60; DMD; FAPs; diagnostic biomarkers; myomiRs.

Figures

Figure 1.
Figure 1.
Expression of BAF60a, BAF60c and miR-206 in FAPs isolated from young mdx mice treated with TSA (Trichostatin A). (A) Expression of BAF60c (Green), in growth medium (GM), was assessed by immunofluorescence. Nuclei were counterstained with DAPI (blue) (Left panel). The percentage of BAF60c positive cells per field was measured and reported as graphed (Right panel). The error bars indicate SEM. (B) Representative images of BAF60a (Red) expression assessed by immunofluorescence in GM (Left panel). Nuclei were counterstained with DAPI (blue). The right panel shows the percentage of BAF60a positive cells per field (Right panel). The error bars indicate SEM. (C) miR-206 expression in GM was evaluated by immunohistochemical staining of miR-206 (Blue/Purple). Nuclei were counterstained with DAPI (Blue for immunofluorescence or Yellow for phase contrast) (Left panel). (D) Myogenic differentiation potential was assessed by myosin heavy chain (MHC) immunofluorescence (Green) and adipogenic differentiation potential by Oil-Red O staining (Red). miR-206 expression in DM was evaluated by immunohistochemical staining. Nuclei were counterstained with DAPI (Blue for immunofluorescence or Yellow for phase contrast).
Figure 2.
Figure 2.
myomiRs as biomarkers of HDACi activity in DMD. FAPs are key cellular determinants of DMD progression. In pathologic conditions these cells differentiate into adipocytes and fibroblasts leading to muscle degeneration. This process is driven by BAF60a and BAF60b-based SWI/SNF complex that promotes the expression of fibro-adipogenic genes. As a consequence of muscle wasting, myomiRs are released in the circulation and have been proposed as biomarkers of DMD disease (Left panel). HDACi treatment at early stages promotes compensatory regeneration by inducing in FAPs BAF60c, which directs myogenesis, and up-regulating myomiRs, which suppress the fibro- adipogenesis In this context, a local increase of myomiRs inversely correlates with the decrease of circulating myomiRs, suggesting that the ratio between circulating/local myomiRs could provide an accurate biomarker of disease progression and response to HDACi (Right panel).

References

    1. Mendell JR, Boué DR, Martin PT. The con- genital muscular dystrophies: recent advances and molecular insights. Pediatr Dev Pathol 2006; 9:427-43; PMID:17163796;
    1. Dalkilic I, Kunkel LM. Muscular dystrophies: genes to pathogenesis. Curr Opin Genet Dev 2003; 13:231-38; PMID:12787784;
    1. Serrano AL, Mann CJ, Vidal B, Ardite E, Perdiguero E, Muñoz-Cánoves P. Cellular and molecular mechanisms regulating fibrosis in skeletal muscle repair and disease. Curr Top Dev Biol 2011; 96:167-201; PMID:21621071;
    1. Mozzetta C, Consalvi S, Saccone V, Tierney M, Diamantini A, Mitchell KJ, Marazzi G, Borsellino G, Battistini L, Sassoon D, et al. . Fibroadipogenic progenitors mediate the ability of HDAC inhibitors to promote regeneration in dystrophic muscles of young, but not old Mdx mice. EMBO Mol Med 2013; 5(4):626-39; PMID:23505062;
    1. Desguerre I, Mayer M, Leturcq F, Barbet JP, Gherardi RK, Christov C. Endomysial fibrosis in Duchenne muscular dystrophy: a marker of poor outcome associated with macrophage alternative activation. J Neuropathol Exp Neurol 2009; 68(7):762-73; PMID:19535995;
    1. Joe AW, Yi L, Natarajan A, Le Grand F, So L, Wang J, Rudnicki MA, Rossi FM. Muscle injury activates resident fibroadipogenic progenitors that facilitate myogenesis. Nat Cell Biol 2010; 2:153-63; PMID:20081841;
    1. Uezumi A, Fukada S, Yamamoto N, Takeda S, Tsuchida K. Mesenchymal progenitors distinct from satellite cells contribute to ectopic fat cell formation in skeletal muscle. Nat Cell Biol 2010; 2:143-52; PMID:20081842;
    1. Judson RN, Zhang RH, Rossi FM. Tissue-resident mesenchymal stemprogenitor cells in skeletal muscle: collaborators or saboteurs? FEBS J 2013; 280(17):4100-8; PMID:23763717;
    1. Malecova B, Puri PL. “Mix of Mics”- phenotypic and biological heterogeneity of “Multipotent” muscle interstitial cells (MICs). J Stem Cell Res Ther 2012; (Suppl 11):004; PMID:24634800;
    1. Heredia JE, Mukundan L, Chen FM, Mueller AA, Deo RC, Locksley RM, Rando TA, Chawla A. Type 2 innate signals stimulate fibroadipogenic progenitors to facilitate muscle regeneration. Cell 2013;153(2):376-88; PMID:23582327;
    1. Uezumi A, Ito T, Morikawa D, Shimizu N, Yoneda T, Segawa M, Yamaguchi M, Ogawa R, Matev MM, Miyagoe-Suzuki Y, et al. . Fibrosis and adipogenesis originate from a common mesenchymal progenitor in skeletal muscle. J Cell Sci 2011; 124:3654-64; PMID:22045730;
    1. Giordani L, Puri PL. Epigenetic control of skeletal muscle regeneration: integrating genetic determinants and environmental changes. FEBS J 2013; 280(17):4014-25; PMID:23745685;
    1. Minetti GC, Colussi C, Adami R, Serra C, Mozzetta C, Parente V, Fortuni S, Straino S, Sampaolesi M, Di Padova M, et al. . Functional and morphological recovery of dystrophic muscles in mice treated with deacetylase inhibitors. Nat Med 2006; 12(10):1147-50; PMID:16980968;
    1. Colussi C, Mozzetta C, Gurtner A, Illi B, Straino S, Ragone G, Pescatori M, Zaccagnini G, Rosati G, Minetti G, et al. . HDAC2 blockade by nitric oxide and histone deacetylase inhibitors reveals a common target in Duchenne muscular dystrophy treatment. Proc Natl Acad Sci 2008; 105(49):19183-7; PMID:19047631;
    1. Consalvi S, Mozzetta C, Bettica P, Germani M, Fiorentini F, Del Bene F, Rocchetti M, Leoni F, Monzani V, Mascagni P, et al. . Preclinical studies in the mdx mouse model of duchenne muscular dystrophy with the histone deacetylase inhibitor givinostat. Mol Med 2013; 19:79-87; PMID:23552722;
    1. Vianello S, Consolaro F, Bich C, Cancela JM, Roulot M, Lanchec E, Touboul D, Brunelle A, Israël M, Benoit E, et al. . Low doses of arginine butyrate derivatives improve dystrophic phenotype and restore membrane integrity in DMD models. FASEB J 2014; 28(6):2603-19; PMID:24604079;
    1. Johnson NM, Farr GH, Maves L. The HDAC inhibitor TSA ameliorates a zebrafish model of duchenne muscular dystrophy. PLoS Curr 2013; PMID:24459606;
    1. Saccone V, Consalvi S, Giordani L, Mozzetta C, Barozzi I, Sandoná M, Ryan T, Rojas-Muñoz A, Madaro L, Fasanaro P, et al. HDAC-regulated myomiRs control BAF60 variant exchange and direct the functional phenotype of fibro-adipogenic progenitors in dystrophic muscles. Genes Dev 2014; 28(8):841-57; PMID:24682306;
    1. Wu JI, Lessard J, Crabtree GR. Understanding the words of chromatin regulation. Cell 2009; 136(2):200-6; PMID:19167321;
    1. Puri PL, Mercola M. BAF60 A, B, and Cs of muscle determination and renewal. Genes Dev 2012; 26(24):2673-83; PMID:23222103;
    1. de la Serna IL, Ohkawa Y, Imbalzano AN. Chromatin remodelling in mammalian differentiation: lessons from ATP-dependent remodellers. Nat Rev Genet 2006; 7:461-73; PMID:16708073;
    1. Simone C, Forcales SV, Hill DA, Imbalzano AN, Latella L, Puri PL. p38 pathway targets SWI-SNF chromatin-remodeling complex to muscle-specific loci. Nat Genet 2004; 36:738-43; PMID:15208625;
    1. Wang W, Xue Y, Zhou S, Kuo A, Cairns BR, Crabtree GR. Diversity and specialization of mammalian SWISNF complexes. Genes Dev 1996; 10(17):2117-30; PMID:8804307;
    1. Albini S, Coutinho P, Malecova B, Giordani L, Savchenko A, Forcales SV, Puri PL. Epigenetic reprogramming of human embryonic stem cells into skeletal muscle cells and generation of contractile myospheres. Cell Rep 2013; 3:661-70; PMID:23478022;
    1. Lickert H, Takeuchi JK, Von Both I, Walls JR, McAuliffe F, Adamson SL, Henkelman RM, Wrana JL, Rossant J, Bruneau BG. Baf60c is essential for function of BAF chromatin remodelling complexes in heart development. Nature 2004; 432:107-112; PMID:15525990;
    1. Forcales SV, Albini S, Giordani L, Malecova B, Cignolo L, Chernov A, Coutinho P, Saccone V, Consalvi S, Williams R, et al. . Signal-dependent incorporation of MyoDBAF60c into Brg1-based SWISNF chromatin-remodelling complex. EMBO J 2011; 31:301-16; PMID:22068056;
    1. Li S, Liu C, Li N, Hao T, Han T, Hill DE, Vidal M, Lin JD. Genome-wide coactivation analysis of PGC-1a identifies BAF60a as a regulator of hepatic lipid metabolism. Cell Metab 2008; 8:105-117; PMID:18680712;
    1. Cacchiarelli D, Martone J, Girardi E, Cesana M, Incitti T, Morlando M, Nicoletti C, Santini T, Sthandier O, Barberi L, et al. . MicroRNAs involved in molecular circuitries relevant for the duchenne muscular dystrophy pathogenesis are controlled by the dystrophinnNOS pathway. Cell Metab 2010; 12:341-51; PMID:20727829;
    1. Sokol NS, Ambros V. Mesodermally expressed Drosophila microRNA-1 is regulated by Twist and is required in muscles during larval growth. Genes Dev 2005; 19(19):2343-54; PMID:16166373;
    1. Chen JF, Mandel EM, Thomson JM, Wu Q, Callis TE, Hammond SM, Conlon FL, Wang DZ. The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nat Genet 2006; 38(2):228-33; PMID:16380711;
    1. Kim HK, Lee YS, Sivaprasad U, Malhotra A, Dutta A. Muscle-specific microRNA miR-206 promotes muscle differentiation. J Cell Biol 2006; 174(5):677-87; PMID:16923828;
    1. Vignier N, Amor F, Fogel P, Duvallet A, Poupiot J, et al. . Distinctive serum miRNA profile in mouse models of striated muscular pathologies. PLoS One 2013; 8(2):e55281; PMID:23418438;
    1. Roberts TC, Blomberg KEM, McClorey G, Andaloussi SE, Godfrey C, et al. . Expression analysis in multiple muscle groups and serum reveals complexity in the MicroRNA transcriptome of the mdx mouse with implications for therapy. Mol Ther - Nucleic Acids 2012. 1:e39; PMID:23344181
    1. Cacchiarelli D, Legnini I, Martone J, Cazzella V, D’Amico A, Bertini E, Bozzoni I. miRNAs as serum biomarkers for Duchenne muscular dystrophy. EMBO Mol Med 2011; 3(5):258-65; PMID:21425469;
    1. Eisenberg I, Alexander MS, Kunkel LM. miRNAS in normal and diseased skeletal muscle. J Cell Mol Med 2009; 13(1):2-11; PMID:19175696;
    1. Williams AH, Liu N, van Rooij E, Olson EN. MicroRNA control of muscle development and disease. Curr Opin Cell Biol 2009; 21(3):461-9; PMID:19278845;
    1. Greco S, De Simone M, Colussi C, Zaccagnini G, Fasanaro P, Pescatori M, Cardani R, Perbellini R, Isaia E, Sale P, et al. . Common micro-RNA signature in skeletal muscle damage and regeneration induced by Duchenne muscular dystrophy and acute ischemia. FASEB J 2009; 23(10):3335-46; PMID:19528256;
    1. Goljanek-Whysall K, Mok GF, Fahad Alrefaei A, Kennerley N, Wheeler GN, Münsterberg A. MyomiR-dependent switching of BAF60 variant incorporation into Brg1 chromatin remodeling complexes during embryo myogenesis. Development 2014; 141(17):3378-87; PMID:25078649;
    1. Forterre A, Jalabert A, Chikh K, Pesenti S, Euthine V, Granjon A, Errazuriz E, Lefai E, Vidal H, Rome S. Myotube-derived exosomal miRNAs downregulate Sirtuin1 in myoblasts during muscle cell differentiation. Cell Cycle 2014; 13(1):78-89; PMID:24196440;
    1. He WA, Calore F, Londhe P, Canella A, Guttridge DC, Croce CM. Microvesicles containing miRNAs promote muscle cell death in cancer cachexia via TLR7. Proc Natl Acad Sci U S A 2014; 111(12):4525-9; PMID:24616506;
    1. Lessard J, Wu JI, Ranish JA, Wan M, Winslow MM, Staahl BT, Wu H, Aebersold R, Graef IA, Crabtree GR. An essential switch in subunit composition of a chromatin remodeling complex during neural development. Neuron 2007; 55:201-15; PMID:17640523;
    1. Yoo AS, Sun AX, Li L, Shcheglovitov A, Portmann T, Li Y, Lee-Messer C, Dolmetsch RE, Tsien RW, Crabtree GR. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature 2011; 476:228-31; PMID:21753754;
    1. Mizuno H, Nakamura A, Aoki Y, Ito N, Kishi S, Yamamoto K, Sekiguchi M, Takeda S, Hashido K. Identification of muscle-specific microRNAs in serum of muscular dystrophy animal models: promising novel blood-based markers for muscular dystrophy. PLoS One 2011; 6(3):e18388; PMID:21479190;
    1. Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol 2011. 13:423-33; PMID:21423178;
    1. Roberts TC, Godfrey C, McClorey G, Vader P, Briggs D, et al. . Extracellular microRNAs are dynamic non-vesicular biomarkers of muscle turnover. Nucl Acids Res 2013. 41:9500-13; PMID:23945935;
    1. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A, et al. . Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA 2008; 105:10513-8; PMID:18663219;
    1. Vlassov AV, Magdaleno S, Setterquist R, Conrad R. Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim Biophys Acta 2012; 1820:940-8; PMID:22503788;
    1. Borges FT, Melo SA, Ozdemir BC, Kato N, Revuelta I, Miller CA, Gattone VH, Lebleu VS, Kalluri R. TGF-β1-containing exosomes from injured epithelial cells activate fibroblasts to initiate tissue regenerative responses and fibrosis. J Am Soc Nephrol. 2013; 24:385-92; PMID:23274427;
    1. Roberts TC, Coenen-Stass AM, Betts CA, Wood MJ. Detection and quantification of extracellular microRNAs in murine biofluids. Biol Proced Online 2014; 16(1):5; PMID:24629058;
    1. Roberts TC, Coenen-Stass AM, Wood MJ. Assessment of RT-qPCR normalization strategies for accurate quantification of extracellular microRNAs in murine serum. PLoS One 2014; 9(2):e89237; PMID:24586621;
    1. Goyenvalle A, Babbs A, Wright J, Wilkins V, Powell D, et al. . Rescue of severely affected dystrophinutrophin-deficient mice through scAAV-U7snRNA-mediated exon skipping. Hum Mol Genet 2012; 21:2559-71; PMID:22388933;

Source: PubMed

3
Abonnieren