Renin-angiotensin-aldosterone system inhibitors improve membrane stability and change gene-expression profiles in dystrophic skeletal muscles.

Angiotensin-converting enzyme inhibitors (ACEi) and mineralocorticoid receptor (MR) antagonists are FDA-approved drugs that inhibit the renin-angiotensin-aldosterone system (RAAS) and are used to treat heart failure. Combined treatment with the ACEi lisinopril and the nonspecific MR antagonist spironolactone surprisingly improves skeletal muscle, in addition to heart function and pathology in a Duchenne muscular dystrophy (DMD) mouse model. We recently demonstrated that MR is present in all limb and respiratory muscles and functions as a steroid hormone receptor in differentiated normal human skeletal muscle fibers. The goals of the current study were to begin to define cellular and molecular mechanisms mediating the skeletal muscle efficacy of RAAS inhibitor treatment. We also compared molecular changes resulting from RAAS inhibition with those resulting from the current DMD standard-of-care glucocorticoid treatment. Direct assessment of muscle membrane integrity demonstrated improvement in dystrophic mice treated with lisinopril and spironolactone compared with untreated mice. Short-term treatments of dystrophic mice with specific and nonspecific MR antagonists combined with lisinopril led to overlapping gene-expression profiles with beneficial regulation of metabolic processes and decreased inflammatory gene expression. Glucocorticoids increased apoptotic, proteolytic, and chemokine gene expression that was not changed by RAAS inhibitors in dystrophic mice. Microarray data identified potential genes that may underlie RAAS inhibitor treatment efficacy and the side effects of glucocorticoids. Direct effects of RAAS inhibitors on membrane integrity also contribute to improved pathology of dystrophic muscles. Together, these data will inform clinical development of MR antagonists for treating skeletal muscles in DMD.

[1]  S. Raman,et al.  Similar efficacy from specific and non-specific mineralocorticoid receptor antagonist treatment of muscular dystrophy mice. , 2016, Journal of neuromuscular diseases.

[2]  S. Raman,et al.  The Angiotensin Converting Enzyme Inhibitor Lisinopril Improves Muscle Histopathology but not Contractile Function in a Mouse Model of Duchenne Muscular Dystrophy , 2015, Journal of neuromuscular diseases.

[3]  D. Guttridge,et al.  Mineralocorticoid receptors are present in skeletal muscle and represent a potential therapeutic target , 2015, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[4]  E. Hoffman,et al.  Contemporary Cardiac Issues in Duchenne Muscular Dystrophy , 2015, Circulation.

[5]  K. Oishi,et al.  Atypical expression of circadian clock genes in denervated mouse skeletal muscle , 2015, Chronobiology international.

[6]  K. Esser,et al.  Circadian Rhythms, the Molecular Clock, and Skeletal Muscle , 2015, Journal of biological rhythms.

[7]  S. Raman,et al.  Eplerenone for early cardiomyopathy in Duchenne muscular dystrophy: a randomised, double-blind, placebo-controlled trial , 2015, The Lancet Neurology.

[8]  J. Molkentin,et al.  Myofiber-specific inhibition of TGFβ signaling protects skeletal muscle from injury and dystrophic disease in mice. , 2014, Human molecular genetics.

[9]  I. Jaffe,et al.  Mineralocorticoid receptors in immune cells: Emerging role in cardiovascular disease , 2014, Steroids.

[10]  E. Hoffman,et al.  Asynchronous remodeling is a driver of failed regeneration in Duchenne muscular dystrophy , 2014, The Journal of cell biology.

[11]  Vassilios J. Bezzerides,et al.  Phenotypic screen quantifying differential regulation of cardiac myocyte hypertrophy identifies CITED4 regulation of myocyte elongation. , 2014, Journal of molecular and cellular cardiology.

[12]  J. Oprzadek,et al.  Transcriptional background of beef marbling - novel genes implicated in intramuscular fat deposition. , 2014, Meat science.

[13]  In-kyu Lee,et al.  Deficiency of clusterin exacerbates high-fat diet-induced insulin resistance in male mice. , 2014, Endocrinology.

[14]  S. Raman,et al.  Prednisolone Attenuates Improvement of Cardiac and Skeletal Contractile Function and Histopathology by Lisinopril and Spironolactone in the mdx Mouse Model of Duchenne Muscular Dystrophy , 2014, PloS one.

[15]  H. S. Neto,et al.  EPA protects against muscle damage in the mdx mouse model of Duchenne muscular dystrophy by promoting a shift from the M1 to M2 macrophage phenotype , 2013, Journal of Neuroimmunology.

[16]  J. Ntambi,et al.  SCD1 activity in muscle increases triglyceride PUFA content, exercise capacity, and PPARδ expression in mice[S] , 2013, Journal of Lipid Research.

[17]  E. Hoffman,et al.  VBP15, a novel anti-inflammatory and membrane-stabilizer, improves muscular dystrophy without side effects , 2013, EMBO molecular medicine.

[18]  J. Eeckhoute,et al.  Rev-erb-α modulates skeletal muscle oxidative capacity by regulating mitochondrial biogenesis and autophagy , 2013, Nature Medicine.

[19]  M. Malek,et al.  Similar skeletal muscle angiogenic and mitochondrial signalling following 8 weeks of endurance exercise in mice: discontinuous versus continuous training , 2013, Experimental physiology.

[20]  Xianhua Wang,et al.  Recombinant MG53 Protein Modulates Therapeutic Cell Membrane Repair in Treatment of Muscular Dystrophy , 2012, Science Translational Medicine.

[21]  A. Papavassiliou,et al.  Glucocorticoid receptor signaling in bone cells. , 2012, Trends in molecular medicine.

[22]  E. Hoffman,et al.  Glucocorticoid-Treated Mice Are an Inappropriate Positive Control for Long-Term Preclinical Studies in the mdx Mouse , 2012, PloS one.

[23]  F. Jaisser,et al.  Aldosterone, mineralocorticoid receptor, and heart failure , 2012, Molecular and Cellular Endocrinology.

[24]  Michael L. Johnson,et al.  Comparative effectiveness of different angiotensin-converting enzyme inhibitors on the risk of hospitalization in patients with heart failure. , 2012, Journal of comparative effectiveness research.

[25]  Cindy Hamil,et al.  Evidence‐based path to newborn screening for duchenne muscular dystrophy , 2012, Annals of neurology.

[26]  S. Raman,et al.  Early treatment with lisinopril and spironolactone preserves cardiac and skeletal muscle in Duchenne muscular dystrophy mice. , 2011, Circulation.

[27]  D. Vestal,et al.  The guanylate-binding proteins: emerging insights into the biochemical properties and functions of this family of large interferon-induced guanosine triphosphatase. , 2011, Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research.

[28]  B. Spiegelman,et al.  C/EBPβ Controls Exercise-Induced Cardiac Growth and Protects against Pathological Cardiac Remodeling , 2010, Cell.

[29]  M. Young,et al.  Activation of mineralocorticoid receptors by exogenous glucocorticoids and the development of cardiovascular inflammatory responses in adrenalectomized rats. , 2010, Endocrinology.

[30]  R. Finkel,et al.  Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and pharmacological and psychosocial management , 2010, The Lancet Neurology.

[31]  M. Young,et al.  Mediators of mineralocorticoid receptor-induced profibrotic inflammatory responses in the heart. , 2009, Clinical science.

[32]  J. Zierath,et al.  Relationship between AMPK and the transcriptional balance of clock-related genes in skeletal muscle. , 2008, American journal of physiology. Endocrinology and metabolism.

[33]  B. Pedersen,et al.  Calprotectin is released from human skeletal muscle tissue during exercise , 2008, The Journal of physiology.

[34]  K. Davies,et al.  Microarray analysis of mdx mice expressing high levels of utrophin: Therapeutic implications for dystrophin deficiency , 2008, Neuromuscular Disorders.

[35]  R. Ransohoff,et al.  Haploinsufficiency of utrophin gene worsens skeletal muscle inflammation and fibrosis in mdx mice , 2008, Journal of the Neurological Sciences.

[36]  F. Holsboer,et al.  Direct targeting of hippocampal neurons for apoptosis by glucocorticoids is reversible by mineralocorticoid receptor activation , 2005, Molecular Psychiatry.

[37]  J. D. Porter,et al.  Dissection of temporal gene expression signatures of affected and spared muscle groups in dystrophin-deficient (mdx) mice. , 2003, Human molecular genetics.

[38]  T. Slaga,et al.  Glucocorticoid receptor functions as a potent suppressor of mouse skin carcinogenesis , 2003, Oncogene.

[39]  The Transcription Factor C/EBP (cid:1) Serves as a Master Regulator of Physiologic Cardiac Hypertrophy C/EBPb Controls Exercise-Induced Cardiac Growth and Protects Against Pathological Cardiac Remodeling , 2022 .