1
|
Gunasekaran M, Littel HR, Wells NM, Turner J, Campos G, Venigalla S, Estrella EA, Ghosh PS, Daugherty AL, Stafki SA, Kunkel LM, Foley AR, Donkervoort S, Bönnemann CG, Toledo-Bravo de Laguna L, Nascimento A, Natera-de Benito D, Draper I, Bruels CC, Pacak CA, Kang PB. Effects of HMG CoA reductase (HMGCR) deficiency on skeletal muscle development. FEBS J 2025. [PMID: 39823152 DOI: 10.1111/febs.17406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/18/2024] [Accepted: 12/17/2024] [Indexed: 01/19/2025]
Abstract
Pathogenic variants in HMGCR were recently linked to a limb-girdle muscular dystrophy (LGMD) phenotype. The protein product HMG CoA reductase (HMGCR) catalyzes a key component of the cholesterol synthesis pathway. The two other muscle diseases associated with HMGCR, statin-associated myopathy (SAM) and autoimmune anti-HMGCR myopathy, are not inherited in a Mendelian pattern. Statins inhibit HMGCR activity to generate their cholesterol-lowering effects and are known to cause multiple types of adverse effects on skeletal muscle, while the antibodies associated with anti-HMGCR myopathy specifically target this enzyme. The mechanism linking pathogenic variants in HMGCR with skeletal muscle dysfunction is unclear. We knocked down Hmgcr in mouse skeletal myoblasts, knocked down hmgcr in Drosophila, and expressed three pathogenic HMGCR variants (c.1327C>T, p.Arg443Trp; c.1522_1524delTCT, p.Ser508del; and c.1621G>A, p.Ala541Thr) in Hmgcr knockdown mouse myoblasts. Hmgcr deficiency was associated with decreased proliferation, increased apoptosis, and impaired myotube fusion. Transcriptome sequencing of Hmgcr knockdown versus control myoblasts revealed differential expression involving mitochondrial function, with corresponding differences in cellular oxygen consumption rates. Both ubiquitous and muscle-specific knockdown of hmgcr in Drosophila led to lethality. Overexpression of reference HMGCR cDNA rescued myotube fusion in knockdown cells, whereas overexpression of the pathogenic variants of HMGCR cDNA did not. These results suggest that the three HMGCR-related muscle diseases share disease mechanisms related to skeletal muscle development.
Collapse
Affiliation(s)
- Mekala Gunasekaran
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Hannah R Littel
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Natalya M Wells
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Johnnie Turner
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Gloriana Campos
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Sree Venigalla
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Elicia A Estrella
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Partha S Ghosh
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Audrey L Daugherty
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Seth A Stafki
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Louis M Kunkel
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | | | - Andres Nascimento
- Neuromuscular Unit, Department of Neurology, Hospital Sant Joan de Déu, Barcelona, Spain
- Applied Research in Neuromuscular Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Daniel Natera-de Benito
- Neuromuscular Unit, Department of Neurology, Hospital Sant Joan de Déu, Barcelona, Spain
- Applied Research in Neuromuscular Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Isabelle Draper
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Christine C Bruels
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Christina A Pacak
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Peter B Kang
- Greg Marzolf Jr. Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
2
|
Gunasekaran M, Littel HR, Wells NM, Turner J, Campos G, Venigalla S, Estrella EA, Ghosh PS, Daugherty AL, Stafki SA, Kunkel LM, Foley AR, Donkervoort S, Bönnemann CG, Toledo-Bravo de Laguna L, Nascimento A, Benito DND, Draper I, Bruels CC, Pacak CA, Kang PB. Effects of HMGCR deficiency on skeletal muscle development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.591934. [PMID: 38903061 PMCID: PMC11188090 DOI: 10.1101/2024.05.06.591934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Pathogenic variants in HMGCR were recently linked to a limb-girdle muscular dystrophy (LGMD) phenotype. The protein product HMG CoA reductase (HMGCR) catalyzes a key component of the cholesterol synthesis pathway. The two other muscle diseases associated with HMGCR, statin-associated myopathy (SAM) and autoimmune anti-HMGCR myopathy, are not inherited in a Mendelian pattern. The mechanism linking pathogenic variants in HMGCR with skeletal muscle dysfunction is unclear. We knocked down Hmgcr in mouse skeletal myoblasts, knocked down hmgcr in Drosophila, and expressed three pathogenic HMGCR variants (c.1327C>T, p.Arg443Trp; c.1522_1524delTCT, p.Ser508del; and c.1621G>A, p.Ala541Thr) in Hmgcr knockdown mouse myoblasts. Hmgcr deficiency was associated with decreased proliferation, increased apoptosis, and impaired myotube fusion. Transcriptome sequencing of Hmgcr knockdown versus control myoblasts revealed differential expression involving mitochondrial function, with corresponding differences in cellular oxygen consumption rates. Both ubiquitous and muscle-specific knockdown of hmgcr in Drosophila led to lethality. Overexpression of reference HMGCR cDNA rescued myotube fusion in knockdown cells, whereas overexpression of the pathogenic variants of HMGCR cDNA did not. These results suggest that the three HMGCR-related muscle diseases share disease mechanisms related to skeletal muscle development.
Collapse
|
3
|
Okada R, Son SM, Fresquez Z, Formanek B, Mertz K, Buser Z, Wang JC. Association of Hyperlipidemia With Perioperative Complications in Posterior Cervical Spine Fusion: A Comparative Retrospective Study. Clin Spine Surg 2023; 36:E457-E463. [PMID: 37482645 DOI: 10.1097/bsd.0000000000001497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 06/21/2023] [Indexed: 07/25/2023]
Abstract
STUDY DESIGN A retrospective database study. OBJECTIVES The purpose of the current study was to investigate the impact of hyperlipidemia (HLD) on the incidence of perioperative complications associated with posterior cervical spine fusion (PCF). BACKGROUND HLD is a very common disease that leads to atherosclerosis. Therefore, it can cause fatal diseases as well as lifestyle-related diseases. The possible impact of HLD on outcomes after PCF has not yet been investigated. METHODS Patients with cervical degeneration underwent initial PCF from 2010 through the third quarter of 2020 using the MSpine subset of the PearlDiver Patient Record Database. The incidence of perioperative complications was queried using relevant ICD-9, 10, and CPT codes. χ 2 analysis was performed in age-, sex-, and Charlson Comorbidity Index (CCI)-matched populations to compare between non-HLD and HLD patients in each single-level and multilevel PCF. RESULTS Through propensity score matching, 1600 patients each in the HLD and non-HLD groups were analyzed in the single-level PCF, 6855 patients were analyzed in the multilevel PCF were analyzed. The comorbidity of HLD significantly decreased the incidence of respiratory failure in single-level PCF (OR=0.58, P <0.01). In the multilevel PCF, the presence of HLD increased the incidence of cervicalgia (OR=1.26, P =0.030). On the contrary, the incident of spinal cord injury (OR=0.72, P <0.01), dysphagia (OR=0.81, P =0.023), respiratory failure (OR=0.85, P =0.030), pneumonia (OR=0.70, P =0.045), neurological bladder (OR=0.84, P =0.041), and urinary tract infection (OR=0.85, P =0.021) in the HLD group were significantly lower than those in non-HLD group. CONCLUSIONS In the current study, the presence of HLD significantly increased the incidence of postoperative cervicalgia in multilevel PCF. On the other hand, the incidence of some complications was significantly decreased with HLD. Further studies are needed taking into account other factors such as the treatment of HLD, its efficacy, and intraoperative events. LEVEL OF EVIDENCE Level III.
Collapse
Affiliation(s)
- Rintaro Okada
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Seung Min Son
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Zoe Fresquez
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Blake Formanek
- University of Queensland School of Medicine, Ochsner Clinical School, Queensland, Australia
| | - Kevin Mertz
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Zorica Buser
- Department of Orthopaedic Surgery, Grossman School of Medicine, New York University, New York, NY
| | - Jeffrey C Wang
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
4
|
Dallevet CA, Benveniste O, Allenbach Y. Pathogenesis and Treatment in IMNM. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2023. [DOI: 10.1007/s40674-023-00201-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
5
|
Wang Y, Lu J, Liu Y. Skeletal Muscle Regeneration in Cardiotoxin-Induced Muscle Injury Models. Int J Mol Sci 2022; 23:ijms232113380. [PMID: 36362166 PMCID: PMC9657523 DOI: 10.3390/ijms232113380] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Skeletal muscle injuries occur frequently in daily life and exercise. Understanding the mechanisms of regeneration is critical for accelerating the repair and regeneration of muscle. Therefore, this article reviews knowledge on the mechanisms of skeletal muscle regeneration after cardiotoxin-induced injury. The process of regeneration is similar in different mouse strains and is inhibited by aging, obesity, and diabetes. Exercise, microcurrent electrical neuromuscular stimulation, and mechanical loading improve regeneration. The mechanisms of regeneration are complex and strain-dependent, and changes in functional proteins involved in the processes of necrotic fiber debris clearance, M1 to M2 macrophage conversion, SC activation, myoblast proliferation, differentiation and fusion, and fibrosis and calcification influence the final outcome of the regenerative activity.
Collapse
|
6
|
Pathophysiological Mechanisms and Treatment of Dermatomyositis and Immune Mediated Necrotizing Myopathies: A Focused Review. Int J Mol Sci 2022; 23:ijms23084301. [PMID: 35457124 PMCID: PMC9030619 DOI: 10.3390/ijms23084301] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 12/15/2022] Open
Abstract
Idiopathic inflammatory myopathies (IIM), collectively known as myositis, are a composite group of rare autoimmune diseases affecting mostly skeletal muscle, although other organs or tissues may also be involved. The main clinical feature of myositis is subacute, progressive, symmetrical muscle weakness in the proximal arms and legs, whereas subtypes of myositis may also present with extramuscular features, such as skin involvement, arthritis or interstitial lung disease (ILD). Established subgroups of IIM include dermatomyositis (DM), immune-mediated necrotizing myopathy (IMNM), anti-synthetase syndrome (ASyS), overlap myositis (OM) and inclusion body myositis (IBM). Although these subgroups have overlapping clinical features, the widespread variation in the clinical manifestations of IIM suggests different pathophysiological mechanisms. Various components of the immune system are known to be important immunopathogenic pathways in IIM, although the exact pathophysiological mechanisms causing the muscle damage remain unknown. Current treatment, which consists of glucocorticoids and other immunosuppressive or immunomodulating agents, often fails to achieve a sustained beneficial response and is associated with various adverse effects. New therapeutic targets have been identified that may improve outcomes in patients with IIM. A better understanding of the overlapping and diverging pathophysiological mechanisms of the major subgroups of myositis is needed to optimize treatment. The aim of this review is to report on recent advancements regarding DM and IMNM.
Collapse
|
7
|
Simvastatin Downregulates Cofilin and Stathmin to Inhibit Skeletal Muscle Cells Migration. Int J Mol Sci 2022; 23:ijms23052848. [PMID: 35269994 PMCID: PMC8911248 DOI: 10.3390/ijms23052848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/27/2022] [Accepted: 03/02/2022] [Indexed: 01/27/2023] Open
Abstract
Statins are the most effective therapeutic agents for reducing cholesterol synthesis. Given their widespread use, many adverse effects from statins have been reported; of these, musculoskeletal complications occurred in 15% of patients after receiving statins for 6 months, and simvastatin was the most commonly administered statin among these cases. This study investigated the negative effects of simvastatin on skeletal muscle cells. We performed RNA sequencing analysis to determine gene expression in simvastatin-treated cells. Cell proliferation and migration were examined through cell cycle analysis and the transwell filter migration assay, respectively. Cytoskeleton rearrangement was examined through F-actin and tubulin staining. Western blot analysis was performed to determine the expression of cell cycle-regulated and cytoskeleton-related proteins. Transfection of small interfering RNAs (siRNAs) was performed to validate the role of cofilin and stathmin in the simvastatin-mediated inhibition of cell migration. The results revealed that simvastatin inhibited the proliferation and migration of skeletal muscle cells and affected the rearrangement of F-actin and tubulin. Simvastatin reduced the expression of cofilin and stathmin. The knockdown of both cofilin and stathmin by specific siRNA synergistically impaired cell migration. In conclusion, our results indicated that simvastatin inhibited skeletal muscle cell migration by reducing the expressions of cofilin and stathmin.
Collapse
|
8
|
Allenbach Y, Benveniste O, Stenzel W, Boyer O. Immune-mediated necrotizing myopathy: clinical features and pathogenesis. Nat Rev Rheumatol 2020; 16:689-701. [PMID: 33093664 DOI: 10.1038/s41584-020-00515-9] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2020] [Indexed: 12/11/2022]
Abstract
Immune-mediated necrotizing myopathy (IMNM) is a group of inflammatory myopathies that was distinguished from polymyositis in 2004. Most IMNMs are associated with anti-signal recognition particle (anti-SRP) or anti-3-hydroxy-3-methylglutaryl-coA reductase (anti-HMGCR) myositis-specific autoantibodies, although ~20% of patients with IMNM remain seronegative. These associations have led to three subclasses of IMNM: anti-SRP-positive IMNM, anti-HMGCR-positive IMNM and seronegative IMNM. IMNMs are frequently rapidly progressive and severe, displaying high serum creatine kinase levels, and failure to treat IMNMs effectively may lead to severe muscle impairment. In patients with seronegative IMNM, disease can be concomitant with cancer. Research into IMNM pathogenesis has shown that anti-SRP and anti-HMGCR autoantibodies cause weakness and myofibre necrosis in mice, suggesting that, as well as being diagnostic biomarkers of IMNM, they may play a key role in disease pathogenesis. Therapeutically, treatments such as rituximab or intravenous immunoglobulins can now be discussed for IMNM, and targeted therapies, such as anticomplement therapeutics, may be a future option for patients with refractory disease.
Collapse
Affiliation(s)
- Yves Allenbach
- Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Inserm U974, Department of Internal Medicine and Clinical Immunology, Pitié-Salpêtrière University Hospital, Paris, France
| | - Olivier Benveniste
- Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Inserm U974, Department of Internal Medicine and Clinical Immunology, Pitié-Salpêtrière University Hospital, Paris, France.
| | - Werner Stenzel
- Department of Neuropathology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Olivier Boyer
- Normandie University, UNIROUEN, Inserm U1234, Department of Immunology and Biotherapy, Rouen University Hospital, Rouen, France
| |
Collapse
|
9
|
ProNGF/p75NTR Axis Drives Fiber Type Specification by Inducing the Fast-Glycolytic Phenotype in Mouse Skeletal Muscle Cells. Cells 2020; 9:cells9102232. [PMID: 33023189 PMCID: PMC7599914 DOI: 10.3390/cells9102232] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022] Open
Abstract
Despite its undisputable role in the homeostatic regulation of the nervous system, the nerve growth factor (NGF) also governs the relevant cellular processes in other tissues and organs. In this study, we aimed at assessing the expression and the putative involvement of NGF signaling in skeletal muscle physiology. To reach this objective, we employed satellite cell-derived myoblasts as an in vitro culture model. In vivo experiments were performed on Tibialis anterior from wild-type mice and an mdx mouse model of Duchenne muscular dystrophy. Targets of interest were mainly assessed by means of morphological, Western blot and qRT-PCR analysis. The results show that proNGF is involved in myogenic differentiation. Importantly, the proNGF/p75NTR pathway orchestrates a slow-to-fast fiber type transition by counteracting the expression of slow myosin heavy chain and that of oxidative markers. Concurrently, proNGF/p75NTR activation facilitates the induction of fast myosin heavy chain and of fast/glycolytic markers. Furthermore, we also provided evidence that the oxidative metabolism is impaired in mdx mice, and that these alterations are paralleled by a prominent buildup of proNGF and p75NTR. These findings underline that the proNGF/p75NTR pathway may play a crucial role in fiber type determination and suggest its prospective modulation as an innovative therapeutic approach to counteract muscle disorders.
Collapse
|
10
|
Barrons R, Woods JA, Humphries R. Statin Associated Autoimmune Myonecrosis: Case Report With Delayed Onset and Treatment Challenges. J Pharm Pract 2020; 35:129-134. [PMID: 32924771 DOI: 10.1177/0897190020958223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE A case of delayed statin associated autoimmune myopathy (SAAM) is presented along with review of clinical findings and treatment strategies. SUMMARY A 54 year old male presented with proximal extremity weakness, difficulty ambulating, and dysphagia. Symptoms began when restarting atorvastatin 40 mg daily for a recent NSTEMI, following 10 years of statin use, interrupted after diagnosis of NASH. Relevant labs included CK of 13,618 IU/L, ALT/ AST of 568/407 IU/L, while additional liver, renal, and toxicology tests were normal. Following treatment response to prednisone 40 mg daily for 3 days, outpatient testing for anti-HMGCR antibodies was ordered.Twelve days from discharge, the patient was readmitted for myalgia and dysphagia, CK = 6042 IU/L, ALT/AST = 360/112 IU/L, and positive anti-HMGCR antibodies. Newly diagnosed with SAAM, symptoms improved with methylprednisolone and intravenous immunoglobulin (IVIG), continuing outpatient as daily prednisone and monthly IVIG. Four days later, the patient relapsed with worsened weakness and dysphagia, CK = 5812 IU/L, and ALT/AST = 647/337 IU/L. After response to methylprednisolone and rituximab, the patient was discharged on a corticosteroid taper, biweekly rituximab, and monthly IVIG. Two weeks later, a final admission involved a syncopal episode and fall, with a CK = 1461 IU/L. Treatment included IVIG, rituximab, and corticosteroid taper, which lead to remission for greater than 6 months. CONCLUSION Statin associated autoimmune myopathy occurred when restarting atorvastatin, following 10 years of statin use. Clinical findings and positive anti-HMGCR antibodies confirmed the diagnosis. Recurrent relapses required triple combination therapy including addition of rituximab to achieve remission.
Collapse
|
11
|
The impact of statins on physical activity and exercise capacity: an overview of the evidence, mechanisms, and recommendations. Eur J Appl Physiol 2020; 120:1205-1225. [PMID: 32248287 DOI: 10.1007/s00421-020-04360-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 03/24/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE Statins are among the most widely prescribed medications worldwide. Considered the 'gold-standard' treatment for cardiovascular disease (CVD), statins inhibit HMG-CoA reductase to ultimately reduce serum LDL-cholesterol levels. Unfortunately, the main adverse event of statin use is the development of muscle-associated problems, referred to as SAMS (statin-associated muscle symptoms). While regular moderate physical activity also decreases CVD risk, there is apprehension that physical activity may induce and/or exacerbate SAMS. While much work has gone into identifying the epidemiology of SAMS, only recent research has focused on the extent to which these muscle symptoms are accompanied by functional declines. The purpose of this review is to provide an overview of possible mechanisms underlying SAMS and summarize current evidence regarding the relationship between statin treatment, physical activity, exercise capacity, and SAMS development. METHODS PubMed and Google Scholar databases were used to search the most relevant and up-to-date peer-reviewed research on the topic. RESULTS The mechanism(s) behind SAMS, including altered mitochondrial metabolism, reduced coenzyme Q10 levels, reduced vitamin D levels, impaired calcium homeostasis, elevated extracellular glutamate, and genetic polymorphisms, still lack consensus and remain up for debate. Our summation of the evidence leads us to suggest that the etiology of SAMS development is likely multifactorial. Our review also demonstrates that there is limited evidence for statins impairing exercise adaptations or reducing exercise capacity for the majority of the investigated populations. CONCLUSION The available evidence indicates that the benefits of engaging in physical activity while on statin medication largely outweigh the risks.
Collapse
|
12
|
Hoppstädter J, Valbuena Perez JV, Linnenberger R, Dahlem C, Legroux TM, Hecksteden A, Tse WKF, Flamini S, Andreas A, Herrmann J, Herr C, Müller R, Meyer T, Bals R, Riccardi C, Bruscoli S, Kiemer AK. The glucocorticoid-induced leucine zipper mediates statin-induced muscle damage. FASEB J 2020; 34:4684-4701. [PMID: 32030813 DOI: 10.1096/fj.201902557rrr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/21/2020] [Accepted: 01/21/2020] [Indexed: 12/31/2022]
Abstract
Statins, the most prescribed class of drugs for the treatment of hypercholesterolemia, can cause muscle-related adverse effects. It has been shown that the glucocorticoid-induced leucine zipper (GILZ) plays a key role in the anti-myogenic action of dexamethasone. In the present study, we aimed to evaluate the role of GILZ in statin-induced myopathy. Statins induced GILZ expression in C2C12 cells, primary murine myoblasts/myotubes, primary human myoblasts, and in vivo in zebrafish embryos and human quadriceps femoris muscle. Gilz induction was mediated by FOXO3 activation and binding to the Gilz promoter, and could be reversed by the addition of geranylgeranyl, but not farnesyl, pyrophosphate. Atorvastatin decreased Akt phosphorylation and increased cleaved caspase-3 levels in myoblasts. This effect was reversed in myoblasts from GILZ knockout mice. Similarly, myofibers isolated from knockout animals were more resistant toward statin-induced cell death than their wild-type counterparts. Statins also impaired myoblast differentiation, and this effect was accompanied by GILZ induction. The in vivo relevance of our findings was supported by the observation that gilz overexpression in zebrafish embryos led to impaired embryonic muscle development. Taken together, our data point toward GILZ as an essential mediator of the molecular mechanisms leading to statin-induced muscle damage.
Collapse
Affiliation(s)
- Jessica Hoppstädter
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany.,Department of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | | | - Rebecca Linnenberger
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Charlotte Dahlem
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Thierry M Legroux
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Anne Hecksteden
- Institute of Sports and Preventive Medicine, Saarland University, Saarbrücken, Germany
| | - William K F Tse
- Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Sara Flamini
- Department of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Anastasia Andreas
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Jennifer Herrmann
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Christian Herr
- Department of Internal Medicine V-Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Rolf Müller
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Tim Meyer
- Institute of Sports and Preventive Medicine, Saarland University, Saarbrücken, Germany
| | - Robert Bals
- Department of Internal Medicine V-Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Carlo Riccardi
- Department of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Stefano Bruscoli
- Department of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Alexandra K Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| |
Collapse
|
13
|
Abstract
There is now overwhelming evidence to support lowering LDL-c (low-density lipoprotein cholesterol) to reduce cardiovascular morbidity and mortality. Statins are a class of drugs frequently prescribed to lower cholesterol. However, in spite of their wide-spread use, discontinuation and nonadherence remains a major gap in both the primary and secondary prevention of atherosclerotic cardiovascular disease. The major reason for statin discontinuation is because of the development of statin-associated muscle symptoms, but a range of other statin-induced side effects also exist. Although the mechanisms behind these side effects have not been fully elucidated, there is an urgent need to identify those at increased risk of developing side effects as well as provide alternative treatment strategies. In this article, we review the mechanisms and clinical importance of statin toxicity and focus on the evaluation and management of statin-associated muscle symptoms.
Collapse
Affiliation(s)
- Natalie C Ward
- From the School of Public Health, Curtin University, Perth, Western Australia, Australia (N.C.W.).,School of Medicine, University of Western Australia, Perth, Australia (N.C.W., G.F.W.)
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, Australia (N.C.W., G.F.W.).,Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Western Australia, Australia (G.F.W.)
| | - Robert H Eckel
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora (R.H.E.)
| |
Collapse
|
14
|
Fracassi A, Marangoni M, Rosso P, Pallottini V, Fioramonti M, Siteni S, Segatto M. Statins and the Brain: More than Lipid Lowering Agents? Curr Neuropharmacol 2019; 17:59-83. [PMID: 28676012 PMCID: PMC6341496 DOI: 10.2174/1570159x15666170703101816] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/24/2017] [Accepted: 06/26/2017] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Statins represent a class of medications widely prescribed to efficiently treat dyslipidemia. These drugs inhibit 3-βhydroxy 3β-methylglutaryl Coenzyme A reductase (HMGR), the rate-limiting enzyme of mevalonate (MVA) pathway. Besides cholesterol, MVA pathway leads to the production of several other compounds, which are essential in the regulation of a plethora of biological activities, including in the central nervous system. For these reasons, statins are able to induce pleiotropic actions, and acquire increased interest as potential and novel modulators in brain processes, especially during pathological conditions. OBJECTIVE The purpose of this review is to summarize and examine the current knowledge about pharmacokinetic and pharmacodynamic properties of statins in the brain. In addition, effects of statin on brain diseases are discussed providing the most up-to-date information. METHODS Relevant scientific information was identified from PubMed database using the following keywords: statins and brain, central nervous system, neurological diseases, neurodegeneration, brain tumors, mood, stroke. RESULTS 315 scientific articles were selected and analyzed for the writing of this review article. Several papers highlighted that statin treatment is effective in preventing or ameliorating the symptomatology of a number of brain pathologies. However, other studies failed to demonstrate a neuroprotective effect. CONCLUSION Even though considerable research studies suggest pivotal functional outcomes induced by statin therapy, additional investigation is required to better determine the pharmacological effectiveness of statins in the brain, and support their clinical use in the management of different neuropathologies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marco Segatto
- Address correspondence to this author at the Department of Sense Organs, Sapienza University, viale del Policlinico 155, 00186 Rome, Italy; E-mail:
| |
Collapse
|
15
|
Simvastatin Impairs the Inflammatory and Repair Phases of the Postinjury Skeletal Muscle Regeneration. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7617312. [PMID: 30519583 PMCID: PMC6241344 DOI: 10.1155/2018/7617312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 10/08/2018] [Accepted: 10/17/2018] [Indexed: 12/27/2022]
Abstract
Background Recent clinical data have suggested that the chronic use of high-lipophilic statins impairs the regenerative capacity of skeletal muscle. Because this activity of statins is poorly understood, we aimed to investigate the effect of simvastatin (SIM) on postinjury myofibre regeneration. Methods The porcine model was used in this study. The animals were divided into two groups: nontreated (control; n=24) and SIM-treated (40 mg/day; n=24). On the 15th day (day 0) of the experiment, a bupivacaine hydrochloride- (BPVC-) induced muscle injury was established, and the animals were sacrificed in the following days after muscle injury. The degree of regeneration was assessed based on histopathological and immunohistochemical examinations. The presence and degree of extravasation, necrosis, and inflammation in the inflammatory phase were assessed, whereas the repair phase was evaluated based on the numbers of muscle precursor cells (MPCs), myotube and young myofibres. Results In the inflammatory phase, SIM increased the distribution and prolonged the period of extravasation, prolonged the duration of necrosis, and prolonged and enhanced the infiltration of inflammatory cells. In the repair phase, SIM delayed and prolonged the activity of MPCs, delayed myotube formation, and delayed and decreased the formation of young myofibres. Our results indicated that SIM did not improve blood vessel stabilization at the site of the injury, did not exert an anti-inflammatory effect, prolonged and enhanced the inflammatory response, and impaired MPC activity, differentiation, and fusion. Moreover, SIM appeared to reduce M1 macrophage activity, resulting in slower removal of necrotic debris and sustained necrosis. Conclusion This study shows that SIM negatively affects the inflammatory and repair phases of the postinjury muscle regeneration. These findings are unique, strengthen the available knowledge on the side effects of SIM, and provide evidence showing that statin therapy is associated with an increased risk of impairment of the regenerative capacity of muscle.
Collapse
|
16
|
Ladislau L, Arouche-Delaperche L, Allenbach Y, Benveniste O. Potential Pathogenic Role of Anti-Signal Recognition Protein and Anti-3-hydroxy-3-methylglutaryl-CoA Reductase Antibodies in Immune-Mediated Necrotizing Myopathies. Curr Rheumatol Rep 2018; 20:56. [PMID: 30074107 DOI: 10.1007/s11926-018-0763-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW This review provides an overview of the potential pathogenic roles of anti-SRP and anti-HMGCR in IMNM over the past 5 years. RECENT FINDINGS Idiopathic inflammatory myopathies (IIM) are a group of acquired autoimmune disorders that mainly affect the skeletal muscle tissue. Classification criteria of IIM are comprised of polymyositis, dermatomyositis, inclusion body myositis and immune-mediated necrotizing myopathies. One important hallmark of autoimmune diseases is the detection of autoantibodies in patient sera. The anti-SRP (signal recognition particle) and anti-HMGCR (3-hydroxy-3-methylglutaryl coenzyme A reductase) antibodies are specifically associated with IMNM patients, and their detection has been described as related to disease severity. The muscles of IMNM patients are characterized by necrosis, atrophy and regenerating fibres with sparse inflammatory infiltrates. Although an important correlation between autoantibody titres, creatine kinase levels and disease progression/severity has been described in the last few years, the potential pathogenic roles of these autoantibodies have only recently been described.
Collapse
Affiliation(s)
- Leandro Ladislau
- UPMC Univ Paris 06, INSERM UMRS_974, Center of Research in Myology, AP-HP, Department of Internal Medicine & Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013, Paris, France
| | - Louiza Arouche-Delaperche
- UPMC Univ Paris 06, INSERM UMRS_974, Center of Research in Myology, AP-HP, Department of Internal Medicine & Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013, Paris, France
| | - Yves Allenbach
- UPMC Univ Paris 06, INSERM UMRS_974, Center of Research in Myology, AP-HP, Department of Internal Medicine & Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013, Paris, France
| | - Olivier Benveniste
- UPMC Univ Paris 06, INSERM UMRS_974, Center of Research in Myology, AP-HP, Department of Internal Medicine & Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013, Paris, France.
| |
Collapse
|
17
|
Hydrogen Sulfide Alleviates Lipopolysaccharide-Induced Diaphragm Dysfunction in Rats by Reducing Apoptosis and Inflammation through ROS/MAPK and TLR4/NF- κB Signaling Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9647809. [PMID: 29977458 PMCID: PMC5994286 DOI: 10.1155/2018/9647809] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/11/2018] [Accepted: 04/29/2018] [Indexed: 12/16/2022]
Abstract
Diaphragm dysfunction is an important clinical problem worldwide. Hydrogen sulfide (H2S) is involved in many physiological and pathological processes in mammals. However, the effect and mechanism of H2S in diaphragm dysfunction have not been fully elucidated. In this study, we detected that the level of H2S was decreased in lipopolysaccharide- (LPS-) treated L6 cells. Treatment with H2S increased the proliferation and viability of LPS-treated L6 cells. We found that H2S decreased reactive oxygen species- (ROS-) induced apoptosis through the mitogen-activated protein kinase (MAPK) signaling pathway in LPS-treated L6 cells. Administration of H2S alleviated LPS-induced inflammation by mediating the toll-like receptor-4 (TLR-4)/nuclear factor-kappa B (NF-κB) signaling pathway in L6 cells. Furthermore, H2S improved diaphragmatic function and structure through the reduction of inflammation and apoptosis in the diaphragm of septic rats. In conclusion, these findings indicate that H2S ameliorates LPS-induced diaphragm dysfunction in rats by reducing apoptosis and inflammation through ROS/MAPK and TLR4/NF-κB signaling pathways. Novel slow-releasing H2S donors can be designed and applied for the treatment of diaphragm dysfunction.
Collapse
|
18
|
Selva-O'Callaghan A, Alvarado-Cardenas M, Pinal-Fernández I, Trallero-Araguás E, Milisenda JC, Martínez MÁ, Marín A, Labrador-Horrillo M, Juárez C, Grau-Junyent JM. Statin-induced myalgia and myositis: an update on pathogenesis and clinical recommendations. Expert Rev Clin Immunol 2018; 14:215-224. [PMID: 29473763 DOI: 10.1080/1744666x.2018.1440206] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Musculoskeletal manifestations are well-recognized side effects of treatment with statins. New advances in this field have appeared in recent years. This review focuses on the diagnosis of these conditions and their underlying pathogenesis, in particular immune-mediated necrotizing myopathy. Areas covered: Clinical phenotypes including rhabdomyolysis, myalgia and/or mild hyperCKemia, self-limited toxin statin myopathy, and immune-mediated necrotizing myopathy are herein described. Therapeutic recommendations and a diagnostic algorithm in statin-associated myopathy are also proposed. The etiology and pathogenesis of statin-induced myopathy has mainly focused on the anti-HMGCR antibodies and the responsibility of the immune-mediated necrotizing myopathy is discussed. The fact that patients who have not been exposed to statins may develop statin-associated autoimmune myopathy with anti-HMGCR antibodies is also addressed. The literature search strategy included terms identified by searches of PubMed between 1969 and December 2017. The search terms 'myositis', 'statin-induced autoimmune myopathy', 'immune-mediate necrotizing myopathy', 'statins', 'muscular manifestations', and 'anti-HMGCR antibodies' were used. Expert commentary: Full characterization of the known phenotypes of statin toxicity and the specific role of the anti-HMGCR in those exposed and not exposed (i.e. juvenile forms) to statins and in some types of neoplasms is of paramount relevance.
Collapse
Affiliation(s)
- Albert Selva-O'Callaghan
- a Systemic Autoimmune Diseases Unit, Vall d'Hebron General Hospital , Universitat Autonoma de Barcelona , Barcelona , Spain
| | - Marcelo Alvarado-Cardenas
- a Systemic Autoimmune Diseases Unit, Vall d'Hebron General Hospital , Universitat Autonoma de Barcelona , Barcelona , Spain
| | - Iago Pinal-Fernández
- b National Institutes of Health, Muscle Diseases , NIAMS , Bethesda , MD , USA.,c Department of Neurology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Ernesto Trallero-Araguás
- d Rheumatology Unit, Vall d'Hebron General Hospital , Universitat Autonoma de Barcelona , Barcelona , Spain
| | - José Cesar Milisenda
- e Internal Medicine Department, Hospital Clinic , Universitat de Barcelona , CIBERER , Barcelona , Spain
| | - María Ángeles Martínez
- f Immunology Department, Hospital de la Santa Creu i Sant Pau , Universitat Autonoma de Barcelona , Barcelona , Spain
| | - Ana Marín
- g Immunology Department, Vall d'Hebron General Hospital , Universitat Autonoma de Barcelona , Barcelona , Spain
| | - Moisés Labrador-Horrillo
- a Systemic Autoimmune Diseases Unit, Vall d'Hebron General Hospital , Universitat Autonoma de Barcelona , Barcelona , Spain.,g Immunology Department, Vall d'Hebron General Hospital , Universitat Autonoma de Barcelona , Barcelona , Spain
| | - Cándido Juárez
- f Immunology Department, Hospital de la Santa Creu i Sant Pau , Universitat Autonoma de Barcelona , Barcelona , Spain
| | - Josep María Grau-Junyent
- e Internal Medicine Department, Hospital Clinic , Universitat de Barcelona , CIBERER , Barcelona , Spain
| |
Collapse
|
19
|
Rebalka IA, Cao AW, Raleigh MJ, Henriksbo BD, Coleman SK, Schertzer JD, Hawke TJ. Statin Therapy Negatively Impacts Skeletal Muscle Regeneration and Cutaneous Wound Repair in Type 1 Diabetic Mice. Front Physiol 2017; 8:1088. [PMID: 29311999 PMCID: PMC5742241 DOI: 10.3389/fphys.2017.01088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022] Open
Abstract
Those with diabetes invariably develop complications including cardiovascular disease (CVD). To reduce their CVD risk, diabetics are generally prescribed cholesterol-lowering 3-hydroxy-methylglutaryl coenzyme A reductase inhibitors (i.e., statins). Statins inhibit cholesterol biosynthesis, but also reduce the synthesis of a number of mevalonate pathway intermediates, leading to several cholesterol-independent effects. One of the pleiotropic effects of statins is the reduction of the anti-fibrinolytic hormone plasminogen activator inhibitor-1 (PAI-1). We have previously demonstrated that a PAI-1 specific inhibitor alleviated diabetes-induced delays in skin and muscle repair. Here we tested if statin administration, through its pleiotropic effects on PAI-1, could improve skin and muscle repair in a diabetic rodent model. Six weeks after diabetes onset, adult male streptozotocin-induced diabetic (STZ), and WT mice were assigned to receive control chow or a diet enriched with 600 mg/kg Fluvastatin. Tibialis anterior muscles were injured via Cardiotoxin injection to induce skeletal muscle injury. Punch biopsies were administered on the dorsal scapular region to induce injury of skin. Twenty-four days after the onset of statin therapy (10 days post-injury), tissues were harvested and analyzed. PAI-1 levels were attenuated in statin-treated diabetic tissue when compared to control-treated tissue, however no differences were observed in non-diabetic tissue as a result of treatment. Muscle and skin repair were significantly attenuated in Fluvastatin-treated STZ-diabetic mice as demonstrated by larger wound areas, less mature granulation tissue, and an increased presence of smaller regenerating muscle fibers. Despite attenuating PAI-1 levels in diabetic tissue, Fluvastatin treatment impaired cutaneous healing and skeletal muscle repair in STZ-diabetic mice.
Collapse
Affiliation(s)
- Irena A Rebalka
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Andrew W Cao
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Matthew J Raleigh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Brandyn D Henriksbo
- Department of Biochemistry and Biomedical Sciences and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Samantha K Coleman
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
20
|
Mohassel P, Foley AR, Donkervoort S, Fequiere PR, Pak K, Bönnemann CG, Mammen AL. Anti-3-hydroxy-3-methylglutaryl-coenzyme a reductase necrotizing myopathy masquerading as a muscular dystrophy in a child. Muscle Nerve 2017; 56:1177-1181. [DOI: 10.1002/mus.25567] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 12/20/2016] [Accepted: 01/06/2017] [Indexed: 11/08/2022]
Affiliation(s)
- Payam Mohassel
- National Institute of Neurological Disorders and Stroke; National Institutes of Health; Bethesda Maryland USA
| | - A. Reghan Foley
- National Institute of Neurological Disorders and Stroke; National Institutes of Health; Bethesda Maryland USA
| | - Sandra Donkervoort
- National Institute of Neurological Disorders and Stroke; National Institutes of Health; Bethesda Maryland USA
| | - Pierre R. Fequiere
- Department of Pediatrics, Division of Pediatric Neurology; University of Alabama at Birmingham; Birmingham Alabama USA
| | - Katherine Pak
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health; Building 50, Room 1146, Bethesda Maryland 20892 USA
| | - Carsten G. Bönnemann
- National Institute of Neurological Disorders and Stroke; National Institutes of Health; Bethesda Maryland USA
| | - Andrew L. Mammen
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health; Building 50, Room 1146, Bethesda Maryland 20892 USA
| |
Collapse
|
21
|
Arouche-Delaperche L, Allenbach Y, Amelin D, Preusse C, Mouly V, Mauhin W, Tchoupou GD, Drouot L, Boyer O, Stenzel W, Butler-Browne G, Benveniste O. Pathogenic role of anti-signal recognition protein and anti-3-Hydroxy-3-methylglutaryl-CoA reductase antibodies in necrotizing myopathies: Myofiber atrophy and impairment of muscle regeneration in necrotizing autoimmune myopathies. Ann Neurol 2017; 81:538-548. [DOI: 10.1002/ana.24902] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 12/27/2017] [Accepted: 02/09/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Louiza Arouche-Delaperche
- Pierre and Marie Curie University, Sorbonne Universities, National Institute of Health and Medical Research, National Center for Scientific Research, Myology Research Center; Pitié-Salpêtrière University Hospital; Paris France
| | - Yves Allenbach
- Pierre and Marie Curie University, Sorbonne Universities, National Institute of Health and Medical Research, National Center for Scientific Research, Myology Research Center; Pitié-Salpêtrière University Hospital; Paris France
- Department of Internal Medicine and Clinical Immunology, University Hospital Department of Inflammation, Immunopathology, and Biotherapy, Pitié-Salpêtrière University Hospital; Public Hospital Network of Paris; Paris France
| | - Damien Amelin
- Pierre and Marie Curie University, Sorbonne Universities, National Institute of Health and Medical Research, National Center for Scientific Research, Myology Research Center; Pitié-Salpêtrière University Hospital; Paris France
| | - Corinna Preusse
- Department of Neuropathology; Charité-Universitätsmedizin; Berlin Germany
| | - Vincent Mouly
- Pierre and Marie Curie University, Sorbonne Universities, National Institute of Health and Medical Research, National Center for Scientific Research, Myology Research Center; Pitié-Salpêtrière University Hospital; Paris France
| | - Wladimir Mauhin
- Pierre and Marie Curie University, Sorbonne Universities, National Institute of Health and Medical Research, National Center for Scientific Research, Myology Research Center; Pitié-Salpêtrière University Hospital; Paris France
| | - Gaelle Dzangue Tchoupou
- Pierre and Marie Curie University, Sorbonne Universities, National Institute of Health and Medical Research, National Center for Scientific Research, Myology Research Center; Pitié-Salpêtrière University Hospital; Paris France
| | - Laurent Drouot
- Department of Immunology; University of Normandy UNIROUEN, National Institute of Health and Medical Research U1234, Rouen University Hospital; Rouen France
| | - Olivier Boyer
- Department of Immunology; University of Normandy UNIROUEN, National Institute of Health and Medical Research U1234, Rouen University Hospital; Rouen France
| | - Werner Stenzel
- Department of Neuropathology; Charité-Universitätsmedizin; Berlin Germany
| | - Gillian Butler-Browne
- Pierre and Marie Curie University, Sorbonne Universities, National Institute of Health and Medical Research, National Center for Scientific Research, Myology Research Center; Pitié-Salpêtrière University Hospital; Paris France
| | - Olivier Benveniste
- Pierre and Marie Curie University, Sorbonne Universities, National Institute of Health and Medical Research, National Center for Scientific Research, Myology Research Center; Pitié-Salpêtrière University Hospital; Paris France
- Department of Internal Medicine and Clinical Immunology, University Hospital Department of Inflammation, Immunopathology, and Biotherapy, Pitié-Salpêtrière University Hospital; Public Hospital Network of Paris; Paris France
| |
Collapse
|
22
|
Muntean DM, Thompson PD, Catapano AL, Stasiolek M, Fabis J, Muntner P, Serban MC, Banach M. Statin-associated myopathy and the quest for biomarkers: can we effectively predict statin-associated muscle symptoms? Drug Discov Today 2016; 22:85-96. [PMID: 27634340 DOI: 10.1016/j.drudis.2016.09.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/28/2016] [Accepted: 09/05/2016] [Indexed: 12/21/2022]
Abstract
Over the past three decades, statins have become the cornerstone of prevention and treatment of atherosclerotic cardiovascular and metabolic diseases. Albeit generally well tolerated, these drugs can elicit a variety of muscle-associated symptoms that represent the most important reason for treatment discontinuation. Statin-associated myopathy has been systematically underestimated by randomized controlled trials as compared with the incidence observed in clinical practice and obtained from patient registries. There are several reasons for this discrepancy, among which the lack of reliable diagnostic tests and a validated questionnaire to assess muscle symptoms are recognized as unmet needs. Here, we review the cellular and molecular mechanisms underlying statin-associated myopathy and discuss the experimental and clinical data on various biomarkers to diagnose and predict muscle-related complaints.
Collapse
Affiliation(s)
- Danina M Muntean
- Department of Pathophysiology Functional Sciences, Victor Babeş University of Medicine and Pharmacy of Timisoara, Timisoara, Romania; Center for Translational Research and Systems Medicine, Victor Babeş University of Medicine and Pharmacy of Timisoara, Timisoara, Romania
| | - Paul D Thompson
- Division of Cardiology, Hartford Hospital, Hartford, CT, USA
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, IRCCS Multimedica, Milan, Italy
| | - Mariusz Stasiolek
- Department of Neurology, Polish Mother's Memorial Hospital-Research Institute in Lodz, Lodz, Poland
| | - Jaroslaw Fabis
- Department of Arthroscopy, Minimally Invasive Surgery and Sports Traumatology, Medical University of Lodz, Poland
| | - Paul Muntner
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Maria-Corina Serban
- Department of Pathophysiology Functional Sciences, Victor Babeş University of Medicine and Pharmacy of Timisoara, Timisoara, Romania; Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Poland; Healthy Aging Research Centre (HARC), Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
23
|
Mancini GJ, Baker S, Bergeron J, Fitchett D, Frohlich J, Genest J, Gupta M, Hegele RA, Ng D, Pearson GJ, Pope J, Tashakkor AY. Diagnosis, Prevention, and Management of Statin Adverse Effects and Intolerance: Canadian Consensus Working Group Update (2016). Can J Cardiol 2016; 32:S35-65. [DOI: 10.1016/j.cjca.2016.01.003] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/03/2016] [Accepted: 01/05/2016] [Indexed: 12/24/2022] Open
|
24
|
Martínez-Redondo V, Jannig PR, Correia JC, Ferreira DMS, Cervenka I, Lindvall JM, Sinha I, Izadi M, Pettersson-Klein AT, Agudelo LZ, Gimenez-Cassina A, Brum PC, Dahlman-Wright K, Ruas JL. Peroxisome Proliferator-activated Receptor γ Coactivator-1 α Isoforms Selectively Regulate Multiple Splicing Events on Target Genes. J Biol Chem 2016; 291:15169-84. [PMID: 27231350 DOI: 10.1074/jbc.m115.705822] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Indexed: 11/06/2022] Open
Abstract
Endurance and resistance exercise training induces specific and profound changes in the skeletal muscle transcriptome. Peroxisome proliferator-activated receptor γ coactivator-1 α (PGC-1α) coactivators are not only among the genes differentially induced by distinct training methods, but they also participate in the ensuing signaling cascades that allow skeletal muscle to adapt to each type of exercise. Although endurance training preferentially induces PGC-1α1 expression, resistance exercise activates the expression of PGC-1α2, -α3, and -α4. These three alternative PGC-1α isoforms lack the arginine/serine-rich (RS) and RNA recognition motifs characteristic of PGC-1α1. Discrete functions for PGC-1α1 and -α4 have been described, but the biological role of PGC-1α2 and -α3 remains elusive. Here we show that different PGC-1α variants can affect target gene splicing through diverse mechanisms, including alternative promoter usage. By analyzing the exon structure of the target transcripts for each PGC-1α isoform, we were able to identify a large number of previously unknown PGC-1α2 and -α3 target genes and pathways in skeletal muscle. In particular, PGC-1α2 seems to mediate a decrease in the levels of cholesterol synthesis genes. Our results suggest that the conservation of the N-terminal activation and repression domains (and not the RS/RNA recognition motif) is what determines the gene programs and splicing options modulated by each PGC-1α isoform. By using skeletal muscle-specific transgenic mice for PGC-1α1 and -α4, we could validate, in vivo, splicing events observed in in vitro studies. These results show that alternative PGC-1α variants can affect target gene expression both quantitatively and qualitatively and identify novel biological pathways under the control of this system of coactivators.
Collapse
Affiliation(s)
- Vicente Martínez-Redondo
- From the Department of Physiology and Pharmacology, Molecular and Cellular Exercise Physiology Unit and
| | - Paulo R Jannig
- From the Department of Physiology and Pharmacology, Molecular and Cellular Exercise Physiology Unit and School of Physical Education and Sport, University of São Paulo, 05508-030 São Paulo, Brazil, and
| | - Jorge C Correia
- From the Department of Physiology and Pharmacology, Molecular and Cellular Exercise Physiology Unit and
| | - Duarte M S Ferreira
- From the Department of Physiology and Pharmacology, Molecular and Cellular Exercise Physiology Unit and
| | - Igor Cervenka
- From the Department of Physiology and Pharmacology, Molecular and Cellular Exercise Physiology Unit and
| | - Jessica M Lindvall
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, SE-141 83 Huddinge, Sweden
| | - Indranil Sinha
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, SE-141 83 Huddinge, Sweden
| | - Manizheh Izadi
- From the Department of Physiology and Pharmacology, Molecular and Cellular Exercise Physiology Unit and
| | - Amanda T Pettersson-Klein
- From the Department of Physiology and Pharmacology, Molecular and Cellular Exercise Physiology Unit and
| | - Leandro Z Agudelo
- From the Department of Physiology and Pharmacology, Molecular and Cellular Exercise Physiology Unit and
| | - Alfredo Gimenez-Cassina
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Patricia C Brum
- School of Physical Education and Sport, University of São Paulo, 05508-030 São Paulo, Brazil, and
| | - Karin Dahlman-Wright
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, SE-141 83 Huddinge, Sweden
| | - Jorge L Ruas
- From the Department of Physiology and Pharmacology, Molecular and Cellular Exercise Physiology Unit and
| |
Collapse
|
25
|
Sato H, Funaki A, Kimura Y, Sumitomo M, Yoshida H, Fukata H, Ueno K. Ethanol extract of Cyclolepis genistoides D. Don (palo azul) induces formation of myotubes, which involves differentiation of C2C12 myoblast cells. Nutr Res 2016; 36:731-41. [PMID: 27262535 DOI: 10.1016/j.nutres.2016.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/12/2016] [Accepted: 02/25/2016] [Indexed: 01/08/2023]
Abstract
In this study, we examined the cell differentiation effect of an ethanol extract of Cyclolepis genistoides D. Don, a herbaceous perennial belonging to the family Asteraceae (vernacular name: palo azul). Palo azul has numerous physiological effects that contribute to the prevention of metabolic syndromes, although the mechanism remains unclear. We previously suggested that palo azul has antidiabetic activity via an adipose differentiation effect. Here, we focused on whether palo azul promoted the differentiation of myoblasts. The mouse muscle myoblast cell line C2C12 was cultured and differentiated using horse serum with or without an ethanol extract of palo azul (12.5-200 μg/mL). Quantitative real-time polymerase chain reaction was performed to evaluate differentiation markers, including insulin-like growth factor-1 and myogenin. To evaluate myotube formation, myosin heavy-chain (MHC) expression and localization were detected by immunohistochemistry. Palo azul increased the expression of the differentiation markers. Furthermore, immunohistochemistry analysis revealed increased formation of MHC myotubes after palo azul treatment along with increased diameter and fusion indices of the myotubes. The expression level of MHC was also increased. In conclusion, palo azul may increase muscle mass in the body and improve insulin resistance conditions by facilitating the formation of myotubes by promoting myocyte differentiation.
Collapse
Affiliation(s)
- Hiromi Sato
- Department of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba-shi, Chiba 260-8675, Japan.
| | - Asami Funaki
- Department of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba-shi, Chiba 260-8675, Japan
| | - Yuki Kimura
- Department of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba-shi, Chiba 260-8675, Japan
| | - Mai Sumitomo
- Department of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba-shi, Chiba 260-8675, Japan
| | - Hiroya Yoshida
- IHM Inc, 7-22-17, Nishigotanda, Shinagawa-ku, Tokyo 141-0031, Japan
| | - Hideki Fukata
- JPD Co. Ltd, 7-98, Kitaitami, Itami-shi, Hyogo 664-0831, Japan
| | - Koichi Ueno
- Center of Preventive Medical Science, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba-shi, Chiba 260-8675, Japan
| |
Collapse
|
26
|
|
27
|
Peric D, Barragan I, Giraud-Triboult K, Egesipe AL, Meyniel-Schicklin L, Cousin C, Lotteau V, Petit V, Touhami J, Battini JL, Sitbon M, Pinset C, Ingelman-Sundberg M, Laustriat D, Peschanski M. Cytostatic Effect of Repeated Exposure to Simvastatin: A Mechanism for Chronic Myotoxicity Revealed by the Use of Mesodermal Progenitors Derived from Human Pluripotent Stem Cells. Stem Cells 2015; 33:2936-48. [DOI: 10.1002/stem.2107] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 06/06/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Delphine Peric
- INSERM U861; I-Stem, Evry Cedex Paris France
- UEVE U861; I-Stem, Evry Cedex Paris France
| | - Isabel Barragan
- Department of Physiology and Pharmacology; Karolinska Institute; Stockholm Sweden
| | | | - Anne-Laure Egesipe
- INSERM U861; I-Stem, Evry Cedex Paris France
- UEVE U861; I-Stem, Evry Cedex Paris France
| | - Laurène Meyniel-Schicklin
- CIRI, International Center for Infectiology Research; Université de Lyon; Lyon France
- INSERM U1111; Lyon France
| | | | - Vincent Lotteau
- CIRI, International Center for Infectiology Research; Université de Lyon; Lyon France
- INSERM U1111; Lyon France
| | | | - Jawida Touhami
- Institut de Génétique Moléculaire de Montpellier, CNRS, UMR5535; Université de Montpellier; Montpellier France
| | - Jean-Luc Battini
- Institut de Génétique Moléculaire de Montpellier, CNRS, UMR5535; Université de Montpellier; Montpellier France
| | - Marc Sitbon
- Institut de Génétique Moléculaire de Montpellier, CNRS, UMR5535; Université de Montpellier; Montpellier France
| | - Christian Pinset
- INSERM U861; I-Stem, Evry Cedex Paris France
- UEVE U861; I-Stem, Evry Cedex Paris France
| | | | | | - Marc Peschanski
- INSERM U861; I-Stem, Evry Cedex Paris France
- UEVE U861; I-Stem, Evry Cedex Paris France
| |
Collapse
|
28
|
Nevalainen M, Metsikkö K. Fluvastatin delays propagation of viral infection in isolated rat FDB myofibers but does not affect exocytic membrane trafficking. Cell Biol Int 2015; 39:1307-16. [PMID: 26123964 DOI: 10.1002/cbin.10509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/25/2015] [Indexed: 11/06/2022]
Abstract
We have utilized the enveloped viral model to study the effect of fluvastatin on membrane trafficking in isolated rat myofibers. Our immunofluorescence studies constantly showed that infections in myofibers, which were treated with fluvastatin prior and during the infection with either vesicular stomatitis virus (VSV) or influenza A virus, propagated more slowly than in control myofibers without drug treatment. Experiments with a virus expressing Dad1 tagged with green fluorescent protein (GFP-Dad1) showed that fluvastatin did not affect its distribution within the ER/SR network and immunofluorescence staining for GM130 did not show any marked effect on the structure of the Golgi components. Furthermore, fluvastatin did not inhibit trafficking of the chimeric transport marker VSV temperature sensitive G protein (tsG-GFP) from the ER to the Golgi. We next subjected VSV infected myofibers for pulse-chase labeling experiments and found that fluvastatin did not slow down the ER-to-Golgi trafficking or Golgi to plasma membrane trafficking of the viral glycoprotein. These studies show that fluvastatin inhibited the propagation of viral infection in skeletal myofibers but no adverse effect on the exocytic trafficking could be demonstrated. These results suggest that other effects of statins rather than inhibition of ER-to-Golgi trafficking might be behind the myotoxic effects of the statins.
Collapse
Affiliation(s)
- Mika Nevalainen
- Division of Cancer Research and Translational Medicine, Department of Anatomy and Cell Biology, Faculty of Medicine, University of Oulu, Oulu, Finland.,Division of Musculoskeletal Imaging and Intervention, Department of Radiology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Kalervo Metsikkö
- Division of Cancer Research and Translational Medicine, Department of Anatomy and Cell Biology, Faculty of Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
29
|
Pellegrini M, Bulzomi P, Galluzzo P, Lecis M, Leone S, Pallottini V, Marino M. Naringenin modulates skeletal muscle differentiation via estrogen receptor α and β signal pathway regulation. GENES AND NUTRITION 2014; 9:425. [PMID: 25156241 DOI: 10.1007/s12263-014-0425-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 07/29/2014] [Indexed: 01/13/2023]
Abstract
Several experiments sustain healthful benefits of the flavanone naringenin (Nar) against chronic diseases including its protective effects against estrogen-related cancers. These experiments encourage Nar use in replacing estrogen treatment in post-menopausal women avoiding the serious side effects ascribed to this hormone. However, at the present, scarce data are available on the impact of Nar on E2-regulated cell functions. This study was aimed at determining the impact of Nar on the estrogen receptor (ERα and β)-dependent signals important for 17β-estradiol (E2) effect in muscle cells (rat L6 myoblasts, mouse C2C12 myoblasts, and mouse skeletal muscle satellite cells). Dietary relevant concentration of Nar delays the appearance of skeletal muscle differentiation markers (i.e., GLUT4 translocation, myogenin, and both fetal and slow MHC isoforms) and impairs E2 effects specifically hampering ERα ability to activate AKT. Intriguingly, Nar effects are specific for E2-initiating signals because IGF-I-induced AKT activation, and myoblast differentiation markers were not affected by Nar treatment. Only 7 days after Nar stimulation, early myoblast differentiation markers (i.e., myogenin, and fetal MHC) start to be accumulated in myoblasts. On the other hand, Nar stimulation activates, via ERβ, the phosphorylation of p38/MAPK involved in reducing the reactive oxygen species formation in skeletal muscle cells. As a whole, data reported here strongly sustain that although Nar action mechanisms include the impairment of ERα signals which drive muscle cells to differentiation, the effects triggered by Nar in the presence of ERβ could balance this negative effect avoiding the toxic effects produced by oxidative stress .
Collapse
Affiliation(s)
- Marco Pellegrini
- Department of Sciences, Biomedical and Technology Science Section, University Roma Tre, Viale G. Marconi 446, 00146, Roma, Italy
| | | | | | | | | | | | | |
Collapse
|