1
|
Tasfaout H, Halbert CL, McMillen TS, Allen JM, Reyes TR, Flint GV, Grimm D, Hauschka SD, Regnier M, Chamberlain JS. Split intein-mediated protein trans-splicing to express large dystrophins. Nature 2024; 632:192-200. [PMID: 39020181 PMCID: PMC11335042 DOI: 10.1038/s41586-024-07710-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/12/2024] [Indexed: 07/19/2024]
Abstract
Gene replacement using adeno-associated virus (AAV) vectors is a promising therapeutic approach for many diseases1,2. However, this therapeutic modality is challenged by the packaging capacity of AAVs (approximately 4.7 kilobases)3, limiting its application for disorders involving large coding sequences, such as Duchenne muscular dystrophy, with a 14 kilobase messenger RNA. Here we developed a new method for expressing large dystrophins by utilizing the protein trans-splicing mechanism mediated by split inteins. We identified several split intein pairs that efficiently join two or three fragments to generate a large midi-dystrophin or the full-length protein. We show that delivery of two or three AAVs into dystrophic mice results in robust expression of large dystrophins and significant physiological improvements compared with micro-dystrophins. Moreover, using the potent myotropic AAVMYO4, we demonstrate that low total doses (2 × 1013 viral genomes per kg) are sufficient to express large dystrophins in striated muscles body-wide with significant physiological corrections in dystrophic mice. Our data show a clear functional superiority of large dystrophins over micro-dystrophins that are being tested in clinical trials. This method could benefit many patients with Duchenne or Becker muscular dystrophy, regardless of genotype, and could be adapted to numerous other disorders caused by mutations in large genes that exceed the AAV capacity.
Collapse
Affiliation(s)
- Hichem Tasfaout
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA.
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, USA.
| | - Christine L Halbert
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, USA
| | - Timothy S McMillen
- Department of Bioengineering, College of Engineering and School of Medicine, University of Washington, Seattle, WA, USA
| | - James M Allen
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, USA
| | - Theodore R Reyes
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, USA
| | - Galina V Flint
- Department of Bioengineering, College of Engineering and School of Medicine, University of Washington, Seattle, WA, USA
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty and Faculty of Engineering Sciences, Center for Integrative Infectious Disease Research (CIID), University of Heidelberg, Heidelberg, Germany
- BioQuant, University of Heidelberg, Heidelberg, Germany
- German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), Heidelberg, Germany
| | - Stephen D Hauschka
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, USA
- Department of Biochemistry, University of Washington School of Medicine, Seattle, WA, USA
| | - Michael Regnier
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, USA
- Department of Bioengineering, College of Engineering and School of Medicine, University of Washington, Seattle, WA, USA
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA
| | - Jeffrey S Chamberlain
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA.
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, USA.
- Department of Biochemistry, University of Washington School of Medicine, Seattle, WA, USA.
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA.
| |
Collapse
|
2
|
Gandhi S, Sweeney HL, Hart CC, Han R, Perry CGR. Cardiomyopathy in Duchenne Muscular Dystrophy and the Potential for Mitochondrial Therapeutics to Improve Treatment Response. Cells 2024; 13:1168. [PMID: 39056750 PMCID: PMC11274633 DOI: 10.3390/cells13141168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive neuromuscular disease caused by mutations to the dystrophin gene, resulting in deficiency of dystrophin protein, loss of myofiber integrity in skeletal and cardiac muscle, and eventual cell death and replacement with fibrotic tissue. Pathologic cardiac manifestations occur in nearly every DMD patient, with the development of cardiomyopathy-the leading cause of death-inevitable by adulthood. As early cardiac abnormalities are difficult to detect, timely diagnosis and appropriate treatment modalities remain a challenge. There is no cure for DMD; treatment is aimed at delaying disease progression and alleviating symptoms. A comprehensive understanding of the pathophysiological mechanisms is crucial to the development of targeted treatments. While established hypotheses of underlying mechanisms include sarcolemmal weakening, upregulation of pro-inflammatory cytokines, and perturbed ion homeostasis, mitochondrial dysfunction is thought to be a potential key contributor. Several experimental compounds targeting the skeletal muscle pathology of DMD are in development, but the effects of such agents on cardiac function remain unclear. The synergistic integration of small molecule- and gene-target-based drugs with metabolic-, immune-, or ion balance-enhancing compounds into a combinatorial therapy offers potential for treating dystrophin deficiency-induced cardiomyopathy, making it crucial to understand the underlying mechanisms driving the disorder.
Collapse
Affiliation(s)
- Shivam Gandhi
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
| | - H. Lee Sweeney
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA; (H.L.S.); (C.C.H.)
- Myology Institute, University of Florida, Gainesville, FL 32610, USA
| | - Cora C. Hart
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA; (H.L.S.); (C.C.H.)
- Myology Institute, University of Florida, Gainesville, FL 32610, USA
| | - Renzhi Han
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Christopher G. R. Perry
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
3
|
Earl CC, Javier AJ, Richards AM, Markham LW, Goergen CJ, Welc SS. Functional cardiac consequences of β-adrenergic stress-induced injury in the mdx mouse model of Duchenne muscular dystrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589650. [PMID: 38659739 PMCID: PMC11042272 DOI: 10.1101/2024.04.15.589650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Cardiomyopathy is the leading cause of death in Duchenne muscular dystrophy (DMD), however, in the mdx mouse model of DMD, the cardiac phenotype differs from that seen in DMD-associated cardiomyopathy. Although some have used pharmacologic stress to enhance the cardiac phenotype in the mdx model, many methods lead to high mortality, variable cardiac outcomes, and do not recapitulate the structural and functional cardiac changes seen in human disease. Here, we describe a simple and effective method to enhance the cardiac phenotype model in mdx mice using advanced 2D and 4D high-frequency ultrasound to monitor cardiac dysfunction progression in vivo. For our study, mdx and wild-type (WT) mice received daily low-dose (2 mg/kg/day) isoproterenol injections for 10 days. Histopathologic assessment showed that isoproterenol treatment increased myocyte injury, elevated serum cardiac troponin I levels, and enhanced fibrosis in mdx mice. Ultrasound revealed reduced ventricular function, decreased wall thickness, increased volumes, and diminished cardiac reserve in mdx mice compared to wild-type. Our findings highlight the utility of low-dose isoproterenol in mdx mice as a valuable model for exploring therapies targeting DMD-associated cardiac complications.
Collapse
Affiliation(s)
- Conner C. Earl
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette IN, USA
- Indiana University School of Medicine, IN, USA
| | - Areli J. Javier
- Musculoskeletal Health Sciences Program, Indiana University School of Medicine, Indianapolis, IN USA
| | - Alyssa M. Richards
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette IN, USA
| | - Larry W. Markham
- Division of Pediatric Cardiology, Riley Children’s Hospital at Indiana University Health, Indiana University School of Medicine, Indianapolis, IN
| | - Craig J. Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette IN, USA
- Indiana University School of Medicine, IN, USA
| | - Steven S. Welc
- Division of Pediatric Cardiology, Riley Children’s Hospital at Indiana University Health, Indiana University School of Medicine, Indianapolis, IN
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis IN, USA
| |
Collapse
|
4
|
Haffner V, Nourian Z, Boerman EM, Lambert MD, Hanft LM, Krenz M, Baines CP, Duan D, McDonald KS, Domeier TL. Calcium handling dysfunction and cardiac damage following acute ventricular preload challenge in the dystrophin-deficient mouse heart. Am J Physiol Heart Circ Physiol 2023; 325:H1168-H1177. [PMID: 37737731 PMCID: PMC10907071 DOI: 10.1152/ajpheart.00265.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023]
Abstract
Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy and is caused by mutations in the dystrophin gene. Dystrophin deficiency is associated with structural and functional changes of the muscle cell sarcolemma and/or stretch-induced ion channel activation. In this investigation, we use mice with transgenic cardiomyocyte-specific expression of the GCaMP6f Ca2+ indicator to test the hypothesis that dystrophin deficiency leads to cardiomyocyte Ca2+ handling abnormalities following preload challenge. α-MHC-MerCreMer-GCaMP6f transgenic mice were developed on both a wild-type (WT) or dystrophic (Dmdmdx-4Cv) background. Isolated hearts of 3-7-mo male mice were perfused in unloaded Langendorff mode (0 mmHg) and working heart mode (preload = 20 mmHg). Following a 30-min preload challenge, hearts were perfused in unloaded Langendorff mode with 40 μM blebbistatin, and GCaMP6f was imaged using confocal fluorescence microscopy. Incidence of premature ventricular complexes (PVCs) was monitored before and following preload elevation at 20 mmHg. Hearts of both wild-type and dystrophic mice exhibited similar left ventricular contractile function. Following preload challenge, dystrophic hearts exhibited a reduction in GCaMP6f-positive cardiomyocytes and an increase in number of cardiomyocytes exhibiting Ca2+ waves/overload. Incidence of cardiac arrhythmias was low in both wild-type and dystrophic hearts during unloaded Langendorff mode. However, after preload elevation to 20-mmHg hearts of dystrophic mice exhibited an increased incidence of PVCs compared with hearts of wild-type mice. In conclusion, these data indicate susceptibility to preload-induced Ca2+ overload, ventricular damage, and ventricular dysfunction in male Dmdmdx-4Cv hearts. Our data support the hypothesis that cardiomyocyte Ca2+ overload underlies cardiac dysfunction in muscular dystrophy.NEW & NOTEWORTHY The mechanisms of cardiac disease progression in muscular dystrophy are complex and poorly understood. Using a transgenic mouse model with cardiomyocyte-specific expression of the GCaMP6f Ca2+ indicator, the present study provides further support for the Ca2+-overload hypothesis of disease progression and ventricular arrhythmogenesis in muscular dystrophy.
Collapse
Affiliation(s)
- Vivian Haffner
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Zahra Nourian
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Erika M Boerman
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Michelle D Lambert
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Laurin M Hanft
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
- The Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Christopher P Baines
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- The Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Kerry S McDonald
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Medicine, School of Medicine, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
5
|
Yang TL, Ting J, Lin MR, Chang WC, Shih CM. Identification of Genetic Variants Associated with Severe Myocardial Bridging through Whole-Exome Sequencing. J Pers Med 2023; 13:1509. [PMID: 37888120 PMCID: PMC10608235 DOI: 10.3390/jpm13101509] [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: 09/06/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
Myocardial bridging (MB) is a congenital coronary artery anomaly and an important cause of angina. The genetic basis of MB is currently unknown. This study used a whole-exome sequencing technique and analyzed genotypic differences. Eight coronary angiography-confirmed cases of severe MB and eight age- and sex-matched control patients were investigated. In total, 139 rare variants that are potentially pathogenic for severe MB were identified in 132 genes. Genes with multiple rare variants or co-predicted by ClinVar and CADD/REVEL for severe MB were collected, from which heart-specific genes were selected under the guidance of tissue expression levels. Functional annotation indicated significant genetic associations with abnormal skeletal muscle mass, cardiomyopathies, and transmembrane ion channels. Candidate genes were reviewed regarding the functions and locations of each individual gene product. Among the gene candidates for severe MB, rare variants in DMD, SGCA, and TTN were determined to be the most crucial. The results suggest that altered anchoring proteins on the cell membrane and intracellular sarcomere unit of cardiomyocytes play a role in the development of the missed trajectory of coronary vessels. Additional studies are required to support the diagnostic application of cardiac sarcoglycan and dystroglycan complexes in patients with severe MB.
Collapse
Affiliation(s)
- Tsung-Lin Yang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Jafit Ting
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan; (J.T.); (M.-R.L.)
| | - Min-Rou Lin
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan; (J.T.); (M.-R.L.)
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan; (J.T.); (M.-R.L.)
- Master’ Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
- Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Chun-Ming Shih
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
6
|
Rok M, Wong TWY, Maino E, Ahmed A, Yang G, Hyatt E, Lindsay K, Fatehi S, Marks R, Delgado-Olguín P, Ivakine EA, Cohn RD. Prevention of early-onset cardiomyopathy in Dmd exon 52-54 deletion mice by CRISPR-Cas9-mediated exon skipping. Mol Ther Methods Clin Dev 2023; 30:246-258. [PMID: 37545481 PMCID: PMC10403712 DOI: 10.1016/j.omtm.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/12/2023] [Indexed: 08/08/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a disease with a life-threatening trajectory resulting from mutations in the dystrophin gene, leading to degeneration of skeletal muscle and fibrosis of cardiac muscle. The overwhelming majority of mutations are multiexonic deletions. We previously established a dystrophic mouse model with deletion of exons 52-54 in Dmd that develops an early-onset cardiac phenotype similar to DMD patients. Here we employed CRISPR-Cas9 delivered intravenously by adeno-associated virus (AAV) vectors to restore functional dystrophin expression via excision or skipping of exon 55. Exon skipping with a solitary guide significantly improved editing outcomes and dystrophin recovery over dual guide excision. Some improvements to genomic and transcript editing levels were observed when the guide dose was enhanced, but dystrophin restoration did not improve considerably. Editing and dystrophin recovery were restricted primarily to cardiac tissue. Remarkably, our exon skipping approach completely prevented onset of the cardiac phenotype in treated mice up to 12 weeks. Thus, our results demonstrate that intravenous delivery of a single-cut CRISPR-Cas9-mediated exon skipping therapy can prevent heart dysfunction in DMD in vivo.
Collapse
Affiliation(s)
- Matthew Rok
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Tatianna Wai Ying Wong
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Eleonora Maino
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Abdalla Ahmed
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Grace Yang
- Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Elzbieta Hyatt
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Kyle Lindsay
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Sina Fatehi
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Ryan Marks
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Paul Delgado-Olguín
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Heart & Stroke Richard Lewar Centre of Excellence, Toronto, ON, Canada
| | - Evgueni A. Ivakine
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Ronald D. Cohn
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
7
|
Hwang HS, Kahmini AR, Prascak J, Cejas-Carbonell A, Valera IC, Champion S, Corrigan M, Mumbi F, Parvatiyar MS. Sarcospan Deficiency Increases Oxidative Stress and Arrhythmias in Hearts after Acute Ischemia-Reperfusion Injury. Int J Mol Sci 2023; 24:11868. [PMID: 37511627 PMCID: PMC10380899 DOI: 10.3390/ijms241411868] [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: 05/27/2023] [Revised: 07/14/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
The protein sarcospan (SSPN) is an integral member of the dystrophin-glycoprotein complex (DGC) and has been shown to be important in the heart during the development and the response to acute stress. In this study, we investigated the role of SSPN in the cardiac response to acute ischemia-reperfusion (IR) injury in SSPN-deficient (SSPN-/-) mice. First, the hemodynamic response of SSPN-/- mice was tested and was similar to SSPN+/+ (wild-type) mice after isoproterenol injection. Using the in situ Langendorff perfusion method, SSPN-/- hearts were subjected to IR injury and found to have increased infarct size and arrhythmia susceptibility compared to SSPN+/+. Ca2+ handling was assessed in single cardiomyocytes and diastolic Ca2+ levels were increased after acute β-AR stimulation in SSPN+/+ but not SSPN-/-. It was also found that SSPN-/- cardiomyocytes had reduced Ca2+ SR content compared to SSPN+/+ but similar SR Ca2+ release. Next, we used qRT-PCR to examine gene expression of Ca2+ handling proteins after acute IR injury. SSPN-/- hearts showed a significant decrease in L-type Ca2+ channels and a significant increase in Ca2+ release channel (RyR2) expression. Interestingly, under oxidizing conditions reminiscent of IR, SSPN-/- cardiomyocytes, had increased H2O2-induced reactive oxygen species production compared to SSPN+/+. Examination of oxidative stress proteins indicated that NADPH oxidase 4 and oxidized CAMKII were increased in SSPN-/- hearts after acute IR injury. These results suggest that increased arrhythmia susceptibility in SSPN-/- hearts post-IR injury may arise from alterations in Ca2+ handling and a reduced capacity to regulate oxidative stress pathways.
Collapse
Affiliation(s)
- Hyun Seok Hwang
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Aida Rahimi Kahmini
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Julia Prascak
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Alexis Cejas-Carbonell
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Isela C Valera
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Samantha Champion
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Mikayla Corrigan
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Florence Mumbi
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Michelle S Parvatiyar
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| |
Collapse
|
8
|
Petrof BJ, Podolsky T, Bhattarai S, Tan J, Ding J. Trained immunity as a potential target for therapeutic immunomodulation in Duchenne muscular dystrophy. Front Immunol 2023; 14:1183066. [PMID: 37398642 PMCID: PMC10309206 DOI: 10.3389/fimmu.2023.1183066] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
Dysregulated inflammation involving innate immune cells, particularly of the monocyte/macrophage lineage, is a key contributor to the pathogenesis of Duchenne muscular dystrophy (DMD). Trained immunity is an evolutionarily ancient protective mechanism against infection, in which epigenetic and metabolic alterations confer non-specific hyperresponsiveness of innate immune cells to various stimuli. Recent work in an animal model of DMD (mdx mice) has shown that macrophages exhibit cardinal features of trained immunity, including the presence of innate immune system "memory". The latter is reflected by epigenetic changes and durable transmissibility of the trained phenotype to healthy non-dystrophic mice by bone marrow transplantation. Mechanistically, it is suggested that a Toll-like receptor (TLR) 4-regulated, memory-like capacity of innate immunity is induced at the level of the bone marrow by factors released from the damaged muscles, leading to exaggerated upregulation of both pro- and anti-inflammatory genes. Here we propose a conceptual framework for the involvement of trained immunity in DMD pathogenesis and its potential to serve as a new therapeutic target.
Collapse
Affiliation(s)
- Basil J. Petrof
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Tom Podolsky
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Salyan Bhattarai
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Jiahui Tan
- Department of Biostatistics and Systems Biology, School of Public Health, Sun Yat-sen University, Shenzhen, China
| | - Jun Ding
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
9
|
Li C, Wilborn J, Pittman S, Daw J, Alonso-Pérez J, Díaz-Manera J, Weihl CC, Haller G. Comprehensive functional characterization of SGCB coding variants predicts pathogenicity in limb-girdle muscular dystrophy type R4/2E. J Clin Invest 2023; 133:e168156. [PMID: 37317968 PMCID: PMC10266784 DOI: 10.1172/jci168156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/27/2023] [Indexed: 06/16/2023] Open
Abstract
Genetic testing is essential for patients with a suspected hereditary myopathy. More than 50% of patients clinically diagnosed with a myopathy carry a variant of unknown significance in a myopathy gene, often leaving them without a genetic diagnosis. Limb-girdle muscular dystrophy (LGMD) type R4/2E is caused by mutations in β-sarcoglycan (SGCB). Together, β-, α-, γ-, and δ-sarcoglycan form a 4-protein transmembrane complex (SGC) that localizes to the sarcolemma. Biallelic loss-of-function mutations in any subunit can lead to LGMD. To provide functional evidence for the pathogenicity of missense variants, we performed deep mutational scanning of SGCB and assessed SGC cell surface localization for all 6,340 possible amino acid changes. Variant functional scores were bimodally distributed and perfectly predicted pathogenicity of known variants. Variants with less severe functional scores more often appeared in patients with slower disease progression, implying a relationship between variant function and disease severity. Amino acid positions intolerant to variation mapped to points of predicted SGC interactions, validated in silico structural models, and enabled accurate prediction of pathogenic variants in other SGC genes. These results will be useful for clinical interpretation of SGCB variants and improving diagnosis of LGMD; we hope they enable wider use of potentially life-saving gene therapy.
Collapse
Affiliation(s)
| | - Jackson Wilborn
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | - Jorge Alonso-Pérez
- Neuromuscular Disease Unit, Neurology Department, Hospital Universitario Nuestra Señora de Candelaria, Fundación Canaria Instituto de Investigación Sanitaria de Canarias, Tenerife, Spain
| | - Jordi Díaz-Manera
- John Walton Muscular Dystrophy Research Center, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | | | - Gabe Haller
- Department of Neurology and
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
10
|
George TG, Hanft LM, Krenz M, Domeier TL, McDonald KS. Dystrophic cardiomyopathy: role of the cardiac myofilaments. Front Physiol 2023; 14:1207658. [PMID: 37362434 PMCID: PMC10288979 DOI: 10.3389/fphys.2023.1207658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023] Open
Abstract
Dystrophic cardiomyopathy arises from mutations in the dystrophin gene. Dystrophin forms part of the dystrophin glycoprotein complex and is postulated to act as a membrane stabilizer, protecting the sarcolemma from contraction-induced damage. Duchenne muscular dystrophy (DMD) is the most severe dystrophinopathy, caused by a total absence of dystrophin. Patients with DMD present with progressive skeletal muscle weakness and, because of treatment advances, a cardiac component of the disease (i.e., dystrophic cardiomyopathy) has been unmasked later in disease progression. The role that myofilaments play in dystrophic cardiomyopathy is largely unknown and, as such, this study aimed to address cardiac myofilament function in a mouse model of muscular dystrophy. To assess the effects of DMD on myofilament function, isolated permeabilized cardiomyocytes of wild-type (WT) littermates and Dmdmdx-4cv mice were attached between a force transducer and motor and subjected to contractile assays. Maximal tension and rates of force development (indexed by the rate constant, k tr) were similar between WT and Dmdmdx-4cv cardiac myocyte preparations. Interestingly, Dmdmdx-4cv cardiac myocytes exhibited greater sarcomere length dependence of peak power output compared to WT myocyte preparations. These results suggest dystrophin mitigates length dependence of activation and, in the absence of dystrophin, augmented sarcomere length dependence of myocyte contractility may accelerate ventricular myocyte contraction-induced damage and contribute to dystrophic cardiomyopathy. Next, we assessed if mavacamten, a small molecule modulator of thick filament activation, would mitigate contractile properties observed in Dmdmdx-4cv permeabilized cardiac myocyte preparations. Mavacamten decreased maximal tension and k tr in both WT and Dmdmdx-4cv cardiac myocytes, while also normalizing the length dependence of peak power between WT and Dmdmdx-4cv cardiac myocyte preparations. These results highlight potential benefits of mavacamten (i.e., reduced contractility while maintaining exquisite sarcomere length dependence of power output) as a treatment for dystrophic cardiomyopathy associated with DMD.
Collapse
|
11
|
Marcadet L, Juracic ES, Khan N, Bouredji Z, Yagita H, Ward LM, Tupling AR, Argaw A, Frenette J. RANKL Inhibition Reduces Cardiac Hypertrophy in mdx Mice and Possibly in Children with Duchenne Muscular Dystrophy. Cells 2023; 12:1538. [PMID: 37296659 PMCID: PMC10253225 DOI: 10.3390/cells12111538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Cardiomyopathy has become one of the leading causes of death in patients with Duchenne muscular dystrophy (DMD). We recently reported that the inhibition of the interaction between the receptor activator of nuclear factor κB ligand (RANKL) and receptor activator of nuclear factor κB (RANK) significantly improves muscle and bone functions in dystrophin-deficient mdx mice. RANKL and RANK are also expressed in cardiac muscle. Here, we investigate whether anti-RANKL treatment prevents cardiac hypertrophy and dysfunction in dystrophic mdx mice. Anti-RANKL treatment significantly reduced LV hypertrophy and heart mass, and maintained cardiac function in mdx mice. Anti-RANKL treatment also inhibited NFκB and PI3K, two mediators implicated in cardiac hypertrophy. Furthermore, anti-RANKL treatment increased SERCA activity and the expression of RyR, FKBP12, and SERCA2a, leading possibly to an improved Ca2+ homeostasis in dystrophic hearts. Interestingly, preliminary post hoc analyses suggest that denosumab, a human anti-RANKL, reduced left ventricular hypertrophy in two patients with DMD. Taken together, our results indicate that anti-RANKL treatment prevents the worsening of cardiac hypertrophy in mdx mice and could potentially maintain cardiac function in teenage or adult patients with DMD.
Collapse
Affiliation(s)
- Laetitia Marcadet
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada; (L.M.); (Z.B.); (A.A.)
| | - Emma Sara Juracic
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, ON N2L 3G1, Canada; (E.S.J.); (A.R.T.)
| | - Nasrin Khan
- The Ottawa Pediatric Bone Health Research Group, Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada; (N.K.); (L.M.W.)
| | - Zineb Bouredji
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada; (L.M.); (Z.B.); (A.A.)
| | - Hideo Yagita
- Department of Immunology, School of Medicine, Juntendo University, Tokyo 113-8421, Japan;
| | - Leanne M. Ward
- The Ottawa Pediatric Bone Health Research Group, Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada; (N.K.); (L.M.W.)
- The Department of Pediatrics, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - A. Russell Tupling
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, ON N2L 3G1, Canada; (E.S.J.); (A.R.T.)
| | - Anteneh Argaw
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada; (L.M.); (Z.B.); (A.A.)
| | - Jérôme Frenette
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada; (L.M.); (Z.B.); (A.A.)
- Department of Rehabilitation, Université Laval, Quebec City, QC G1V 0A6, Canada
| |
Collapse
|
12
|
Uryash A, Mijares A, Estève E, Adams JA, Lopez JR. Smooth Muscle Cells of Dystrophic (mdx) Mice Are More Susceptible to Hypoxia; The Protective Effect of Reducing Ca 2+ Influx. Biomedicines 2023; 11:biomedicines11020623. [PMID: 36831159 PMCID: PMC9953629 DOI: 10.3390/biomedicines11020623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is an inherited muscular disorder caused by mutations in the dystrophin gene. DMD patients have hypoxemic events due to sleep-disordered breathing. We reported an anomalous regulation of resting intracellular Ca2+ ([Ca2+]i) in vascular smooth muscle cells (VSMCs) from a mouse (mdx) model of DMD. We investigated the effect of hypoxia on [Ca2+]i in isolated and quiescent VSMCs from C57BL/10SnJ (WT) and C57BL/10ScSn-Dmd (mdx) male mice. [Ca2+]i was measured using Ca2+-selective microelectrodes under normoxic conditions (95% air, 5% CO2) and after hypoxia (glucose-free solution aerated with 95% N2-5% CO2 for 30 min). [Ca2+]i in mdx VSMCs was significantly elevated compared to WT under normoxia. Hypoxia-induced [Ca2+]i overload, which was significantly greater in mdx than in WT VSMCs. A low Ca2+ solution caused a reduction in [Ca2+]i and prevented [Ca2+]i overload secondary to hypoxia. Nifedipine (10 µM), a Ca2+ channel blocker, did not modify resting [Ca2+]i in VSMCs but partially prevented the hypoxia-induced elevation of [Ca2+]i in both genotypes. SAR7334 (1 µM), an antagonist of TRPC3 and TRPC6, reduced the basal and [Ca2+]i overload caused by hypoxia. Cell viability, assessed by tetrazolium salt (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, was significantly reduced in mdx compared to WT VSMCs. Pretreatment with SAR7341 increases cell viability in normoxic mdx (p < 0.001) and during hypoxia in WT and mdx VSMCs. These results provide evidence that the lack of dystrophin makes VSMCs more susceptible to hypoxia-induced [Ca2+]i overload, which appears to be mediated by increased Ca2+ entry through L-type Ca2+ and TRPC channels.
Collapse
Affiliation(s)
- Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami, FL 33140, USA
| | - Alfredo Mijares
- Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas, Caracas 21827, Venezuela
| | - Eric Estève
- PhyMedExp, University of Montpellier, CNRS, INSERM, CHRU Montpellier, 34090 Montpellier, France
- Univ. Grenoble Alpes, CNRS, TIMC-IMAG/PRETA (UMR 5525), 38000 Grenoble, France
| | - Jose A. Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami, FL 33140, USA
| | - Jose R. Lopez
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
- Correspondence: ; Tel.: +1-305-674-2727
| |
Collapse
|
13
|
Ion Channels of the Sarcolemma and Intracellular Organelles in Duchenne Muscular Dystrophy: A Role in the Dysregulation of Ion Homeostasis and a Possible Target for Therapy. Int J Mol Sci 2023; 24:ijms24032229. [PMID: 36768550 PMCID: PMC9917149 DOI: 10.3390/ijms24032229] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by the absence of the dystrophin protein and a properly functioning dystrophin-associated protein complex (DAPC) in muscle cells. DAPC components act as molecular scaffolds coordinating the assembly of various signaling molecules including ion channels. DMD shows a significant change in the functioning of the ion channels of the sarcolemma and intracellular organelles and, above all, the sarcoplasmic reticulum and mitochondria regulating ion homeostasis, which is necessary for the correct excitation and relaxation of muscles. This review is devoted to the analysis of current data on changes in the structure, functioning, and regulation of the activity of ion channels in striated muscles in DMD and their contribution to the disruption of muscle function and the development of pathology. We note the prospects of therapy based on targeting the channels of the sarcolemma and organelles for the correction and alleviation of pathology, and the problems that arise in the interpretation of data obtained on model dystrophin-deficient objects.
Collapse
|
14
|
Chey YCJ, Arudkumar J, Aartsma-Rus A, Adikusuma F, Thomas PQ. CRISPR applications for Duchenne muscular dystrophy: From animal models to potential therapies. WIREs Mech Dis 2023; 15:e1580. [PMID: 35909075 PMCID: PMC10078488 DOI: 10.1002/wsbm.1580] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/28/2022] [Accepted: 06/30/2022] [Indexed: 01/31/2023]
Abstract
CRISPR gene-editing technology creates precise and permanent modifications to DNA. It has significantly advanced our ability to generate animal disease models for use in biomedical research and also has potential to revolutionize the treatment of genetic disorders. Duchenne muscular dystrophy (DMD) is a monogenic muscle-wasting disease that could potentially benefit from the development of CRISPR therapy. It is commonly associated with mutations that disrupt the reading frame of the DMD gene that encodes dystrophin, an essential scaffolding protein that stabilizes striated muscles and protects them from contractile-induced damage. CRISPR enables the rapid generation of various animal models harboring mutations that closely simulates the wide variety of mutations observed in DMD patients. These models provide a platform for the testing of sequence-specific interventions like CRISPR therapy that aim to reframe or skip DMD mutations to restore functional dystrophin expression. This article is categorized under: Congenital Diseases > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Yu C J Chey
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Jayshen Arudkumar
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Fatwa Adikusuma
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.,CSIRO Synthetic Biology Future Science Platform, Canberra, Australia
| | - Paul Q Thomas
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.,South Australian Genome Editing (SAGE), South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| |
Collapse
|
15
|
The role of the dystrophin glycoprotein complex in muscle cell mechanotransduction. Commun Biol 2022; 5:1022. [PMID: 36168044 PMCID: PMC9515174 DOI: 10.1038/s42003-022-03980-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022] Open
Abstract
Dystrophin is the central protein of the dystrophin-glycoprotein complex (DGC) in skeletal and heart muscle cells. Dystrophin connects the actin cytoskeleton to the extracellular matrix (ECM). Severing the link between the ECM and the intracellular cytoskeleton has a devastating impact on the homeostasis of skeletal muscle cells, leading to a range of muscular dystrophies. In addition, the loss of a functional DGC leads to progressive dilated cardiomyopathy and premature death. Dystrophin functions as a molecular spring and the DGC plays a critical role in maintaining the integrity of the sarcolemma. Additionally, evidence is accumulating, linking the DGC to mechanosignalling, albeit this role is still less understood. This review article aims at providing an up-to-date perspective on the DGC and its role in mechanotransduction. We first discuss the intricate relationship between muscle cell mechanics and function, before examining the recent research for a role of the dystrophin glycoprotein complex in mechanotransduction and maintaining the biomechanical integrity of muscle cells. Finally, we review the current literature to map out how DGC signalling intersects with mechanical signalling pathways to highlight potential future points of intervention, especially with a focus on cardiomyopathies. A review of the function of the Dystrophic Glycoprotein Complex (DGC) in mechanosignaling provides an overview of the various components of DGC and potential mechanopathogenic mechanisms, particularly as they relate to muscular dystrophy.
Collapse
|
16
|
Wu Y, Guo X, Han T, Feng K, Zhang P, Xu Y, Yang Y, Xia Y, Chen Y, Xi J, Yang H, Wan X, Kang J. Cmarr/miR-540-3p axis promotes cardiomyocyte maturation transition by orchestrating Dtna expression. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 29:481-497. [PMID: 36035750 PMCID: PMC9382425 DOI: 10.1016/j.omtn.2022.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 07/20/2022] [Indexed: 10/31/2022]
Abstract
The immature phenotype of embryonic stem cell-derived cardiomyocytes (ESC-CMs) limits their application. However, the molecular mechanisms of cardiomyocyte maturation remain largely unexplored. This study found that overexpression of long noncoding RNA (lncRNA)-Cmarr, which was highly expressed in cardiomyocytes, promoted the maturation change and physiological maturation of mouse ESC-CMs (mESC-CMs). Moreover, transplantation of cardiac patch overexpressing Cmarr exhibited better retention of mESC-CMs, reduced infarct area by enhancing vascular density in the host heart, and improved cardiac function in mice after myocardial infarction. Mechanism studies identified that Cmarr acted as a competitive endogenous RNA to impede the repression of miR-540-3p on Dtna expression and promoted the binding of the dystrophin-glycoprotein complex (DGC) and yes-associated protein (YAP), which in turn reduced the proportion of nuclear YAP and the expression of YAP target genes. Therefore, this study revealed the function and mechanism of Cmarr in promoting cardiomyocyte maturation and provided a lncRNA that can be used as a functional factor in the construction of cardiac patches for the treatment of myocardial infarction.
Collapse
|
17
|
Raccah BH, Biton B, Amir O, Gotsman I, Nahman D, Matok I. Anti-Remodeling Cardiac Therapy in Patients With Duchenne Muscular Dystrophy, Meta-Analysis Study. Front Pharmacol 2022; 12:769896. [PMID: 35126112 PMCID: PMC8811374 DOI: 10.3389/fphar.2021.769896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/21/2021] [Indexed: 12/04/2022] Open
Abstract
Background: Almost all Duchenne muscular dystrophy (DMD) patients that reach their 30s present cardiomyopathy. As a result, this population remains under-treated. There is no sufficient proof of the efficacy of anti-remodeling cardiac therapy for DMD cardiomyopathy (DMDCM). We aim to assess the efficacy of anti-remodeling cardiac therapy for DMDCM by using meta-analysis. Methods: PubMed (MEDLINE), Embase, and Cochrane library were searched through January 2021. Randomized control trials, case-control studies, and observational studies that reported assessments of cardiovascular outcomes and death of participants using angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, beta-blockers, mineralocorticoid-receptor antagonists and Ivabradine, were included. The primary outcome was total mortality. Secondary outcomes included changes in left ventricular ejection fraction (LVEF), serum natriuretic peptide levels (BNP), and heart rate (HR). Data were extracted for eligibility by two independent reviewers. Random-effects meta-analysis was used to pool results. Results: Twelve studies with 439 patients were included in our meta-analysis. Treated patients have lower HR, mean difference of −17 beats per minute (CI [−25]–[−9], p < 0.01). The LVEF was improved in treated patients, with a mean difference of LVEF of 3.77% (CI 0.44–7.12, p < 0.03). Although mortality rates did not reach statistical significance there was a trend for total mortality reduction (hazard ratio 0.36, CI (0.1–1.25), p = 0.107) and for BNP reduction (SSMD: 0.141, CI ([−0.19]–[0.47]), p = 0.3). Conclusion: Pharmacologic treatment for DMDCM patients is associated with decreased HR and improved LVEF. Therefore, DMDCM patients may benefit from implementing guideline therapy for HF.
Collapse
Affiliation(s)
- Bruria Hirsh Raccah
- Division of Clinical Pharmacy, Faculty of Medicine, School of Pharmacy, Institute for Drug Research, the Hebrew University of Jerusalem, Jerusalem, Israel
- Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Bar Biton
- Division of Clinical Pharmacy, Faculty of Medicine, School of Pharmacy, Institute for Drug Research, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Offer Amir
- Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University, Jerusalem, Israel
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Israel Gotsman
- Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Dean Nahman
- Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Ilan Matok
- Division of Clinical Pharmacy, Faculty of Medicine, School of Pharmacy, Institute for Drug Research, the Hebrew University of Jerusalem, Jerusalem, Israel
- *Correspondence: Ilan Matok,
| |
Collapse
|
18
|
Petrof BJ. Macrophage plasticity in Duchenne muscular dystrophy: a nexus of pathological remodelling with therapeutic implications. J Physiol 2021; 600:3455-3464. [PMID: 34904234 DOI: 10.1113/jp281673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/03/2021] [Indexed: 12/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by chronic skeletal muscle necrosis, leading to muscle regeneration failure and fibrosis. Although macrophages (MPs) are normally essential for muscle regeneration, dysregulated MP function promotes pathological muscle remodelling. Infiltrating MPs can be predominantly pro-inflammatory (M1 biased), anti-inflammatory (M2 biased) or of a mixed phenotype and can originate from the adult bone marrow (monocyte dependent) or embryonic precursors (monocyte independent). In mdx mice (genetic model of DMD) lacking either Toll-like receptor (Tlr) 2 or Tlr4, it is found that MP infiltration of dystrophic muscle is significantly reduced and that the MP phenotype is shifted toward a more anti-inflammatory profile. This is accompanied by significant improvements in muscle histology and force production. Lack of the chemokine receptor CCR2, which impedes monocyte release from the bone marrow, leads to similar beneficial effects in mdx mice. Evidence was also found for Tlr4-regulated induction of trained innate immunity in MPs cultured from the bone marrow of mdx mice before their entry into the muscle. These MPs exhibit epigenetic and metabolic alterations, accompanied by non-specific hyper-responsiveness to multiple stimuli, which is manifested by potentiated upregulation of both pro- and anti-inflammatory genes. In summary, exaggerated recruitment of monocyte-derived MPs and signs of trained innate immunity at the level of the bone marrow are features of the immunophenotype associated with dystrophic muscle disease. These phenomena are regulated by Toll-like receptors that bind endogenous damage-associated molecular pattern (DAMP) molecules, suggesting that DAMP release from dystrophic muscles modulates MP plasticity at the bone marrow level through Toll-like receptor-driven mechanisms.
Collapse
Affiliation(s)
- Basil J Petrof
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Respiratory Division, Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
19
|
Cellular pathology of the human heart in Duchenne muscular dystrophy (DMD): lessons learned from in vitro modeling. Pflugers Arch 2021; 473:1099-1115. [DOI: 10.1007/s00424-021-02589-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
|
20
|
Dudley RWR, Comtois AS, St-Pierre DH, Danialou G. Early administration of L-arginine in mdx neonatal mice delays the onset of muscular dystrophy in tibialis anterior (TA) muscle. FASEB Bioadv 2021; 3:639-651. [PMID: 34377959 PMCID: PMC8332474 DOI: 10.1096/fba.2020-00104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 02/17/2021] [Accepted: 04/15/2021] [Indexed: 12/04/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disorder that results in the absence of dystrophin, a cytoskeletal protein. Individuals with this disease experience progressive muscle destruction, which leads to muscle weakness. Studies have been conducted to find solutions for the relief of individuals with this disease, several of which have shown that utrophin, a protein closely related to dystrophin, when overexpressed in mdx neonatal mice (the murine model of DMD), is able to prevent the progressive muscle destruction observed in the absence of dystrophin. Furthermore, recent studies have shown that L‐arginine induces utrophin upregulation in adult mdx mice. We hypothesized that L‐arginine treatment also induces utrophin upregulation to prevent the development of muscle weakness in neonatal mdx mice. Hence, L‐arginine should also prevent progressive muscle destruction via utrophin upregulation in mdx neonatal mice. Mdx neonatal mice were injected intraperitoneally daily with 800 mg/kg of L‐arginine for 6 weeks, whereas control mice were injected with a physiological saline. The following experiments were performed on the tibialis anterior (TA) muscle: muscle contractility and resistance to mechanical stress; central nucleation and peripheral nucleation, utrophin, and creatine kinase quantification as well as a nitric oxide (NO) assay. Our findings show that early administration of L‐arginine in mdx neonatal mice prevents the destruction of the tibialis anterior (TA) muscle. However, this improvement was related to nitric oxide (NO) production rather than the expected utrophin upregulation.
Collapse
Affiliation(s)
- Roy W R Dudley
- Meakins Christie Laboratories McGill University Montreal QC Canada
| | - Alain S Comtois
- Département des Sciences de l'Activité Physique Université du Québec à Montréal (UQAM Montreal QC Canada.,Groupe de Recherche en Activité Physique Adaptée UQAM Montreal QC Canada
| | - David H St-Pierre
- Département des Sciences de l'Activité Physique Université du Québec à Montréal (UQAM Montreal QC Canada.,Groupe de Recherche en Activité Physique Adaptée UQAM Montreal QC Canada.,Centre de Recherche du CHU Sainte-Justine Montréal QC Canada
| | - Gawiyou Danialou
- Meakins Christie Laboratories McGill University Montreal QC Canada.,Département des Sciences de l'Activité Physique Université du Québec à Montréal (UQAM Montreal QC Canada.,Royal Military College Saint-Jean Saint-Jean-sur-Richelieu QC Canada
| |
Collapse
|
21
|
Lopez MA, Bontiff S, Adeyeye M, Shaibani AI, Alexander MS, Wynd S, Boriek AM. Mechanics of dystrophin deficient skeletal muscles in very young mice and effects of age. Am J Physiol Cell Physiol 2021; 321:C230-C246. [PMID: 33979214 DOI: 10.1152/ajpcell.00155.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The MDX mouse is an animal model of Duchenne muscular dystrophy, a human disease marked by an absence of the cytoskeletal protein, dystrophin. We hypothesized that 1) dystrophin serves a complex mechanical role in skeletal muscles by contributing to passive compliance, viscoelastic properties, and contractile force production and 2) age is a modulator of passive mechanics of skeletal muscles of the MDX mouse. Using an in vitro biaxial mechanical testing apparatus, we measured passive length-tension relationships in the muscle fiber direction as well as transverse to the fibers, viscoelastic stress-relaxation curves, and isometric contractile properties. To avoid confounding secondary effects of muscle necrosis, inflammation, and fibrosis, we used very young 3-wk-old mice whose muscles reflected the prefibrotic and prenecrotic state. Compared with controls, 1) muscle extensibility and compliance were greater in both along fiber direction and transverse to fiber direction in MDX mice and 2) the relaxed elastic modulus was greater in dystrophin-deficient diaphragms. Furthermore, isometric contractile muscle stress was reduced in the presence and absence of transverse fiber passive stress. We also examined the effect of age on the diaphragm length-tension relationships and found that diaphragm muscles from 9-mo-old MDX mice were significantly less compliant and less extensible than those of muscles from very young MDX mice. Our data suggest that the age of the MDX mouse is a determinant of the passive mechanics of the diaphragm; in the prefibrotic/prenecrotic stage, muscle extensibility and compliance, as well as viscoelasticity, and muscle contractility are altered by loss of dystrophin.
Collapse
Affiliation(s)
- Michael A Lopez
- Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Pediatrics, University of Alabama Birmingham, Birmingham, Alabama
| | - Sherina Bontiff
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Mary Adeyeye
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Aziz I Shaibani
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Matthew S Alexander
- Department of Pediatrics, University of Alabama Birmingham, Birmingham, Alabama
| | - Shari Wynd
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Aladin M Boriek
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
22
|
Mareedu S, Million ED, Duan D, Babu GJ. Abnormal Calcium Handling in Duchenne Muscular Dystrophy: Mechanisms and Potential Therapies. Front Physiol 2021; 12:647010. [PMID: 33897454 PMCID: PMC8063049 DOI: 10.3389/fphys.2021.647010] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/02/2021] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked muscle-wasting disease caused by the loss of dystrophin. DMD is associated with muscle degeneration, necrosis, inflammation, fatty replacement, and fibrosis, resulting in muscle weakness, respiratory and cardiac failure, and premature death. There is no curative treatment. Investigations on disease-causing mechanisms offer an opportunity to identify new therapeutic targets to treat DMD. An abnormal elevation of the intracellular calcium (Cai2+) concentration in the dystrophin-deficient muscle is a major secondary event, which contributes to disease progression in DMD. Emerging studies have suggested that targeting Ca2+-handling proteins and/or mechanisms could be a promising therapeutic strategy for DMD. Here, we provide an updated overview of the mechanistic roles the sarcolemma, sarcoplasmic/endoplasmic reticulum, and mitochondria play in the abnormal and sustained elevation of Cai2+ levels and their involvement in DMD pathogenesis. We also discuss current approaches aimed at restoring Ca2+ homeostasis as potential therapies for DMD.
Collapse
Affiliation(s)
- Satvik Mareedu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Emily D Million
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, United States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, United States.,Department of Biomedical, Biological & Chemical Engineering, The University of Missouri, Columbia, MO, United States
| | - Gopal J Babu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| |
Collapse
|
23
|
Valera IC, Wacker AL, Hwang HS, Holmes C, Laitano O, Landstrom AP, Parvatiyar MS. Essential roles of the dystrophin-glycoprotein complex in different cardiac pathologies. Adv Med Sci 2021; 66:52-71. [PMID: 33387942 DOI: 10.1016/j.advms.2020.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/12/2020] [Accepted: 12/17/2020] [Indexed: 12/20/2022]
Abstract
The dystrophin-glycoprotein complex (DGC), situated at the sarcolemma dynamically remodels during cardiac disease. This review examines DGC remodeling as a common denominator in diseases affecting heart function and health. Dystrophin and the DGC serve as broad cytoskeletal integrators that are critical for maintaining stability of muscle membranes. The presence of pathogenic variants in genes encoding proteins of the DGC can cause absence of the protein and/or alterations in other complex members leading to muscular dystrophies. Targeted studies have allowed the individual functions of affected proteins to be defined. The DGC has demonstrated its dynamic function, remodeling under a number of conditions that stress the heart. Beyond genetic causes, pathogenic processes also impinge on the DGC, causing alterations in the abundance of dystrophin and associated proteins during cardiac insult such as ischemia-reperfusion injury, mechanical unloading, and myocarditis. When considering new therapeutic strategies, it is important to assess DGC remodeling as a common factor in various heart diseases. The DGC connects the internal F-actin-based cytoskeleton to laminin-211 of the extracellular space, playing an important role in the transmission of mechanical force to the extracellular matrix. The essential functions of dystrophin and the DGC have been long recognized. DGC based therapeutic approaches have been primarily focused on muscular dystrophies, however it may be a beneficial target in a number of disorders that affect the heart. This review provides an account of what we now know, and discusses how this knowledge can benefit persistent health conditions in the clinic.
Collapse
Affiliation(s)
- Isela C Valera
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Amanda L Wacker
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Hyun Seok Hwang
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Christina Holmes
- Department of Chemical and Biomedical Engineering, Florida A&M University-Florida State University College of Engineering, Tallahassee, FL, USA
| | - Orlando Laitano
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Andrew P Landstrom
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, USA; Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Michelle S Parvatiyar
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
24
|
Kincl V, Panovský R, Pešl M, Máchal J, Juříková L, Haberlová J, Masárová L. Echocardiographic signs of subclinical cardiac function impairment in Duchenne dystrophy gene carriers. Sci Rep 2020; 10:20794. [PMID: 33247228 PMCID: PMC7695725 DOI: 10.1038/s41598-020-77882-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 11/17/2020] [Indexed: 11/16/2022] Open
Abstract
To assess subclinical cardiac function impairment in Duchenne dystrophy (DMD) female carriers. Forty-four female subjects proved as DMD carriers underwent echocardiographic examination including tissue Doppler imaging (TDI) of mitral and tricuspid annulus. Seventeen age-matched healthy female subjects served as controls. A significant differences in peak systolic annular velocity (Sa) between carriers and controls were found for lateral and septal part of the mitral annulus and for tricuspid annulus (0.09 vs. 0.11 m/s, p < 0.001, 0.08 vs. 0.09 m/s, p < 0.01 and 0.13 vs. 0.14 m/s, p = 0.02 respectively). There was also difference in early diastolic velocity (Ea) of the septal part of the mitral annulus (0.11 vs. 0.13 m/s, p = 0.03). The subclinical deterioration of systolic function is presented even in asymptomatic DMD female carriers.
Collapse
Affiliation(s)
- Vladimír Kincl
- Department of Internal Medicine/Cardiology, Faculty of Medicine, St. Anne's University Hospital, Masaryk University, Brno, Czech Republic. .,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
| | - Roman Panovský
- Department of Internal Medicine/Cardiology, Faculty of Medicine, St. Anne's University Hospital, Masaryk University, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Martin Pešl
- Department of Internal Medicine/Cardiology, Faculty of Medicine, St. Anne's University Hospital, Masaryk University, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.,Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Máchal
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.,Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lenka Juříková
- Department of Pediatric Neurology, Faculty of Medicine, University Hospital Brno, Masaryk University, Brno, Czech Republic
| | - Jana Haberlová
- Department of Pediatric Neurology, Second Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Lucia Masárová
- Department of Internal Medicine/Cardiology, Faculty of Medicine, St. Anne's University Hospital, Masaryk University, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| |
Collapse
|
25
|
Pecorari I, Mestroni L, Sbaizero O. Current Understanding of the Role of Cytoskeletal Cross-Linkers in the Onset and Development of Cardiomyopathies. Int J Mol Sci 2020; 21:E5865. [PMID: 32824180 PMCID: PMC7461563 DOI: 10.3390/ijms21165865] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 07/28/2020] [Accepted: 08/10/2020] [Indexed: 01/08/2023] Open
Abstract
Cardiomyopathies affect individuals worldwide, without regard to age, sex and ethnicity and are associated with significant morbidity and mortality. Inherited cardiomyopathies account for a relevant part of these conditions. Although progresses have been made over the years, early diagnosis and curative therapies are still challenging. Understanding the events occurring in normal and diseased cardiac cells is crucial, as they are important determinants of overall heart function. Besides chemical and molecular events, there are also structural and mechanical phenomena that require to be investigated. Cell structure and mechanics largely depend from the cytoskeleton, which is composed by filamentous proteins that can be cross-linked via accessory proteins. Alpha-actinin 2 (ACTN2), filamin C (FLNC) and dystrophin are three major actin cross-linkers that extensively contribute to the regulation of cell structure and mechanics. Hereby, we review the current understanding of the roles played by ACTN2, FLNC and dystrophin in the onset and progress of inherited cardiomyopathies. With our work, we aim to set the stage for new approaches to study the cardiomyopathies, which might reveal new therapeutic targets and broaden the panel of genes to be screened.
Collapse
Affiliation(s)
- Ilaria Pecorari
- Department of Engineering and Architecture, University of Trieste, 34127 Trieste, Italy;
| | - Luisa Mestroni
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, 34127 Trieste, Italy;
| |
Collapse
|
26
|
Garbincius JF, Merz LE, Cuttitta AJ, Bayne KV, Schrade S, Armstead EA, Converso-Baran KL, Whitesall SE, D'Alecy LG, Michele DE. Enhanced dimethylarginine degradation improves coronary flow reserve and exercise tolerance in Duchenne muscular dystrophy carrier mice. Am J Physiol Heart Circ Physiol 2020; 319:H582-H603. [PMID: 32762558 DOI: 10.1152/ajpheart.00333.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked disease caused by null mutations in dystrophin and characterized by muscle degeneration. Cardiomyopathy is common and often prevalent at similar frequency in female DMD carriers irrespective of whether they manifest skeletal muscle disease. Impaired muscle nitric oxide (NO) production in DMD disrupts muscle blood flow regulation and exaggerates postexercise fatigue. We show that circulating levels of endogenous methylated arginines including asymmetric dimethylarginine (ADMA), which act as NO synthase inhibitors, are elevated by acute necrotic muscle damage and in chronically necrotic dystrophin-deficient mice. We therefore hypothesized that excessive ADMA impairs muscle NO production and diminishes exercise tolerance in DMD. We used transgenic expression of dimethylarginine dimethylaminohydrolase 1 (DDAH), which degrades methylated arginines, to investigate their contribution to exercise-induced fatigue in DMD. Although infusion of exogenous ADMA was sufficient to impair exercise performance in wild-type mice, transgenic DDAH expression did not rescue exercise-induced fatigue in dystrophin-deficient male mdx mice. Surprisingly, DDAH transgene expression did attenuate exercise-induced fatigue in dystrophin-heterozygous female mdx carrier mice. Improved exercise tolerance was associated with reduced heart weight and improved cardiac β-adrenergic responsiveness in DDAH-transgenic mdx carriers. We conclude that DDAH overexpression increases exercise tolerance in female DMD carriers, possibly by limiting cardiac pathology and preserving the heart's responses to changes in physiological demand. Methylated arginine metabolism may be a new target to improve exercise tolerance and cardiac function in DMD carriers or act as an adjuvant to promote NO signaling alongside therapies that partially restore dystrophin expression in patients with DMD.NEW & NOTEWORTHY Duchenne muscular dystrophy (DMD) carriers are at risk for cardiomyopathy. The nitric oxide synthase inhibitor asymmetric dimethylarginine (ADMA) is released from damaged muscle in DMD and impairs exercise performance. Transgenic expression of dimethylarginine dimethylaminohydrolase to degrade ADMA prevents cardiac hypertrophy, improves cardiac function, and improves exercise tolerance in DMD carrier mice. These findings highlight the relevance of ADMA to muscular dystrophy and have important implications for therapies targeting nitric oxide in patients with DMD and DMD carriers.
Collapse
Affiliation(s)
- Joanne F Garbincius
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Lauren E Merz
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Ashley J Cuttitta
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Kaitlynn V Bayne
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Sara Schrade
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Emily A Armstead
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | | | - Steven E Whitesall
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan.,Physiology Phenotyping Core, University of Michigan, Ann Arbor, Michigan
| | - Louis G D'Alecy
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan.,Physiology Phenotyping Core, University of Michigan, Ann Arbor, Michigan
| | - Daniel E Michele
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan.,Physiology Phenotyping Core, University of Michigan, Ann Arbor, Michigan.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
27
|
Jelinkova S, Vilotic A, Pribyl J, Aimond F, Salykin A, Acimovic I, Pesl M, Caluori G, Klimovic S, Urban T, Dobrovolna H, Soska V, Skladal P, Lacampagne A, Dvorak P, Meli AC, Rotrekl V. DMD Pluripotent Stem Cell Derived Cardiac Cells Recapitulate in vitro Human Cardiac Pathophysiology. Front Bioeng Biotechnol 2020; 8:535. [PMID: 32656189 PMCID: PMC7325914 DOI: 10.3389/fbioe.2020.00535] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 05/04/2020] [Indexed: 12/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe genetic disorder characterized by the lack of functional dystrophin. DMD is associated with progressive dilated cardiomyopathy, eventually leading to heart failure as the main cause of death in DMD patients. Although several molecular mechanisms leading to the DMD cardiomyocyte (DMD-CM) death were described, mostly in mouse model, no suitable human CM model was until recently available together with proper clarification of the DMD-CM phenotype and delay in cardiac symptoms manifestation. We obtained several independent dystrophin-deficient human pluripotent stem cell (hPSC) lines from DMD patients and CRISPR/Cas9-generated DMD gene mutation. We differentiated DMD-hPSC into cardiac cells (CC) creating a human DMD-CC disease model. We observed that mutation-carrying cells were less prone to differentiate into CCs. DMD-CCs demonstrated an enhanced cell death rate in time. Furthermore, ion channel expression was altered in terms of potassium (Kir2.1 overexpression) and calcium handling (dihydropyridine receptor overexpression). DMD-CCs exhibited increased time of calcium transient rising compared to aged-matched control, suggesting mishandling of calcium release. We observed mechanical impairment (hypocontractility), bradycardia, increased heart rate variability, and blunted β-adrenergic response connected with remodeling of β-adrenergic receptors expression in DMD-CCs. Overall, these results indicated that our DMD-CC models are functionally affected by dystrophin-deficiency associated and recapitulate functional defects and cardiac wasting observed in the disease. It offers an accurate tool to study human cardiomyopathy progression and test therapies in vitro.
Collapse
Affiliation(s)
- Sarka Jelinkova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Center ICRC, St. Anne's University Hospital Brno, Brno, Czechia
| | - Aleksandra Vilotic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Jan Pribyl
- CEITEC, Masaryk University, Brno, Czechia
| | - Franck Aimond
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Anton Salykin
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Ivana Acimovic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Martin Pesl
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Center ICRC, St. Anne's University Hospital Brno, Brno, Czechia.,First Department of Internal Medicine-Cardioangiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Guido Caluori
- International Clinical Research Center ICRC, St. Anne's University Hospital Brno, Brno, Czechia.,First Department of Internal Medicine-Cardioangiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Simon Klimovic
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czechia
| | - Tomas Urban
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Hana Dobrovolna
- Department of Clinical Biochemistry, St. Anne's University Hospital of Brno, Brno, Czechia
| | - Vladimir Soska
- Department of Clinical Biochemistry, St. Anne's University Hospital of Brno, Brno, Czechia.,Second Clinic of Internal Medicine, Masaryk University of Brno, Brno, Czechia
| | - Petr Skladal
- First Department of Internal Medicine-Cardioangiology, Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czechia
| | - Alain Lacampagne
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Petr Dvorak
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Center ICRC, St. Anne's University Hospital Brno, Brno, Czechia
| | - Albano C Meli
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia.,PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Vladimir Rotrekl
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Center ICRC, St. Anne's University Hospital Brno, Brno, Czechia
| |
Collapse
|
28
|
Lopez JR, Uryash A, Faury G, Estève E, Adams JA. Contribution of TRPC Channels to Intracellular Ca 2 + Dyshomeostasis in Smooth Muscle From mdx Mice. Front Physiol 2020; 11:126. [PMID: 32153426 PMCID: PMC7044154 DOI: 10.3389/fphys.2020.00126] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/04/2020] [Indexed: 12/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an irreversible muscle disease characterized by a progressive loss of muscle function, decreased ambulation, and ultimately death as a result of cardiac or respiratory failure. DMD is caused by the lack of dystrophin, a protein that is important for membrane stability and signaling in excitable cells. Although vascular smooth muscle cells (VSMCs) dysfunction occurs in many pathological conditions, little is known about vascular smooth muscle function in DMD. We have previously shown that striated muscle cells, as well as neurons isolated from dystrophic (mdx) mice have higher intracellular Ca2+ ([Ca2+]i) and Na+ ([Na+]i) concentrations and decreased cell viability in comparison with wild type (Wt). Experiments were carried out in isolated VSMCs from mdx (a murine model of DMD) and congenic C57BL/10SnJ Wt mice. We found elevated [Ca2+]i and [Na+]i in VSMCs from mdx mice compared to Wt. Exposure to 1-oleoyl-2-acetyl-sn-glycerol (OAG), a TRPC3 and TRPC6 channel activator, induced a greater elevation of [Ca2+]i and [Na+]i in mdx than Wt VSMCs. The OAG induced increases in [Ca2+]i could be abolished by either removal of extracellular Ca2+ or by SAR7334, a blocker of TRPC3 and TRPC 6 channels in both genotypes. Mdx and Wt VSMCs were susceptible to muscle cell stretch-induced elevations of [Ca2+]i and [Na+]i which was completely inhibited by GsMTx-4, a mechanosensitive ion channel inhibitor. Western blots showed a significant upregulation of TRPC1 -3, -6 proteins in mdx VSMCs compare to age-matched Wt. The lack of dystrophin in mdx VSMCs produced a profound alteration of [Ca2+]i and [Na+]i homeostasis that appears to be mediated by TRPC channels. Moreover, we have been able to demonstrate pharmacologically that the enhanced stretch-induced elevation of intracellular [Ca2+] and concomitant cell damage in mdx VSMCs also appears to be mediated through TRPC1, -3 and -6 channel activation.
Collapse
Affiliation(s)
- Jose R Lopez
- Department of Research, Mount Sinai Medical Center, Miami, FL, United States
| | - Arkady Uryash
- Department of Neonatology, Mount Sinai Medical Center, Miami, FL, United States
| | - Gilles Faury
- HP2, CHU Grenoble Alpes, Inserm, University Grenoble Alpes, Grenoble, France
| | - Eric Estève
- HP2, CHU Grenoble Alpes, Inserm, University Grenoble Alpes, Grenoble, France
| | - Jose A Adams
- Department of Neonatology, Mount Sinai Medical Center, Miami, FL, United States
| |
Collapse
|
29
|
Lamhonwah AM, Tein I. Expression of the organic cation/carnitine transporter family (Octn1,-2 and-3) in mdx muscle and heart: Implications for early carnitine therapy in Duchenne muscular dystrophy to improve cellular carnitine homeostasis. Clin Chim Acta 2020; 505:92-97. [PMID: 32070725 DOI: 10.1016/j.cca.2020.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Carnitine is essential for long-chain fatty acid oxidation in muscle and heart. Tissue stores are regulated by organic cation/Cn transporter plasmalemmal Octn2. We previously demonstrated low carnitine in quadriceps/gluteus and heart of adult mdx mice. METHODS We studied protein and mRNA expression of Octn2, mitochondrial Octn1 and peroxisomal Octn3 in adult male C57BL/10ScSn-DMD mdx/J quadriceps, heart, and diaphragm compared to C57BL/10SnJ mice. RESULTS We demonstrated reduction in mOctn2 expression on Western blot and similar expression of mOctn1 and mOctn3 in mdx quadriceps, heart and diaphragm. There was a significant upregulation of mOctn1 and mOctn2 mRNA by qRT-PCR in mdx quadriceps and of mOctn2 and mOctn3 mRNA in mdx heart. We showed upregulation of mdx mOctn1 and mOctn3 mRNA but no increase in protein expression. DISCUSSION Dystrophin deficiency likely disrupts Octn2 expression decreasing muscle carnitine uptake thus contributing to membranotoxic long-chain acyl-CoAs with sarcolemmal and organellar membrane oxidative injury providing a treatment rationale for early L-carnitine in DMD.
Collapse
Affiliation(s)
- Anne-Marie Lamhonwah
- Department of Pediatrics, Division of Neurology, Hospital for Sick Children, University of Toronto, 555 University, Ave., Toronto, Ontario M5G 1X8, Canada; Genetics and Genome Biology Program, The Research Institute, Hospital for Sick Children, University of Toronto, Toronto, Ontario M5G 1X8, Canada
| | - Ingrid Tein
- Department of Pediatrics, Division of Neurology, Hospital for Sick Children, University of Toronto, 555 University, Ave., Toronto, Ontario M5G 1X8, Canada; Genetics and Genome Biology Program, The Research Institute, Hospital for Sick Children, University of Toronto, Toronto, Ontario M5G 1X8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A1, Canada.
| |
Collapse
|
30
|
Law ML, Cohen H, Martin AA, Angulski ABB, Metzger JM. Dysregulation of Calcium Handling in Duchenne Muscular Dystrophy-Associated Dilated Cardiomyopathy: Mechanisms and Experimental Therapeutic Strategies. J Clin Med 2020; 9:jcm9020520. [PMID: 32075145 PMCID: PMC7074327 DOI: 10.3390/jcm9020520] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
: Duchenne muscular dystrophy (DMD) is an X-linked recessive disease resulting in the loss of dystrophin, a key cytoskeletal protein in the dystrophin-glycoprotein complex. Dystrophin connects the extracellular matrix with the cytoskeleton and stabilizes the sarcolemma. Cardiomyopathy is prominent in adolescents and young adults with DMD, manifesting as dilated cardiomyopathy (DCM) in the later stages of disease. Sarcolemmal instability, leading to calcium mishandling and overload in the cardiac myocyte, is a key mechanistic contributor to muscle cell death, fibrosis, and diminished cardiac contractile function in DMD patients. Current therapies for DMD cardiomyopathy can slow disease progression, but they do not directly target aberrant calcium handling and calcium overload. Experimental therapeutic targets that address calcium mishandling and overload include membrane stabilization, inhibition of stretch-activated channels, ryanodine receptor stabilization, and augmentation of calcium cycling via modulation of the Serca2a/phospholamban (PLN) complex or cytosolic calcium buffering. This paper addresses what is known about the mechanistic basis of calcium mishandling in DCM, with a focus on DMD cardiomyopathy. Additionally, we discuss currently utilized therapies for DMD cardiomyopathy, and review experimental therapeutic strategies targeting the calcium handling defects in DCM and DMD cardiomyopathy.
Collapse
Affiliation(s)
- Michelle L. Law
- Department of Family and Consumer Sciences, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA;
| | - Houda Cohen
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Ashley A. Martin
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Addeli Bez Batti Angulski
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
- Correspondence: ; Tel.: +1-612-625-5902; Fax: +1-612-625-5149
| |
Collapse
|
31
|
Esposito G, Carsana A. Metabolic Alterations in Cardiomyocytes of Patients with Duchenne and Becker Muscular Dystrophies. J Clin Med 2019; 8:jcm8122151. [PMID: 31817415 PMCID: PMC6947625 DOI: 10.3390/jcm8122151] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/22/2019] [Accepted: 12/02/2019] [Indexed: 12/14/2022] Open
Abstract
Duchenne and Becker muscular dystrophies (DMD/BMD) result in progressive weakness of skeletal and cardiac muscles due to the deficiency of functional dystrophin. Respiratory failure is a leading cause of mortality in DMD patients; however, improved management of the respiratory symptoms have increased patients' life expectancy, thereby also increasing the clinical relevance of heart disease. In fact, the prevalence of cardiomyopathy, which significantly contributes to mortality in DMD patients, increases with age and disease progression, so that over 95% of adult patients has cardiomyopathy signs. We here review the current literature featuring the metabolic alterations observed in the dystrophic heart of the mdx mouse, i.e., the best-studied animal model of the disease, and discuss their pathophysiological role in the DMD heart. It is well assessed that dystrophin deficiency is associated with pathological alterations of lipid metabolism, intracellular calcium levels, neuronal nitric oxide (NO) synthase localization, and NO and reactive oxygen species production. These metabolic stressors contribute to impair the function of the cardiac mitochondrial bulk, which has a relevant pathophysiological role in the development of cardiomyopathy. In fact, mitochondrial dysfunction becomes more severe as the dystrophic process progresses, thereby indicating it may be both the cause and the consequence of the dystrophic process in the DMD heart.
Collapse
Affiliation(s)
- Gabriella Esposito
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy;
- CEINGE Advanced Biotechnologies, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Antonella Carsana
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy;
- Correspondence:
| |
Collapse
|
32
|
Lionarons JM, Hoogland G, Hendriksen RGF, Faber CG, Hellebrekers DMJ, Van Koeveringe GA, Schipper S, Vles JSH. Dystrophin is expressed in smooth muscle and afferent nerve fibers in the rat urinary bladder. Muscle Nerve 2019; 60:202-210. [PMID: 31095755 PMCID: PMC6771971 DOI: 10.1002/mus.26518] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 05/08/2019] [Accepted: 05/11/2019] [Indexed: 11/10/2022]
Abstract
INTRODUCTION With increasing life expectancy, comorbidities become overt in Duchenne muscular dystrophy (DMD). Although micturition problems are common, bladder function is poorly understood in DMD. We studied dystrophin expression and multiple isoform involvement in the bladder during maturation to gain insights into their roles in micturition. METHODS Dystrophin distribution was evaluated in rat bladders by immunohistochemical colocalization with smooth muscle, interstitial, urothelial, and neuronal markers. Protein levels of Dp140, Dp71, and smooth muscle were quantitated by Western blotting of neonatal to adult rat bladders. RESULTS Dystrophin colocalized with smooth muscle cells and afferent nerve fibers. Dp71 was expressed two- to threefold higher compared with Dp140, independently of age. Age-related muscle mass changes did not influence isoform expression levels. DISCUSSION Dystrophin is expressed in smooth muscle cells and afferent nerve fibers in the urinary bladder, which underscores that micturition problems in DMD may have not solely a myogenic but also a neurogenic origin. Muscle Nerve 60: 202-210, 2019.
Collapse
Affiliation(s)
- Judith M Lionarons
- Department of Neurology, Maastricht University Medical Center, PO Box 5800, 6202 AZ Maastricht, The Netherlands.,School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Govert Hoogland
- School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ruben G F Hendriksen
- Department of Neurology, Maastricht University Medical Center, PO Box 5800, 6202 AZ Maastricht, The Netherlands.,School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Catharina G Faber
- Department of Neurology, Maastricht University Medical Center, PO Box 5800, 6202 AZ Maastricht, The Netherlands.,School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Danique M J Hellebrekers
- Department of Neurology, Maastricht University Medical Center, PO Box 5800, 6202 AZ Maastricht, The Netherlands.,School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Gommert A Van Koeveringe
- School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Urology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Sandra Schipper
- School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Urology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Johan S H Vles
- Department of Neurology, Maastricht University Medical Center, PO Box 5800, 6202 AZ Maastricht, The Netherlands.,School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
33
|
Various effects of AAV9-mediated βARKct gene therapy on the heart in dystrophin-deficient (mdx) mice and δ-sarcoglycan-deficient (Sgcd-/-) mice. Neuromuscul Disord 2019; 29:231-241. [DOI: 10.1016/j.nmd.2018.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 10/21/2018] [Accepted: 12/16/2018] [Indexed: 01/08/2023]
|
34
|
Heier CR, Yu Q, Fiorillo AA, Tully CB, Tucker A, Mazala DA, Uaesoontrachoon K, Srinivassane S, Damsker JM, Hoffman EP, Nagaraju K, Spurney CF. Vamorolone targets dual nuclear receptors to treat inflammation and dystrophic cardiomyopathy. Life Sci Alliance 2019; 2:2/1/e201800186. [PMID: 30745312 PMCID: PMC6371196 DOI: 10.26508/lsa.201800186] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/25/2022] Open
Abstract
Cardiomyopathy is a leading cause of death for Duchenne muscular dystrophy. Here, we find that the mineralocorticoid receptor (MR) and glucocorticoid receptor (GR) can share common ligands but play distinct roles in dystrophic heart and skeletal muscle pathophysiology. Comparisons of their ligand structures indicate that the Δ9,11 modification of the first-in-class drug vamorolone enables it to avoid interaction with a conserved receptor residue (N770/N564), which would otherwise activate transcription factor properties of both receptors. Reporter assays show that vamorolone and eplerenone are MR antagonists, whereas prednisolone is an MR agonist. Macrophages, cardiomyocytes, and CRISPR knockout myoblasts show vamorolone is also a dissociative GR ligand that inhibits inflammation with improved safety over prednisone and GR-specific deflazacort. In mice, hyperaldosteronism activates MR-driven hypertension and kidney phenotypes. We find that genetic dystrophin loss provides a second hit for MR-mediated cardiomyopathy in Duchenne muscular dystrophy model mice, as aldosterone worsens fibrosis, mass and dysfunction phenotypes. Vamorolone successfully prevents MR-activated phenotypes, whereas prednisolone activates negative MR and GR effects. In conclusion, vamorolone targets dual nuclear receptors to treat inflammation and cardiomyopathy with improved safety.
Collapse
Affiliation(s)
- Christopher R Heier
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA .,Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA
| | - Qing Yu
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA
| | - Alyson A Fiorillo
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.,Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA
| | - Christopher B Tully
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA
| | - Asya Tucker
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA
| | - Davi A Mazala
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA
| | | | | | | | - Eric P Hoffman
- AGADA Biosciences Incorporated, Halifax, Nova Scotia, Canada.,ReveraGen BioPharma, Incorporated, Rockville, MD, USA.,School of Pharmacy and Pharmaceutical Sciences, Binghamton University-State University of New York (SUNY), Binghamton, NY, USA
| | - Kanneboyina Nagaraju
- AGADA Biosciences Incorporated, Halifax, Nova Scotia, Canada.,ReveraGen BioPharma, Incorporated, Rockville, MD, USA.,School of Pharmacy and Pharmaceutical Sciences, Binghamton University-State University of New York (SUNY), Binghamton, NY, USA
| | - Christopher F Spurney
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.,Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA.,Division of Cardiology, Children's National Heart Institute, Children's National Medical Center, Washington, DC, USA
| |
Collapse
|
35
|
Jelinkova S, Fojtik P, Kohutova A, Vilotic A, Marková L, Pesl M, Jurakova T, Kruta M, Vrbsky J, Gaillyova R, Valášková I, Frák I, Lacampagne A, Forte G, Dvorak P, Meli AC, Rotrekl V. Dystrophin Deficiency Leads to Genomic Instability in Human Pluripotent Stem Cells via NO Synthase-Induced Oxidative Stress. Cells 2019; 8:cells8010053. [PMID: 30650618 PMCID: PMC6356905 DOI: 10.3390/cells8010053] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/29/2018] [Accepted: 01/11/2019] [Indexed: 11/16/2022] Open
Abstract
Recent data on Duchenne muscular dystrophy (DMD) show myocyte progenitor's involvement in the disease pathology often leading to the DMD patient's death. The molecular mechanism underlying stem cell impairment in DMD has not been described. We created dystrophin-deficient human pluripotent stem cell (hPSC) lines by reprogramming cells from two DMD patients, and also by introducing dystrophin mutation into human embryonic stem cells via CRISPR/Cas9. While dystrophin is expressed in healthy hPSC, its deficiency in DMD hPSC lines induces the release of reactive oxygen species (ROS) through dysregulated activity of all three isoforms of nitric oxide synthase (further abrev. as, NOS). NOS-induced ROS release leads to DNA damage and genomic instability in DMD hPSC. We were able to reduce both the ROS release as well as DNA damage to the level of wild-type hPSC by inhibiting NOS activity.
Collapse
Affiliation(s)
- Sarka Jelinkova
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
- International Clinical Research Center ICRC, St. Anne's University Hospital Brno, 602 00 Brno, Czech Republic.
| | - Petr Fojtik
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
| | - Aneta Kohutova
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
- International Clinical Research Center ICRC, St. Anne's University Hospital Brno, 602 00 Brno, Czech Republic.
| | - Aleksandra Vilotic
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
| | - Lenka Marková
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
| | - Martin Pesl
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
- International Clinical Research Center ICRC, St. Anne's University Hospital Brno, 602 00 Brno, Czech Republic.
- 1st department of Internal Medicine-Cardioangiology, Faculty of Medicine, Masaryk University, 602 00 Brno, Czech Republic.
| | - Tereza Jurakova
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
| | - Miriama Kruta
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
| | - Jan Vrbsky
- International Clinical Research Center ICRC, St. Anne's University Hospital Brno, 602 00 Brno, Czech Republic.
| | - Renata Gaillyova
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
- Department of Clinical Genetics, University hospital Brno, 613 00 Brno, Czech Republic.
| | - Iveta Valášková
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
- Department of Clinical Genetics, University hospital Brno, 613 00 Brno, Czech Republic.
| | - Ivan Frák
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
| | - Alain Lacampagne
- PhyMedExp, INSERM, University of Montpellier, CNRS, 342 95 Montpellier CEDEX 5, France.
| | - Giancarlo Forte
- International Clinical Research Center ICRC, St. Anne's University Hospital Brno, 602 00 Brno, Czech Republic.
| | - Petr Dvorak
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
- International Clinical Research Center ICRC, St. Anne's University Hospital Brno, 602 00 Brno, Czech Republic.
| | - Albano C Meli
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
- PhyMedExp, INSERM, University of Montpellier, CNRS, 342 95 Montpellier CEDEX 5, France.
| | - Vladimir Rotrekl
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
- International Clinical Research Center ICRC, St. Anne's University Hospital Brno, 602 00 Brno, Czech Republic.
| |
Collapse
|
36
|
Farini A, Gowran A, Bella P, Sitzia C, Scopece A, Castiglioni E, Rovina D, Nigro P, Villa C, Fortunato F, Comi GP, Milano G, Pompilio G, Torrente Y. Fibrosis Rescue Improves Cardiac Function in Dystrophin-Deficient Mice and Duchenne Patient-Specific Cardiomyocytes by Immunoproteasome Modulation. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 189:339-353. [PMID: 30448404 DOI: 10.1016/j.ajpath.2018.10.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 09/12/2018] [Accepted: 10/10/2018] [Indexed: 11/30/2022]
Abstract
Patients affected by Duchenne muscular dystrophy (DMD) develop a progressive dilated cardiomyopathy characterized by inflammatory cell infiltration, necrosis, and cardiac fibrosis. Standard treatments consider the use of β-blockers and angiotensin-converting enzyme inhibitors that are symptomatic and unspecific toward DMD disease. Medications that target DMD cardiac fibrosis are in the early stages of development. We found immunoproteasome dysregulation in affected hearts of mdx mice (murine animal model of DMD) and cardiomyocytes derived from induced pluripotent stem cells of patients with DMD. Interestingly, immunoproteasome inhibition ameliorated cardiomyopathy in mdx mice and reduced the development of cardiac fibrosis. Establishing the immunoproteasome inhibition-dependent cardioprotective role suggests the possibility of modulating the immunoproteasome as new and clinically relevant treatment to rescue dilated cardiomyopathy in patients with DMD.
Collapse
Affiliation(s)
- Andrea Farini
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Universitá degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Aoife Gowran
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Pamela Bella
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Universitá degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Clementina Sitzia
- UOC SMEL-1, Scuola di Specializzazione di Patologia Clinica e Biochimica Clinica, Università degli Studi di Milano, Milan, Italy
| | - Alessandro Scopece
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Elisa Castiglioni
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Davide Rovina
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Patrizia Nigro
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Chiara Villa
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Universitá degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Francesco Fortunato
- Neurology Unit, Neuroscience Section, Department of Pathophysiology and Transplantation, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Giacomo Pietro Comi
- Neurology Unit, Neuroscience Section, Department of Pathophysiology and Transplantation, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Giuseppina Milano
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy; Laboratory of Cardiovascular Research, Department of Surgery and Anesthesiology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy; Department of Cardiac Surgery, Centro Cardiologico Monzino-IRCCS, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Yvan Torrente
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Universitá degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy.
| |
Collapse
|
37
|
Bourke JP, Bueser T, Quinlivan R. Interventions for preventing and treating cardiac complications in Duchenne and Becker muscular dystrophy and X-linked dilated cardiomyopathy. Cochrane Database Syst Rev 2018; 10:CD009068. [PMID: 30326162 PMCID: PMC6517009 DOI: 10.1002/14651858.cd009068.pub3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The dystrophinopathies include Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD), and X-linked dilated cardiomyopathy (XLDCM). In recent years, co-ordinated multidisciplinary management for these diseases has improved the quality of care, with early corticosteroid use prolonging independent ambulation, and the routine use of non-invasive ventilation signficantly increasing survival. The next target to improve outcomes is optimising treatments to delay the onset or slow the progression of cardiac involvement and so prolong survival further. OBJECTIVES To assess the effects of interventions for preventing or treating cardiac involvement in DMD, BMD, and XLDCM, using measures of change in cardiac function over six months. SEARCH METHODS On 16 October 2017 we searched the Cochrane Neuromuscular Specialised Register, CENTRAL, MEDLINE and Embase, and on 12 December 2017, we searched two clinical trials registries. We also searched conference proceedings and bibliographies. SELECTION CRITERIA We considered only randomised controlled trials (RCTs), quasi-RCTs and randomised cross-over trials for inclusion. In the Discussion, we reviewed open studies, longitudinal observational studies and individual case reports but only discussed studies that adequately described the diagnosis, intervention, pretreatment, and post-treatment states and in which follow-up lasted for at least six months. DATA COLLECTION AND ANALYSIS Two authors independently reviewed the titles and abstracts identified from the search and performed data extraction. All three authors assessed risk of bias independently, compared results, and decided which trials met the inclusion criteria. They assessed the certainty of evidence using GRADE criteria. MAIN RESULTS We included five studies (N = 205) in the review; four studies included participants with DMD only, and one study included participants with DMD or BMD. All studied different interventions, and meta-analysis was not possible. We found no studies for XLDCM. None of the trials reported cardiac function as improved or stable cardiac versus deteriorated.The randomised first part of a two-part study of perindopril (N = 28) versus placebo (N = 27) in boys with DMD with normal heart function at baseline showed no difference in the number of participants with a left ventricular ejection fraction (LVEF%) of less than 45% after three years of therapy (n = 1 in each group; risk ratio (RR) 1.04, 95% confidence interval (CI) 0.07 to 15.77). This result is uncertain because of study limitations, indirectness and imprecision. In a non-randomised follow-up study, after 10 years, more participants who had received placebo from the beginning had reduced LVEF% (less than 45%). Adverse event rates were similar between the placebo and treatment groups (low-certainty evidence).A study comparing treatment with lisinopril versus losartan in 23 boys newly diagnosed with Duchenne cardiomyopathy showed that after 12 months, both were equally effective in preserving or improving LVEF% (lisinopril 54.6% (standard deviation (SD) 5.19), losartan 55.2% (SD 7.19); mean difference (MD) -0.60% CI -6.67 to 5.47: N = 16). The certainty of evidence was very low because of very serious imprecision and study limitations (risk of bias). Two participants in the losartan group were withdrawn due to adverse events: one participant developed an allergic reaction, and a second exceeded the safety standard with a fall in ejection fraction greater than 10%. Authors reported no other adverse events related to the medication (N = 22; very low-certainty evidence).A study comparing idebenone versus placebo in 21 boys with DMD showed little or no difference in mean change in cardiac function between the two groups from baseline to 12 months; for fractional shortening the mean change was 1.4% (SD 4.1) in the idebenone group and 1.6% (SD 2.6) in the placebo group (MD -0.20%, 95% CI -3.07 to 2.67, N = 21), and for ejection fraction the mean change was -1.9% (SD 9.8) in the idebenone group and 0.4% (SD 5.5) in the placebo group (MD -2.30%, 95% CI -9.18 to 4.58, N = 21). The certainty of evidence was very low because of study limitations and very serious imprecision. Reported adverse events were similar between the treatment and placebo groups (low-certainty evidence).A multicentre controlled study added eplerenone or placebo to 42 patients with DMD with early cardiomyopathy but preserved left ventricular function already established on ACEI or ARB therapy. Results showed that eplerenone slowed the rate of decline of magnetic resonance (MR)-assessed left ventricular circumferential strain at 12 months (eplerenone group median 1.0%, interquartile range (IQR) 0.3 to -2.2; placebo group median 2.2%, IQR 1.3 to -3.1%; P = 0.020). The median decline in LVEF over the same period was also less in the eplerenone group (-1.8%, IQR -2.9 to 6.0) than in the placebo group (-3.7%, IQR -10.8 to 1.0; P = 0.032). We downgraded the certainty of evidence to very low for study limitations and serious imprecision. Serious adverse events were reported in two patients given placebo but none in the treatment group (very low-certainty evidence).A randomised placebo-controlled study of subcutaneous growth hormone in 16 participants with DMD or BMD showed an increase in left ventricular mass after three months' treatment but no significant improvement in cardiac function. The evidence was of very low certainty due to imprecision, indirectness, and study limitations. There were no clinically significant adverse events (very low-certainty evidence).Some studies were at risk of bias, and all were small. Therefore, although there is some evidence from non-randomised data to support the prophylactic use of perindopril for cardioprotection ahead of detectable cardiomyopathy, and for lisinopril or losartan plus eplerenone once cardiomyopathy is detectable, this must be considered of very low certainty. Findings from non-randomised studies, some of which have been long term, have led to the use of these drugs in daily clinical practice. AUTHORS' CONCLUSIONS Based on the available evidence from RCTs, early treatment with ACE inhibitors or ARBs may be comparably beneficial for people with a dystrophinopathy; however, the certainty of evidence is very low. Very low-certainty evidence indicates that adding eplerenone might give additional benefit when early cardiomyopathy is detected. No clinically meaningful effect was seen for growth hormone or idebenone, although the certainty of the evidence is also very low.
Collapse
Affiliation(s)
- John P Bourke
- Freeman HospitalDepartment of CardiologyFreeman RoadNewcastle Upon TyneUKNE7 DN
| | - Teofila Bueser
- King's College LondonFlorence Nightingale Faculty of Nursing & MidwiferyLondonUKSE1 8WA
| | - Rosaline Quinlivan
- UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery and Great Ormond StreetMRC Centre for Neuromuscular Diseases and Dubowitz Neuromuscular CentrePO Box 114LondonUKWC1B 3BN
| | | |
Collapse
|
38
|
Peterson JM, Wang DJ, Shettigar V, Roof SR, Canan BD, Bakkar N, Shintaku J, Gu JM, Little SC, Ratnam NM, Londhe P, Lu L, Gaw CE, Petrosino JM, Liyanarachchi S, Wang H, Janssen PML, Davis JP, Ziolo MT, Sharma SM, Guttridge DC. NF-κB inhibition rescues cardiac function by remodeling calcium genes in a Duchenne muscular dystrophy model. Nat Commun 2018; 9:3431. [PMID: 30143619 PMCID: PMC6109146 DOI: 10.1038/s41467-018-05910-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 07/25/2018] [Indexed: 12/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a neuromuscular disorder causing progressive muscle degeneration. Although cardiomyopathy is a leading mortality cause in DMD patients, the mechanisms underlying heart failure are not well understood. Previously, we showed that NF-κB exacerbates DMD skeletal muscle pathology by promoting inflammation and impairing new muscle growth. Here, we show that NF-κB is activated in murine dystrophic (mdx) hearts, and that cardiomyocyte ablation of NF-κB rescues cardiac function. This physiological improvement is associated with a signature of upregulated calcium genes, coinciding with global enrichment of permissive H3K27 acetylation chromatin marks and depletion of the transcriptional repressors CCCTC-binding factor, SIN3 transcription regulator family member A, and histone deacetylase 1. In this respect, in DMD hearts, NF-κB acts differently from its established role as a transcriptional activator, instead promoting global changes in the chromatin landscape to regulate calcium genes and cardiac function.
Collapse
Affiliation(s)
- Jennifer M Peterson
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Pharmacy and Pharmaceutical Sciences, SUNY Binghamton University, Binghamton, NY, 13902, USA
| | - David J Wang
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Vikram Shettigar
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA
| | - Steve R Roof
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA.,Q Test Labs, Columbus, OH, 43235, USA
| | - Benjamin D Canan
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA
| | - Nadine Bakkar
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Neurobiology, St Joseph's Hospital and Medical Center-Barrow Neurological Institute, Phoenix, AZ, 85013, USA
| | - Jonathan Shintaku
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Jin-Mo Gu
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Biomedical Engineering and Pediatrics, Emory University, Decatur, GA, 30322, USA
| | - Sean C Little
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA.,Bristol-Myers Squibb, Wallingford, CT, 06492, USA
| | - Nivedita M Ratnam
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Priya Londhe
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Leina Lu
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Christopher E Gaw
- The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Jennifer M Petrosino
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Sandya Liyanarachchi
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Huating Wang
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Paul M L Janssen
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA
| | - Jonathan P Davis
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA
| | - Mark T Ziolo
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA
| | - Sudarshana M Sharma
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Denis C Guttridge
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA. .,Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA. .,The Ohio State University Medical Center, Columbus, OH, 43210, USA. .,Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, 29425, USA.
| |
Collapse
|
39
|
Milad N, White Z, Tehrani AY, Sellers S, Rossi FMV, Bernatchez P. Increased plasma lipid levels exacerbate muscle pathology in the mdx mouse model of Duchenne muscular dystrophy. Skelet Muscle 2017; 7:19. [PMID: 28899419 PMCID: PMC5596936 DOI: 10.1186/s13395-017-0135-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/28/2017] [Indexed: 01/11/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is caused by loss of dystrophin expression and leads to severe ambulatory and cardiac function decline. However, the dystrophin-deficient mdx murine model of DMD only develops a very mild form of the disease. Our group and others have shown vascular abnormalities in animal models of MD, a likely consequence of the fact that blood vessels express the same dystrophin-associated glycoprotein complex (DGC) proteins as skeletal muscles. Methods To test the blood vessel contribution to muscle damage in DMD, mdx4cv mice were given elevated lipid levels via apolipoprotein E (ApoE) gene knockout combined with normal chow or lipid-rich Western diets. Ambulatory function and heart function (via echocardiogram) were assessed at 4 and 7 months of age. After sacrifice, muscle histology and aortic staining were used to assess muscle pathology and atherosclerosis development, respectively. Plasma levels of total cholesterol, high-density lipoprotein (HDL), triglycerides, and creatine kinase (CK) were also measured. Results Although there was an increase in left ventricular heart volume in mdx-ApoE mice compared to that in mdx mice, parameters of heart function were not affected. Compared with wild-type and ApoE-null, only mdx-ApoE KO mice showed significant ambulatory dysfunction. Despite no significant difference in plasma CK, histological analyses revealed that elevated plasma lipids in chow- and Western diet-fed mdx-ApoE mice was associated with severe exacerbation of muscle pathology compared to mdx mice: significant increase in myofiber damage and fibrofatty replacement in the gastrocnemius and triceps brachii muscles, more reminiscent of human DMD pathology. Finally, although both ApoE and mdx-ApoE groups displayed increased plasma lipids, mdx-ApoE exhibited atherosclerotic plaque deposition equal to or less than that of ApoE mice. Conclusions Since others have shown that lipid abnormalities correlate with DMD severity, our data suggest that plasma lipids could be primary contributors to human DMD severity and that the notoriously mild phenotype of mdx mice might be attributable in part to their endogenously low plasma lipid profiles. Hence, DMD patients may benefit from lipid-lowering and vascular-targeted therapies. Electronic supplementary material The online version of this article (10.1186/s13395-017-0135-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nadia Milad
- Department of Anaesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, British Columbia, V6T 1Z3, Canada.,Centre for Heart and Lung Innovation, St. Paul's Hospital, 1081 Burrard Street, Rm 166, Vancouver, British Columbia, Canada
| | - Zoe White
- Department of Anaesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, British Columbia, V6T 1Z3, Canada.,Centre for Heart and Lung Innovation, St. Paul's Hospital, 1081 Burrard Street, Rm 166, Vancouver, British Columbia, Canada
| | - Arash Y Tehrani
- Department of Anaesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, British Columbia, V6T 1Z3, Canada.,Centre for Heart and Lung Innovation, St. Paul's Hospital, 1081 Burrard Street, Rm 166, Vancouver, British Columbia, Canada
| | - Stephanie Sellers
- Department of Anaesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, British Columbia, V6T 1Z3, Canada.,Centre for Heart and Lung Innovation, St. Paul's Hospital, 1081 Burrard Street, Rm 166, Vancouver, British Columbia, Canada
| | - Fabio M V Rossi
- Department of Medical Genetics, Centre for Biomedical Research, University of British Columbia, 2222 Health Sciences Mall, Vancouver, British Columbia, Canada
| | - Pascal Bernatchez
- Department of Anaesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, British Columbia, V6T 1Z3, Canada. .,Centre for Heart and Lung Innovation, St. Paul's Hospital, 1081 Burrard Street, Rm 166, Vancouver, British Columbia, Canada.
| |
Collapse
|
40
|
Tsuda T, Fitzgerald KK. Dystrophic Cardiomyopathy: Complex Pathobiological Processes to Generate Clinical Phenotype. J Cardiovasc Dev Dis 2017; 4:jcdd4030014. [PMID: 29367543 PMCID: PMC5715712 DOI: 10.3390/jcdd4030014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/27/2017] [Accepted: 08/30/2017] [Indexed: 02/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD), and X-linked dilated cardiomyopathy (XL-DCM) consist of a unique clinical entity, the dystrophinopathies, which are due to variable mutations in the dystrophin gene. Dilated cardiomyopathy (DCM) is a common complication of dystrophinopathies, but the onset, progression, and severity of heart disease differ among these subgroups. Extensive molecular genetic studies have been conducted to assess genotype-phenotype correlation in DMD, BMD, and XL-DCM to understand the underlying mechanisms of these diseases, but the results are not always conclusive, suggesting the involvement of complex multi-layers of pathological processes that generate the final clinical phenotype. Dystrophin protein is a part of dystrophin-glycoprotein complex (DGC) that is localized in skeletal muscles, myocardium, smooth muscles, and neuronal tissues. Diversity of cardiac phenotype in dystrophinopathies suggests multiple layers of pathogenetic mechanisms in forming dystrophic cardiomyopathy. In this review article, we review the complex molecular interactions involving the pathogenesis of dystrophic cardiomyopathy, including primary gene mutations and loss of structural integrity, secondary cellular responses, and certain epigenetic and other factors that modulate gene expressions. Involvement of epigenetic gene regulation appears to lead to specific cardiac phenotypes in dystrophic hearts.
Collapse
Affiliation(s)
- Takeshi Tsuda
- Nemours Cardiac Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, 1600 Rockland Rd, DE 19803, USA.
| | - Kristi K Fitzgerald
- Nemours Cardiac Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, 1600 Rockland Rd, DE 19803, USA.
| |
Collapse
|
41
|
Sharifi-Sanjani M, Oyster NM, Tichy ED, Bedi KC, Harel O, Margulies KB, Mourkioti F. Cardiomyocyte-Specific Telomere Shortening is a Distinct Signature of Heart Failure in Humans. J Am Heart Assoc 2017; 6:JAHA.116.005086. [PMID: 28882819 PMCID: PMC5634248 DOI: 10.1161/jaha.116.005086] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Telomere defects are thought to play a role in cardiomyopathies, but the specific cell type affected by the disease in human hearts is not yet identified. The aim of this study was to systematically evaluate the cell type specificity of telomere shortening in patients with heart failure in relation to their cardiac disease, age, and sex. Methods and Results We studied cardiac tissues from patients with heart failure by utilizing telomere quantitative fluorescence in situ hybridization, a highly sensitive method with single‐cell resolution. In this study, total of 63 human left ventricular samples, including 37 diseased and 26 nonfailing donor hearts, were stained for telomeres in combination with cardiomyocyte‐ or α‐smooth muscle cell‐specific markers, cardiac troponin T, and smooth muscle actin, respectively, and assessed for telomere length. Patients with heart failure demonstrate shorter cardiomyocyte telomeres compared with nonfailing donors, which is specific only to cardiomyocytes within diseased human hearts and is associated with cardiomyocyte DNA damage. Our data further reveal that hypertrophic hearts with reduced ejection fraction exhibit the shortest telomeres. In contrast to other reported cell types, no difference in cardiomyocyte telomere length is evident with age. However, under the disease state, telomere attrition manifests in both young and older patients with cardiac hypertrophy. Finally, we demonstrate that cardiomyocyte‐telomere length is better sustained in women than men under diseased conditions. Conclusions This study provides the first evidence of cardiomyocyte‐specific telomere shortening in heart failure.
Collapse
Affiliation(s)
| | - Nicholas M Oyster
- Department of Orthopaedic Surgery, University of Connecticut, Storrs, CT
| | - Elisia D Tichy
- Department of Orthopaedic Surgery, University of Connecticut, Storrs, CT
| | - Kenneth C Bedi
- Cardiovascular Institute, University of Connecticut, Storrs, CT
| | - Ofer Harel
- College of Liberal Arts and Sciences, Department of Statistics, University of Connecticut, Storrs, CT
| | | | - Foteini Mourkioti
- Department of Orthopaedic Surgery, University of Connecticut, Storrs, CT .,Cardiovascular Institute, University of Connecticut, Storrs, CT.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
42
|
Nance ME, Hakim CH, Yang NN, Duan D. Nanotherapy for Duchenne muscular dystrophy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 10. [PMID: 28398005 DOI: 10.1002/wnan.1472] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 02/09/2017] [Accepted: 03/11/2017] [Indexed: 12/14/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a lethal X-linked childhood muscle wasting disease caused by mutations in the dystrophin gene. Nanobiotechnology-based therapies (such as synthetic nanoparticles and naturally existing viral and nonviral nanoparticles) hold great promise to replace and repair the mutated dystrophin gene and significantly change the disease course. While a majority of DMD nanotherapies are still in early preclinical development, several [such as adeno-associated virus (AAV)-mediated systemic micro-dystrophin gene therapy] are advancing for phase I clinical trials. Recent regulatory approval of Ataluren (a nonsense mutation read-through chemical) in Europe and Exondys51 (an exon-skipping antisense oligonucleotide drug) in the United States shall offer critical insight in how to move DMD nanotherapy to human patients. Progress in novel, optimized nano-delivery systems may further improve emerging molecular therapeutic modalities for DMD. Despite these progresses, DMD nanotherapy faces a number of unique challenges. Specifically, the dystrophin gene is one of the largest genes in the genome while nanoparticles have an inherent size limitation per definition. Furthermore, muscle is the largest tissue in the body and accounts for 40% of the body mass. How to achieve efficient bodywide muscle targeting in human patients with nanomedication remains a significant translational hurdle. New creative approaches in the design of the miniature micro-dystrophin gene, engineering of muscle-specific synthetic AAV capsids, and novel nanoparticle-mediated exon-skipping are likely to result in major breakthroughs in DMD therapy. WIREs Nanomed Nanobiotechnol 2018, 10:e1472. doi: 10.1002/wnan.1472 This article is categorized under: Biology-Inspired Nanomaterials > Protein and Virus-Based Structures Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Michael E Nance
- Department of Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, USA
| | - Chady H Hakim
- Department of Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, USA.,National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - N Nora Yang
- National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Dongsheng Duan
- Department of Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, USA.,Department of Neurology, University of Missouri, Columbia, MO, USA.,Department of Bioengineering, University of Missouri, Columbia, MO, USA.,Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
| |
Collapse
|
43
|
Yang L, Gregorich ZR, Cai W, Zhang P, Young B, Gu Y, Zhang J, Ge Y. Quantitative Proteomics and Immunohistochemistry Reveal Insights into Cellular and Molecular Processes in the Infarct Border Zone One Month after Myocardial Infarction. J Proteome Res 2017; 16:2101-2112. [PMID: 28347137 DOI: 10.1021/acs.jproteome.7b00107] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Postinfarction remodeling and expansion of the peri-infarct border zone (BZ) directly correlate with mortality following myocardial infarction (MI); however, the cellular and molecular mechanisms underlying remodeling processes in the BZ remain unclear. Herein, we utilized a label-free quantitative proteomics approach in combination with immunohistochemical analyses to gain a better understanding of processes contributing to postinfarction remodeling of the peri-infarct BZ in a swine model of MI with reperfusion. Our analysis uncovered a significant down-regulation of proteins involved in energy metabolism, indicating impaired myocardial energetics and possibly mitochondrial dysfunction, in the peri-scar BZ. An increase in endothelial and vascular smooth muscles cells, as well as up-regulation of proteins implicated in vascular endothelial growth factor (VEGF) signaling and marked changes in the expression of extracellular matrix and subendothelial basement membrane proteins, is indicative of active angiogenesis in the infarct BZ. A pronounced increase in macrophages in the peri-infarct BZ was also observed, and proteomic analysis uncovered evidence of persistent inflammation in this tissue. Additional evidence suggested an increase in cellular proliferation that, concomitant with increased nestin expression, indicates potential turnover of endogenous stem cells in the BZ. A marked up-regulation of pro-apoptotic proteins, as well as the down-regulation of proteins important for adaptation to mechanical, metabolic, and oxidative stress, likely contributes to increased apoptosis in the peri-infarct BZ. The cellular processes and molecular pathways identified herein may have clinical utility for therapeutic intervention aimed at limiting remodeling and expansion of the BZ myocardium and preventing the development of heart failure post-MI.
Collapse
Affiliation(s)
- Libang Yang
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School , Minneapolis, Minnesota 55455, United States
| | | | | | - Patrick Zhang
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School , Minneapolis, Minnesota 55455, United States
| | - Bernice Young
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School , Minneapolis, Minnesota 55455, United States
| | | | - Jianyi Zhang
- Department of Biomedical Engineering, School of Engineering, School of Medicine, University of Alabama at Birmingham , Birmingham, Alabama 35294, United States
| | | |
Collapse
|
44
|
Ballmann C, Denney T, Beyers RJ, Quindry T, Romero M, Selsby JT, Quindry JC. Long-term dietary quercetin enrichment as a cardioprotective countermeasure in mdx mice. Exp Physiol 2017; 102:635-649. [PMID: 28192862 DOI: 10.1113/ep086091] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/02/2017] [Indexed: 12/19/2022]
Abstract
NEW FINDINGS What is the central question of this study? The central question of this study is to understand whether dietary quercetin enrichment attenuates physiologic, histological, and biochemical indices of cardiac pathology. What is the main finding and its importance? Novel findings from this investigation, in comparison to prior published studies, suggest that mouse strain-dependent cardiac outcomes in performance and remodelling exist. Unlike Mdx/Utrn-/+ mice, mdx mice receiving lifelong quercetin treatment did not exhibit improvements cardiac function. Similar to prior work in Mdx/Utrn-/+ mice, histological evidence of remodelling suggests that quercetin consumption may have benefited hearts of mdx mice. Positive outcomes may be related to indirect markers that suggest improved mitochondrial wellbeing and to selected indices of inflammation that were lower in hearts from quercetin-fed mice. Duchenne muscular dystrophy causes a decline in cardiac health, resulting in premature mortality. As a potential countermeasure, quercetin is a polyphenol possessing inherent anti-inflammatory and antioxidant effects that activate proliferator-activated γ coactivator 1α (PGC-1α), increasing the abundance of mitochondrial biogenesis proteins. We investigated the extent to which lifelong 0.2% dietary quercetin enrichment attenuates dystrophic cardiopathology in mdx mice. Dystrophic animals were fed a quercetin-enriched or control diet for 12 months, while control C57 mice were fed a control diet. Cardiac function was assessed via 7 T magnetic resonance imaging at 2, 10 and 14 months. At 14 months, hearts were harvested for histology and Western blotting. The results indicated an mdx strain-dependent decline in cardiac performance at 14 months and that dietary quercetin enrichment did not attenuate functional losses. In contrast, histological analyses provided evidence that quercetin feeding was associated with decreased fibronectin and indirect damage indices (Haematoxylin and Eosin) compared with untreated mdx mice. Dietary quercetin enrichment increased cardiac protein abundance of PGC-1α, cytochrome c, electron transport chain complexes I-V, citrate synthase, superoxide dismutase 2 and glutathione peroxidase (GPX) versus untreated mdx mice. The protein abundance of the inflammatory markers nuclear factor-κB, phosphorylated nuclear factor kappa beta (P-NFκB) and phosphorylated nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha (P-IKBα) was decreased by quercetin compared with untreated mdx mice, while preserving nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha( IKBα) compared with mdx mice. Furthermore, quercetin decreased transforming growth factor-β1, cyclooxygenase-2 (COX2) and macrophage-restricted F4/80 protein (F4/80) versus untreated mdx mice. The data suggest that long-term quercetin enrichment does not impact physiological parameters of cardiac function but improves indices of mitochondrial biogenesis and antioxidant enzymes, facilitates dystrophin-associated glycoprotein complex (DGC) assembly and decreases inflammation in dystrophic hearts.
Collapse
Affiliation(s)
| | - Thomas Denney
- MRI Research Center, Auburn University, Auburn, AL, USA
| | | | | | - Matthew Romero
- School of Kinesiology, Auburn University, Auburn, AL, USA
| | - Joshua T Selsby
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | - John C Quindry
- School of Kinesiology, Auburn University, Auburn, AL, USA
| |
Collapse
|
45
|
Short-term and long-term models of doxorubicin-induced cardiomyopathy in rats: A comparison of functional and histopathological changes. ACTA ACUST UNITED AC 2017; 69:213-219. [PMID: 28153388 DOI: 10.1016/j.etp.2017.01.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 12/28/2016] [Accepted: 01/16/2017] [Indexed: 11/23/2022]
Abstract
OBJECTIVES Doxorubicin (DXR), an anthracyclic antineoplastic agent, is one of the most commonly drug utilized to induce dilated cardiomyopathy (DCM) and heart failure (HF), but the well optimized protocol for cardiomyopathy induction leading to development of cardiac systolic dysfunction is unclear. This study aims to critically compare short-term and long-term DXR injection protocols for the induction of DCM in rats. METHODS Animals were allocated into 3 experimental groups: a ST (short-term DXR injection) group, in which animals received 6 intraperitoneal (i.p.) injections of DXR (2.5mg/kg per dose) over a period of 2 weeks (cumulative dose of 15mg/kg); a LT (long-term DXR injection) group in which animals received weekly i.p. injections of DXR (2mg/kg per dose) over a period of 9 weeks (cumulative dose of 18mg/kg); and a control group in which animals received an appropriate volume of 0.9% saline i.p. All animals were submitted to echocardiography analysis at baseline and after completion treatment. Afterwards, the hearts were collected for conventional light microscopy and collagen quantification. RESULTS Morphological myocardial analysis of both DXR-treated groups showed an identical pattern of swollen and vacuolated cardiomyocytes and disorganization of myofibrils. There was pronounced interstitial fibrosis in both groups of DXR-treated hearts as compared to controls, as assessed by the interstitial collagen volume fraction. There was no difference in interstitial fibrosis between the ST and LT groups. The echocardiography analysis of the LT group showed structural and functional findings compatible with DCM, including increased left ventricular systolic (5.02±0.96mm) and diastolic (7.68±0.96mm) dimensions and reduction of ejection fraction (69.40±8.51%) as compared to the ST group (4.10±0.89mm, 7.32±0.84, and 79.68±7.23%, respectively) and control group (4.07±0.72mm, 7.17±0.68mm and 80.08±4.71%, respectively), ANOVA p<0.01. CONCLUSIONS These results indicate that LT injection of DXR is more effective than ST injection in inducing left ventricular dysfunction and structural cardiac changes resembling those found in dilated cardiomyopathy.
Collapse
|
46
|
Ballmann C, Denney TS, Beyers RJ, Quindry T, Romero M, Amin R, Selsby JT, Quindry JC. Lifelong quercetin enrichment and cardioprotection in Mdx/Utrn+/− mice. Am J Physiol Heart Circ Physiol 2017; 312:H128-H140. [DOI: 10.1152/ajpheart.00552.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/03/2016] [Accepted: 11/03/2016] [Indexed: 11/22/2022]
Abstract
Duchenne Muscular Dystrophy (DMD) is associated with progressive cardiac pathology; however, the SIRT1/PGC1-α activator quercetin may cardioprotect dystrophic hearts. We tested the extent to which long-term 0.2% dietary quercetin enrichment attenuates dystrophic cardiopathology in Mdx/Utrn+/− mice. At 2 mo, Mdx/Utrn+/− mice were fed quercetin-enriched (Mdx/Utrn+/−-Q) or control diet (Mdx/Utrn+/−) for 8 mo. Control C57BL/10 (C57) animals were fed a control diet for 10 mo. Cardiac function was quantified by MRI at 2 and 10 mo. Spontaneous physical activity was quantified during the last week of treatment. At 10 mo hearts were excised for histological and biochemical analysis. Quercetin feeding improved various physiological indexes of cardiac function in diseased animals. Mdx/Utrn+/−-Q also engaged in more high-intensity physical activity than controls. Histological analyses of heart tissues revealed higher expression and colocalization of utrophin and α-sarcoglycan. Lower abundance of fibronectin, cardiac damage (Hematoxylin Eosin-Y), and MMP9 were observed in quercetin-fed vs. control Mdx/Utrn+/− mice. Quercetin evoked higher protein abundance of PGC-1α, cytochrome c, ETC complexes I–V, citrate synthase, SOD2, and GPX compared with control-fed Mdx/Utrn+/−. Quercetin decreased abundance of inflammatory markers including NFκB, TGF-β1, and F4/80 compared with Mdx/Utrn+/−; however, P-NFκB, P-IKBα, IKBα, CD64, and COX2 were similar between groups. Dietary quercetin enrichment improves cardiac function in aged Mdx/Utrn+/− mice and increases mitochondrial protein content and dystrophin glycoprotein complex formation. Histological analyses indicate a marked attenuation in pathological cardiac remodeling and indicate that long-term quercetin consumption benefits the dystrophic heart. NEW & NOTEWORTHY The current investigation provides first-time evidence that quercetin provides physiological cardioprotection against dystrophic pathology and is associated with improved spontaneous physical activity. Secondary findings suggest that quercetin-dependent outcomes are in part due to PGC-1α pathway activation.
Collapse
Affiliation(s)
| | | | | | | | - Matthew Romero
- School of Kinesiology, Auburn University, Auburn, Alabama
| | - Rajesh Amin
- Harrison School of Pharmacy, Auburn University, Auburn, Alabama; and
| | | | | |
Collapse
|
47
|
Taqatqa A, Bokowski J, Al-Kubaisi M, Khalil A, Miranda C, Alaksham H, Fughhi I, Kenny D, Diab KA. The Use of Speckle Tracking Echocardiography for Early Detection of Myocardial Dysfunction in Patients with Duchenne Muscular Dystrophy. Pediatr Cardiol 2016; 37:1422-1428. [PMID: 27452803 DOI: 10.1007/s00246-016-1451-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/09/2016] [Indexed: 11/27/2022]
Abstract
Cardiac complications are the leading cause of death in patients with Duchenne muscular dystrophy (DMD). Two-dimensional echocardiography is the current standard for monitoring of LV systolic function in these patients, but it might not detect early systolic dysfunction. The current study examined the use of speckle tracking echocardiography (STE) to detect early signs of cardiac dysfunction in DMD patients. A retrospective review of charts and offline strain analysis of transthoracic echocardiographic studies of DMD patients at our institution from April 2014 to January 2015 were performed and compared to age-matched healthy male subjects. Nineteen DMD patients (age range 12.6 ± 3.1 years) with normal ejection fraction and shortening fraction were compared with sixteen controls. The global circumferential strain was lower in DMD patients compared with controls (-14.7 ± 4.7 vs. -23.1 ± 2.9 %, respectively, p value: 0.001). Circumferential strain measured at basal, mid-ventricular and apical parasternal short-axis views was lower in DMD patients compared with controls. Segmental circumferential strain was lower in DMD patients in most segments compared with controls. The global longitudinal strain was lower in DMD patients compared with controls (-13.6 ± 5 vs. -18.8 ± 3 %, respectively, p value: 0.001). Segmental longitudinal strain measured in various segments was lower in DMD patients compared with controls. DMD patients can have occult cardiovascular dysfunction as shown by reduction in circumferential and longitudinal strain measurements with STE despite normal standard echocardiographic parameters. The clinical significance of early detection of cardiac dysfunction in these patients warrants further studies.
Collapse
Affiliation(s)
- Anas Taqatqa
- Rush Congenital Echocardiography Laboratory, Rush Center for Congenital Heart Disease, Rush University Medical Center, PAV 667 A 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - John Bokowski
- Rush Congenital Echocardiography Laboratory, Rush Center for Congenital Heart Disease, Rush University Medical Center, PAV 667 A 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - Maytham Al-Kubaisi
- Rush Congenital Echocardiography Laboratory, Rush Center for Congenital Heart Disease, Rush University Medical Center, PAV 667 A 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - Ahmad Khalil
- Rush Congenital Echocardiography Laboratory, Rush Center for Congenital Heart Disease, Rush University Medical Center, PAV 667 A 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - Carlos Miranda
- Rush Congenital Echocardiography Laboratory, Rush Center for Congenital Heart Disease, Rush University Medical Center, PAV 667 A 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - Hamad Alaksham
- Rush Congenital Echocardiography Laboratory, Rush Center for Congenital Heart Disease, Rush University Medical Center, PAV 667 A 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - Ibtihaj Fughhi
- Rush Congenital Echocardiography Laboratory, Rush Center for Congenital Heart Disease, Rush University Medical Center, PAV 667 A 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - Damien Kenny
- Rush Congenital Echocardiography Laboratory, Rush Center for Congenital Heart Disease, Rush University Medical Center, PAV 667 A 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - Karim A Diab
- Rush Congenital Echocardiography Laboratory, Rush Center for Congenital Heart Disease, Rush University Medical Center, PAV 667 A 1653 W. Congress Parkway, Chicago, IL, 60612, USA.
| |
Collapse
|
48
|
Freitas ACS, Figueiredo MJ, Campos EC, Soave DF, Ramos SG, Tanowitz HB, Celes MRN. Activation of Both the Calpain and Ubiquitin-Proteasome Systems Contributes to Septic Cardiomyopathy through Dystrophin Loss/Disruption and mTOR Inhibition. PLoS One 2016; 11:e0166839. [PMID: 27880847 PMCID: PMC5120800 DOI: 10.1371/journal.pone.0166839] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 11/05/2016] [Indexed: 11/18/2022] Open
Abstract
Cardiac dysfunction caused by the impairment of myocardial contractility has been recognized as an important factor contributing to the high mortality in sepsis. Calpain activation in the heart takes place in response to increased intracellular calcium influx resulting in proteolysis of structural and contractile proteins with subsequent myocardial dysfunction. The purpose of the present study was to test the hypothesis that increased levels of calpain in the septic heart leads to disruption of structural and contractile proteins and that administration of calpain inhibitor-1 (N-acetyl-leucinyl-leucinyl-norleucinal (ALLN)) after sepsis induced by cecal ligation and puncture prevents cardiac protein degradation. We also tested the hypothesis that calpain plays a role in the modulation of protein synthesis/degradation through the activation of proteasome-dependent proteolysis and inhibition of the mTOR pathway. Severe sepsis significantly increased heart calpain-1 levels and promoted ubiquitin and Pa28β over-expression with a reduction in the mTOR levels. In addition, sepsis reduced the expression of structural proteins dystrophin and β-dystroglycan as well as the contractile proteins actin and myosin. ALLN administration prevented sepsis-induced increases in calpain and ubiquitin levels in the heart, which resulted in decreased of structural and contractile proteins degradation and basal mTOR expression levels were re-established. Our results support the concept that increased calpain concentrations may be part of an important mechanism of sepsis-induced cardiac muscle proteolysis.
Collapse
Affiliation(s)
- Ana Caroline Silva Freitas
- Department of Pathology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paulo, Brazil
| | - Maria Jose Figueiredo
- Department of Pathology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paulo, Brazil
| | - Erica Carolina Campos
- Department of Physiotherapy, Faculty of Physical Education, Federal University of Uberlandia, Minas Gerais, Brazil
| | - Danilo Figueiredo Soave
- Department of Histology, Embryology and Cellular Biology, Federal University of Goias, Goias, Brazil
| | - Simone Gusmao Ramos
- Department of Pathology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paulo, Brazil
| | - Herbert B. Tanowitz
- Departments of Pathology and medicine, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York, United States of America
| | - Mara Rúbia N. Celes
- Department of Pathology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paulo, Brazil
- Institute of Tropical Pathology and Public Health, Federal University of Goias, Goias, Brazil
- * E-mail: ,
| |
Collapse
|
49
|
Microtubule-Mediated Misregulation of Junctophilin-2 Underlies T-Tubule Disruptions and Calcium Mishandling in mdx Mice. JACC Basic Transl Sci 2016; 1:122-130. [PMID: 27482548 PMCID: PMC4965806 DOI: 10.1016/j.jacbts.2016.02.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cardiac myocytes from the mdx mouse, the mouse model of Duchenne muscular dystrophy, exhibit t-tubule disarray and increased calcium sparks, but a unifying molecular mechanism has not been elucidated. Recently, improper trafficking of junctophilin (JPH)-2 on an altered microtubule network caused t-tubule derangements and calcium mishandling in a pressure-overload heart failure model. Mdx cardiac myocytes have microtubule abnormalities, but how this may affect JPH-2, t-tubules, and calcium handling has not been established. Here, we investigated the hypothesis that an inverse relationship between microtubules and JPH-2 underlies t-tubule disruptions and calcium mishandling in mdx cardiac myocytes. Confocal microscopy revealed t-tubule disorganization in mdx cardiac myocytes. Quantitative Western blot analysis demonstrated JPH-2 was decreased by 75% and showed an inverse hyperbolic relationship with α- and β-tubulin, the individual components of microtubules, in mdx hearts. Colchicine-induced microtubule depolymerization normalized JPH-2 protein levels and localization, corrected t-tubule architecture, and reduced calcium sparks. In summary, these results suggest microtubule-mediated misregulation of JPH-2 causes t-tubule derangements and altered calcium handling in mdx cardiac myocytes. Decreased junctophilin-2 levels are associated with cardiac t-tubule derangements in mdx mice, the mouse model of Duchenne muscular dystrophy (DMD). Reduced junctophilin-2 protein levels correlate with increases in total microtubule content in mdx hearts. Colchicine-mediated microtubule depolymerization increases junctophilin-2 protein levels and improves localization patterns which, in turn, are associated with t-tubule reorganization and reduced calcium sparks. This study identifies microtubule-mediated misregulation of junctophilin-2 as a novel molecular mechanism in Duchenne cardiomyopathy.
Collapse
|
50
|
Abstract
The dystrophin complex stabilizes the plasma membrane of striated muscle cells. Loss of function mutations in the genes encoding dystrophin, or the associated proteins, trigger instability of the plasma membrane, and myofiber loss. Mutations in dystrophin have been extensively cataloged, providing remarkable structure-function correlation between predicted protein structure and clinical outcomes. These data have highlighted dystrophin regions necessary for in vivo function and fueled the design of viral vectors and now, exon skipping approaches for use in dystrophin restoration therapies. However, dystrophin restoration is likely more complex, owing to the role of the dystrophin complex as a broad cytoskeletal integrator. This review will focus on dystrophin restoration, with emphasis on the regions of dystrophin essential for interacting with its associated proteins and discuss the structural implications of these approaches.
Collapse
Affiliation(s)
- Quan Q Gao
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, Illinois, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University, Chicago, Chicago, Illinois, USA
| |
Collapse
|