1
|
Earl CC, Javier AJ, Richards AM, Markham LW, Goergen CJ, Welc SS. Functional cardiac consequences of β-adrenergic stress-induced injury in a model of Duchenne muscular dystrophy. Dis Model Mech 2024; 17:dmm050852. [PMID: 39268580 PMCID: PMC11488649 DOI: 10.1242/dmm.050852] [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: 04/16/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024] Open
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 stimulate injury and enhance cardiac pathology in the mdx model, many methods lead to high mortality with 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. mdx and wild-type mice received daily low-dose (2 mg/kg/day) isoproterenol injections for 10 days. Histopathological 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 compared to wild-type mice. Our findings highlight the utility of challenging mdx mice with low-dose isoproterenol as a valuable model for exploring therapies targeting DMD-associated cardiac pathologies.
Collapse
MESH Headings
- Animals
- Muscular Dystrophy, Duchenne/complications
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/physiopathology
- Mice, Inbred mdx
- Isoproterenol/pharmacology
- Disease Models, Animal
- Fibrosis
- Stress, Physiological/drug effects
- Receptors, Adrenergic, beta/metabolism
- Myocardium/pathology
- Myocardium/metabolism
- Heart/drug effects
- Heart/physiopathology
- Mice
- Male
- Mice, Inbred C57BL
- Troponin I/metabolism
- Troponin I/blood
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/metabolism
- Adrenergic beta-Agonists/pharmacology
Collapse
Affiliation(s)
- Conner C. Earl
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
- Department of Medicine, Indiana University School of Medicine, IN 46202, USA
| | - Areli J. Javier
- Musculoskeletal Health Sciences Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alyssa M. Richards
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Larry W. Markham
- Division of Pediatric Cardiology, Riley Children's Hospital at Indiana University Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Craig J. Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
- Department of Medicine, Indiana University School of Medicine, IN 46202, USA
| | - Steven S. Welc
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, USA
| |
Collapse
|
2
|
Hasegawa J, Nagata T, Ihara K, Tanihata J, Ebihara S, Yoshida-Tanaka K, Yanagidaira M, Ohara M, Sasaki A, Nakayama M, Yamamoto S, Ishii T, Iwata-Hara R, Naito M, Miyata K, Sakaue F, Yokota T. Heteroduplex oligonucleotide technology boosts oligonucleotide splice switching activity of morpholino oligomers in a Duchenne muscular dystrophy mouse model. Nat Commun 2024; 15:7530. [PMID: 39327422 PMCID: PMC11427662 DOI: 10.1038/s41467-024-48204-5] [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: 03/02/2023] [Accepted: 04/23/2024] [Indexed: 09/28/2024] Open
Abstract
The approval of splice-switching oligonucleotides with phosphorodiamidate morpholino oligomers (PMOs) for treating Duchenne muscular dystrophy (DMD) has advanced the field of oligonucleotide therapy. Despite this progress, PMOs encounter challenges such as poor tissue uptake, particularly in the heart, diaphragm, and central nervous system (CNS), thereby affecting patient's prognosis and quality of life. To address these limitations, we have developed a PMOs-based heteroduplex oligonucleotide (HDO) technology. This innovation involves a lipid-ligand-conjugated complementary strand hybridized with PMOs, significantly enhancing delivery to key tissues in mdx mice, normalizing motor functions, muscle pathology, and serum creatine kinase by restoring internal deleted dystrophin expression. Additionally, PMOs-based HDOs normalized cardiac and CNS abnormalities without adverse effects. Our technology increases serum albumin binding to PMOs and improves blood retention and cellular uptake. Here we show that PMOs-based HDOs address the limitations in oligonucleotide therapy for DMD and offer a promising approach for diseases amenable to exon-skipping therapy.
Collapse
Affiliation(s)
- Juri Hasegawa
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
| | - Tetsuya Nagata
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan.
- Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan.
- NucleoTIDE and PepTIDE Drug Discovery Center, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan.
| | - Kensuke Ihara
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
| | - Jun Tanihata
- Department of Cell Physiology, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, 105-8461, Minato-ku, Tokyo, Japan
| | - Satoe Ebihara
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
| | - Kie Yoshida-Tanaka
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
| | - Mitsugu Yanagidaira
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
| | - Masahiro Ohara
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
| | - Asuka Sasaki
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
| | - Miyu Nakayama
- COE for Drug Metabolism, Pharmacokinetics and Modeling, Preclinical and Translational Sciences, Research, Takeda Pharmaceutical Company Limited, 2-26-1, Fujisawa, Kanagawa, 251-8555, Japan
| | - Syunsuke Yamamoto
- COE for Drug Metabolism, Pharmacokinetics and Modeling, Preclinical and Translational Sciences, Research, Takeda Pharmaceutical Company Limited, 2-26-1, Fujisawa, Kanagawa, 251-8555, Japan
| | - Takashi Ishii
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
| | - Rintaro Iwata-Hara
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
| | - Mitsuru Naito
- Department of Materials Engineering, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, 113-8656, Tokyo, Japan
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, 113-8656, Tokyo, Japan
| | - Fumika Sakaue
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan.
- Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan.
- NucleoTIDE and PepTIDE Drug Discovery Center, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Tokyo, Japan.
| |
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
|
Connolly AM, Zaidman CM, Brandsema JF, Phan HC, Tian C, Zhang X, Li J, Eisner MD, Carrier E. Pamrevlumab, a Fully Human Monoclonal Antibody Targeting Connective Tissue Growth Factor, for Non-Ambulatory Patients with Duchenne Muscular Dystrophy. J Neuromuscul Dis 2023:JND230019. [PMID: 37248912 DOI: 10.3233/jnd-230019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a neuromuscular disease stemming from dystrophin gene mutations. Lack of dystrophin leads to progressive muscle damage and replacement of muscle with fibrotic and adipose tissue. Pamrevlumab (FG-3019), a fully human monoclonal antibody that binds to connective tissue growth factor (CTGF), is in Phase III development for treatment of DMD and other diseases. METHODS MISSION (Study 079; NCT02606136) was an open-label, Phase II, single-arm trial of pamrevlumab in 21 non-ambulatory patients with DMD (aged≥12 years, receiving corticosteroids) who received 35-mg/kg intravenous infusions every 2 weeks for 2 years. The primary endpoint was change from baseline in percent predicted forced vital capacity (ppFVC). Secondary endpoints included other pulmonary function tests, upper limb function and strength assessments, and changes in upper arm fat and fibrosis scores on magnetic resonance imaging. RESULTS Fifteen patients completed the trial. Annual change from baseline (SE) in ppFVC was -4.2 (0.7) (95% CI -5.5, -2.8). Rate of decline in ppFVC in pamrevlumab-treated patients was slower than observed in historical published trials of non-ambulatory patients. MISSION participants experienced slower-than-anticipated muscle function declines compared with natural history and historical published trials of non-ambulatory patients with DMD. Pamrevlumab was well-tolerated. Treatment-emergent adverse events were mild to moderate, and none led to study discontinuation. CONCLUSIONS nti-CTGF therapy with pamrevlumab represents a potential treatment for DMD. The lack of internal control group limits the results.
Collapse
Affiliation(s)
- Anne M Connolly
- Nationwide Children's Hospital, Ohio State University College of Medicine, Columbus, OH, USA
| | - Craig M Zaidman
- Department of Neurology, Washington University at St. Louis, St. Louis, MO, USA
| | - John F Brandsema
- Division of Neurology, The Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Han C Phan
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Cuixia Tian
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Jack Li
- FibroGen, Inc., San Francisco, CA, USA
| | | | | |
Collapse
|
5
|
Onódi Z, Szabó PL, Kucsera D, Pokreisz P, Dostal C, Hilber K, Oudit GY, Podesser BK, Ferdinandy P, Varga ZV, Kiss A. Inflammasome Activity in the Skeletal Muscle and Heart of Rodent Models for Duchenne Muscular Dystrophy. Int J Mol Sci 2023; 24:8497. [PMID: 37239853 PMCID: PMC10218525 DOI: 10.3390/ijms24108497] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by wasting of muscles that leads to difficulty moving and premature death, mainly from heart failure. Glucocorticoids are applied in the management of the disease, supporting the hypothesis that inflammation may be driver as well as target. However, the inflammatory mechanisms during progression of cardiac and skeletal muscle dysfunction are still not well characterized. Our objective was to characterize the inflammasomes in myocardial and skeletal muscle in rodent models of DMD. Gastrocnemius and heart samples were collected from mdx mice and DMDmdx rats (3 and 9-10 months). Inflammasome sensors and effectors were assessed by immunoblotting. Histology was used to assess leukocyte infiltration and fibrosis. In gastrocnemius, a tendency towards elevation of gasdermin D irrespective of the age of the animal was observed. The adaptor protein was elevated in the mdx mouse skeletal muscle and heart. Increased cleavage of the cytokines was observed in the skeletal muscle of the DMDmdx rats. Sensor or cytokine expression was not changed in the tissue samples of the mdx mice. In conclusion, inflammatory responses are distinct between the skeletal muscle and heart in relevant models of DMD. Inflammation tends to decrease over time, supporting the clinical observations that the efficacy of anti-inflammatory therapies might be more prominent in the early stage.
Collapse
Affiliation(s)
- Zsófia Onódi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Z.O.)
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Petra Lujza Szabó
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Dániel Kucsera
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Z.O.)
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Péter Pokreisz
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Christopher Dostal
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Karlheinz Hilber
- Department of Neurophysiology & Neuropharmacology, Center for Physiology & Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Gavin Y. Oudit
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Bruno K. Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Z.O.)
- Pharmahungary Group, 6728 Szeged, Hungary
| | - Zoltán V. Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Z.O.)
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
6
|
Ueda J, Saito S. Evaluation of Cardiac Function in Young Mdx Mice Using MRI with Feature Tracking and Self-Gated Magnetic Resonance Cine Imaging. Diagnostics (Basel) 2023; 13:diagnostics13081472. [PMID: 37189573 DOI: 10.3390/diagnostics13081472] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/31/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
This study aimed to evaluate cardiac function in a young mouse model of Duchenne muscular dystrophy (mdx) using cardiac magnetic resonance imaging (MRI) with feature tracking and self-gated magnetic resonance cine imaging. Cardiac function was evaluated in mdx and control mice (C57BL/6JJmsSlc mice) at 8 and 12 weeks of age. Preclinical 7-T MRI was used to capture short-axis, longitudinal two-chamber view and longitudinal four-chamber view cine images of mdx and control mice. Strain values were measured and evaluated from cine images acquired using the feature tracking method. The left ventricular ejection fraction was significantly less (p < 0.01 each) in the mdx group at both 8 (control, 56.6 ± 2.3% mdx, 47.2 ± 7.4%) and 12 weeks (control, 53.9 ± 3.3% mdx, 44.1 ± 2.7%). In the strain analysis, all strain value peaks were significantly less in mdx mice, except for the longitudinal strain of the four-chamber view at both 8 and 12 weeks of age. Strain analysis with feature tracking and self-gated magnetic resonance cine imaging is useful for assessing cardiac function in young mdx mice.
Collapse
Affiliation(s)
- Junpei Ueda
- Department of Medical Physics and Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita 560-0871, Osaka, Japan
| | - Shigeyoshi Saito
- Department of Medical Physics and Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita 560-0871, Osaka, Japan
- Department of Advanced Medical Technologies, National Cardiovascular and Cerebral Research Center, Suita 564-8565, Osaka, Japan
| |
Collapse
|
7
|
Oliveira-Santos A, Dagda M, Burkin DJ. Sunitinib inhibits STAT3 phosphorylation in cardiac muscle and prevents cardiomyopathy in the mdx mouse model of Duchenne muscular dystrophy. Hum Mol Genet 2022; 31:2358-2369. [PMID: 35157045 PMCID: PMC9307308 DOI: 10.1093/hmg/ddac042] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/31/2022] [Accepted: 02/09/2022] [Indexed: 11/14/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal X-linked genetic disorder affecting approximately 1 in 5000 male births worldwide. DMD is caused by mutations in the dystrophin gene. Dystrophin is essential for maintaining muscle cell membrane integrity and stability by linking the cytoskeleton to the extracellular matrix, which protects myofibers from contraction-induced damage. Loss of dystrophin leads to mechanically induced skeletal and cardiac muscle damage. Although the disease is not evident in DMD patients at birth, muscular dystrophy rapidly progresses and results in respiratory and cardiac muscle failure as early as the teenage years. Premature death in DMD patients is due to cardiac arrhythmias and left ventricular dysfunction. Currently, there is no effective treatment for DMD-related cardiac failure. Recently, we have shown that a Food and Drug Administration-approved small molecule, sunitinib, a multi-targeted tyrosine kinase inhibitor can mitigate skeletal muscle disease through an increase in myogenic capacity, cell membrane integrity, and improvement of skeletal muscle function via regulation of STAT3-related signaling pathway. Chronic activation of STAT3 has been shown to promote cardiac hypertrophy and failure. In this study, we examined the effects of long-term sunitinib treatment on cardiac pathology and function. Our results showed sunitinib treatment reduced STAT3 phosphorylation in the heart muscle of mdx mice, improved cardiac electrical function, increased cardiac output and stroke volume, decreased ventricular hypertrophy, reduced cardiomyocytes membrane damage, fibrotic tissue deposition and slightly decreased cardiac inflammation. Together, our studies support the idea that sunitinib could serve as a novel treatment to slow cardiomyopathy progression in DMD. One Sentence Summary In this study, we determined if sunitinib, a Food and Drug Administration-approved drug, could reduce the pathology and improve cardiac function in an animal model for DMD.
Collapse
Affiliation(s)
- Ariany Oliveira-Santos
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno NV 89557, USA
| | - Marisela Dagda
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno NV 89557, USA
| | - Dean J Burkin
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno NV 89557, USA
| |
Collapse
|
8
|
de Zélicourt A, Fayssoil A, Dakouane-Giudicelli M, De Jesus I, Karoui A, Zarrouki F, Lefebvre F, Mansart A, Launay JM, Piquereau J, Tarragó MG, Bonay M, Forand A, Moog S, Piétri-Rouxel F, Brisebard E, Chini CCS, Kashyap S, Fogarty MJ, Sieck GC, Mericskay M, Chini EN, Gomez AM, Cancela JM, de la Porte S. CD38-NADase is a new major contributor to Duchenne muscular dystrophic phenotype. EMBO Mol Med 2022; 14:e12860. [PMID: 35298089 PMCID: PMC9081905 DOI: 10.15252/emmm.202012860] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 01/14/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by progressive muscle degeneration. Two important deleterious features are a Ca2+ dysregulation linked to Ca2+ influxes associated with ryanodine receptor hyperactivation, and a muscular nicotinamide adenine dinucleotide (NAD+) deficit. Here, we identified that deletion in mdx mice of CD38, a NAD+ glycohydrolase‐producing modulators of Ca2+ signaling, led to a fully restored heart function and structure, with skeletal muscle performance improvements, associated with a reduction in inflammation and senescence markers. Muscle NAD+ levels were also fully restored, while the levels of the two main products of CD38, nicotinamide and ADP‐ribose, were reduced, in heart, diaphragm, and limb. In cardiomyocytes from mdx/CD38−/− mice, the pathological spontaneous Ca2+ activity was reduced, as well as in myotubes from DMD patients treated with isatuximab (SARCLISA®) a monoclonal anti‐CD38 antibody. Finally, treatment of mdx and utrophin–dystrophin‐deficient (mdx/utr−/−) mice with CD38 inhibitors resulted in improved skeletal muscle performances. Thus, we demonstrate that CD38 actively contributes to DMD physiopathology. We propose that a selective anti‐CD38 therapeutic intervention could be highly relevant to develop for DMD patients.
Collapse
Affiliation(s)
- Antoine de Zélicourt
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, Versailles, France.,Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, Saclay, France
| | | | | | - Isley De Jesus
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, Versailles, France
| | - Ahmed Karoui
- Signalisation et Physiopathologie Cardiovasculaire, INSERM, UMR-S 1180 - Université Paris-Saclay, Châtenay-Malabry, France
| | - Faouzi Zarrouki
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, Versailles, France
| | - Florence Lefebvre
- Signalisation et Physiopathologie Cardiovasculaire, INSERM, UMR-S 1180 - Université Paris-Saclay, Châtenay-Malabry, France
| | - Arnaud Mansart
- Université Paris-Saclay, UVSQ, Inserm, 2I, Versailles, France
| | - Jean-Marie Launay
- Service de Biochimie, INSERM UMR S942, Hôpital Lariboisière, Paris, France
| | - Jerome Piquereau
- Signalisation et Physiopathologie Cardiovasculaire, INSERM, UMR-S 1180 - Université Paris-Saclay, Châtenay-Malabry, France
| | - Mariana G Tarragó
- Department of Anesthesiology and Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Marcel Bonay
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, Versailles, France
| | - Anne Forand
- Centre de Recherche en Myologie, Faculté de Médecine de la Pitié Salpêtrière, Sorbonne Université-UMRS974-Inserm-Institut de Myologie, Paris, France.,Inovarion, Paris, France
| | - Sophie Moog
- Centre de Recherche en Myologie, Faculté de Médecine de la Pitié Salpêtrière, Sorbonne Université-UMRS974-Inserm-Institut de Myologie, Paris, France.,Inovarion, Paris, France
| | - France Piétri-Rouxel
- Centre de Recherche en Myologie, Faculté de Médecine de la Pitié Salpêtrière, Sorbonne Université-UMRS974-Inserm-Institut de Myologie, Paris, France
| | | | - Claudia C S Chini
- Department of Anesthesiology and Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Sonu Kashyap
- Department of Anesthesiology and Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew J Fogarty
- Department of Anesthesiology and Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Gary C Sieck
- Department of Anesthesiology and Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Mathias Mericskay
- Signalisation et Physiopathologie Cardiovasculaire, INSERM, UMR-S 1180 - Université Paris-Saclay, Châtenay-Malabry, France
| | - Eduardo N Chini
- Department of Anesthesiology and Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Ana Maria Gomez
- Signalisation et Physiopathologie Cardiovasculaire, INSERM, UMR-S 1180 - Université Paris-Saclay, Châtenay-Malabry, France
| | - José-Manuel Cancela
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, Saclay, France
| | | |
Collapse
|
9
|
He X, Liu J, Gu F, Chen J, Lu YW, Ding J, Guo H, Nie M, Kataoka M, Lin Z, Hu X, Chen H, Liao X, Dong Y, Min W, Deng ZL, Pu WT, Huang ZP, Wang DZ. Cardiac CIP protein regulates dystrophic cardiomyopathy. Mol Ther 2022; 30:898-914. [PMID: 34400329 PMCID: PMC8822131 DOI: 10.1016/j.ymthe.2021.08.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 05/24/2021] [Accepted: 08/08/2021] [Indexed: 02/04/2023] Open
Abstract
Heart failure is a leading cause of fatality in Duchenne muscular dystrophy (DMD) patients. Previously, we discovered that cardiac and skeletal-muscle-enriched CIP proteins play important roles in cardiac function. Here, we report that CIP, a striated muscle-specific protein, participates in the regulation of dystrophic cardiomyopathy. Using a mouse model of human DMD, we found that deletion of CIP leads to dilated cardiomyopathy and heart failure in young, non-syndromic mdx mice. Conversely, transgenic overexpression of CIP reduces pathological dystrophic cardiomyopathy in old, syndromic mdx mice. Genome-wide transcriptome analyses reveal that molecular pathways involving fibrogenesis and oxidative stress are affected in CIP-mediated dystrophic cardiomyopathy. Mechanistically, we found that CIP interacts with dystrophin and calcineurin (CnA) to suppress the CnA-Nuclear Factor of Activated T cells (NFAT) pathway, which results in decreased expression of Nox4, a key component of the oxidative stress pathway. Overexpression of Nox4 accelerates the development of dystrophic cardiomyopathy in mdx mice. Our study indicates CIP is a modifier of dystrophic cardiomyopathy and a potential therapeutic target for this devastating disease.
Collapse
Affiliation(s)
- Xin He
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Jianming Liu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Fei Gu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Jinghai Chen
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yao Wei Lu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Jian Ding
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Haipeng Guo
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Critical Care and Emergency Medicine, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Mao Nie
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Masaharu Kataoka
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Zhiqiang Lin
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Huaqun Chen
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Biology, Nanjing Normal University, Nanjing, China
| | - Xinxue Liao
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Yugang Dong
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Wang Min
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhong-Liang Deng
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China.
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
10
|
Laurila PP, Luan P, Wohlwend M, Zanou N, Crisol B, Imamura de Lima T, Goeminne LJE, Gallart-Ayala H, Shong M, Ivanisevic J, Place N, Auwerx J. Inhibition of sphingolipid de novo synthesis counteracts muscular dystrophy. SCIENCE ADVANCES 2022; 8:eabh4423. [PMID: 35089797 PMCID: PMC8797791 DOI: 10.1126/sciadv.abh4423] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 11/01/2021] [Indexed: 05/29/2023]
Abstract
Duchenne muscular dystrophy (DMD), the most common muscular dystrophy, is a severe muscle disorder, causing muscle weakness, loss of independence, and premature death. Here, we establish the link between sphingolipids and muscular dystrophy. Transcripts of sphingolipid de novo biosynthesis pathway are up-regulated in skeletal muscle of patients with DMD and other muscular dystrophies, which is accompanied by accumulation of metabolites of the sphingolipid pathway in muscle and plasma. Pharmacological inhibition of sphingolipid synthesis by myriocin in the mdx mouse model of DMD ameliorated the loss in muscle function while reducing inflammation, improving Ca2+ homeostasis, preventing fibrosis of the skeletal muscle, heart, and diaphragm, and restoring the balance between M1 and M2 macrophages. Myriocin alleviated the DMD phenotype more than glucocorticoids. Our study identifies inhibition of sphingolipid synthesis, targeting multiple pathogenetic pathways simultaneously, as a strong candidate for treatment of muscular dystrophies.
Collapse
Affiliation(s)
- Pirkka-Pekka Laurila
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Peiling Luan
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Martin Wohlwend
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences, Department of Physiology, Faculty of Biology-Medicine, University of Lausanne, Lausanne, Switzerland
| | - Barbara Crisol
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tanes Imamura de Lima
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ludger J. E. Goeminne
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Nicolas Place
- Institute of Sport Sciences, Department of Physiology, Faculty of Biology-Medicine, University of Lausanne, Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
11
|
Deficiency of MMP-10 Aggravates the Diseased Phenotype of Aged Dystrophic Mice. Life (Basel) 2021; 11:life11121398. [PMID: 34947929 PMCID: PMC8705381 DOI: 10.3390/life11121398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/05/2021] [Accepted: 12/08/2021] [Indexed: 01/03/2023] Open
Abstract
Matrix metalloproteinases (MMPs) have been implicated in the progression of muscular dystrophy, and recent studies have reported the role of MMP-10 in skeletal muscle pathology of young dystrophic mice. Nevertheless, its involvement in dystrophin-deficient hearts remains unexplored. Here, we aimed to investigate the involvement of MMP-10 in the progression of severe muscular dystrophy and to characterize MMP-10 loss in skeletal and cardiac muscles of aged dystrophic mice. We examined the histopathological effect of MMP-10 ablation in aged mdx mice, both in the hind limb muscles and heart tissues. We found that MMP-10 loss compromises survival rates of aged mdx mice, with skeletal and cardiac muscles developing a chronic inflammatory response. Our findings indicate that MMP-10 is implicated in severe muscular dystrophy progression, thus identifying a new area of research that could lead to future therapies for dystrophic muscles.
Collapse
|
12
|
Liu Y, Zhu Y, Liu S, Liu J, Li X. NORAD lentivirus shRNA mitigates fibrosis and inflammatory responses in diabetic cardiomyopathy via the ceRNA network of NORAD/miR-125a-3p/Fyn. Inflamm Res 2021; 70:1113-1127. [PMID: 34591118 DOI: 10.1007/s00011-021-01500-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVE Diabetic cardiomyopathy (DCM) is a serious complication of diabetes, but its pathogenesis is still unclear. This study investigated the mechanism of long noncoding RNA (lncRNA) NORAD in DCM. METHODS Male leptin receptor-deficient (db/db) mice and leptin control mice (db/ +) were procured. DCM model was established by subcutaneous injection of angiotensin II (ATII) in db/db mice. NORAD lentivirus shRNA or Adv-miR-125a-3p was administered to analyze cardiac function, fibrosis, serum biochemical indexes, inflammation and fibrosis. Primary cardiomyocytes were extracted and transfected with miR-125a-3p mimic. The competing endogenous RNA (ceRNA) network of NORAD/miR-125a-3p/Fyn was verified. The levels of fibrosis- and inflammation-related factors were measured. RESULTS In db/db mice treated with ATII, the body weight and serum biochemical indexes were increased, while the cardiac function was decreased, and inflammatory cell infiltration and fibrosis were induced. NORAD was upregulated in diabetic and DCM mice. The 4-week intravenous injection of NORAD lentivirus shRNA reduced body weight and serum biochemical indexes, improved cardiac function, and attenuated inflammation and fibrosis in DCM mice. NORAD acted as a sponge to adsorb miR-125a-3p, and miR-125a-3p targeted Fyn. Intravenous injection of miR-125a-3p adenovirus improved cardiac function and fibrosis and reduced inflammatory responses in DCM mice. Co-overexpression of miR-125-3p and Fyn partly reversed the improving effect of miR-125-3p overexpression on cardiac fibrosis in DCM mice. CONCLUSION NORAD lentivirus shRNA improved cardiac function and fibrosis and reduced inflammatory responses in DCM mice via the ceRNA network of NORAD/miR-125a-3p/Fyn. These findings provide a valuable and promising therapeutic target for the treatment of DCM.
Collapse
Affiliation(s)
- Ye Liu
- Department of Endocrinology, The Second Hospital of ShanXi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Yikun Zhu
- Department of Endocrinology, The Second Hospital of ShanXi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Sujun Liu
- Department of Endocrinology, The Second Hospital of ShanXi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Jiong Liu
- Department of Nuclear Medicine, The Second Hospital of ShanXi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Xing Li
- Department of Endocrinology, The Second Hospital of ShanXi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
13
|
Rebolledo DL, Lipson KE, Brandan E. Driving fibrosis in neuromuscular diseases: Role and regulation of Connective tissue growth factor (CCN2/CTGF). Matrix Biol Plus 2021; 11:100059. [PMID: 34435178 PMCID: PMC8377001 DOI: 10.1016/j.mbplus.2021.100059] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Connective tissue growth factor or cellular communication network 2 (CCN2/CTGF) is a matricellular protein member of the CCN family involved in several crucial biological processes. In skeletal muscle, CCN2/CTGF abundance is elevated in human muscle biopsies and/or animal models for diverse neuromuscular pathologies, including muscular dystrophies, neurodegenerative disorders, muscle denervation, and muscle overuse. In this context, CCN2/CTGF is deeply involved in extracellular matrix (ECM) modulation, acting as a strong pro-fibrotic factor that promotes excessive ECM accumulation. Reducing CCN2/CTGF levels or biological activity in pathological conditions can decrease fibrosis, improve muscle architecture and function. In this work, we summarize information about the role of CCN2/CTGF in fibrosis associated with neuromuscular pathologies and the mechanisms and signaling pathways that regulate their expression in skeletal muscle.
Collapse
Affiliation(s)
- Daniela L Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Punta Arenas, Chile
| | | | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Chile.,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile.,Fundación Ciencia y Vida, Santiago, Chile
| |
Collapse
|
14
|
Accelerating the Mdx Heart Histo-Pathology through Physical Exercise. Life (Basel) 2021; 11:life11070706. [PMID: 34357078 PMCID: PMC8306456 DOI: 10.3390/life11070706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023] Open
Abstract
Chronic cardiac muscle inflammation and fibrosis are key features of Duchenne Muscular Dystrophy (DMD). Around 90% of 18-year-old patients already show signs of DMD-related cardiomyopathy, and cardiac failure is rising as the main cause of death among DMD patients. The evaluation of novel therapies for the treatment of dystrophic heart problems depends on the availability of animal models that closely mirror the human pathology. The widely used DMD animal model, the mdx mouse, presents a milder cardiac pathology compared to humans, with a late onset, which precludes large-scale and reliable studies. In this study, we used an exercise protocol to accelerate and worsen the cardiac pathology in mdx mice. The mice were subjected to a 1 h-long running session on a treadmill, at moderate speed, twice a week for 8 weeks. We demonstrate that subjecting young mdx mice (4-week-old) to "endurance" exercise accelerates heart pathology progression, as shown by early fibrosis deposition, increases necrosis and inflammation, and reduces heart function compared to controls. We believe that our exercised mdx model represents an easily reproducible and useful tool to study the molecular and cellular networks involved in dystrophic heart alterations, as well as to evaluate novel therapeutic strategies aimed at ameliorating dystrophic heart pathology.
Collapse
|
15
|
QiShenYiQi Pill Improves Myocardial Hypertrophy Caused by Pressure Overload in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5536723. [PMID: 34221074 PMCID: PMC8225423 DOI: 10.1155/2021/5536723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/17/2021] [Accepted: 06/07/2021] [Indexed: 01/19/2023]
Abstract
Pressure-overloaded myocardial hypertrophy is an independent risk factor for various cardiovascular diseases (CVDs), such as heart failure (HF), arrhythmia, and even sudden death. It is reported that QiShenYiQi pill (QSYQ) is widely used in the treatment of CVDs and can prevent pathological hypertrophy of myocardium, but its specific mechanism is still unclear. In this study, a rat model of myocardial hypertrophy was established through the pressure overload caused by abdominal aortic constriction in Wistar rats. The rats were randomly divided into model group, valsartan group, and QSYQ group, and sham-operated animals served as the control group. At the 4 and 8 weeks of intervention, the general morphology of the heart, myocardial collagen content, collagen volume factor (CVF), collagen type I, collagen type III, myocardial pathological changes, and the expression of ANP, β-MHC, TGF-β1, and CTGF were analyzed, respectively, in order to explore the possible effect of QSYQ on the mechanism of myocardial hypertrophy. We observed that QSYQ could effectively improve myocardial hypertrophy in pressure-overloaded rats, which was related to the regulatory mechanism of TGF-β1 and CTGF.
Collapse
|
16
|
Rebolledo DL, Acuña MJ, Brandan E. Role of Matricellular CCN Proteins in Skeletal Muscle: Focus on CCN2/CTGF and Its Regulation by Vasoactive Peptides. Int J Mol Sci 2021; 22:5234. [PMID: 34063397 PMCID: PMC8156781 DOI: 10.3390/ijms22105234] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/28/2021] [Accepted: 05/12/2021] [Indexed: 02/08/2023] Open
Abstract
The Cellular Communication Network (CCN) family of matricellular proteins comprises six proteins that share conserved structural features and play numerous biological roles. These proteins can interact with several receptors or soluble proteins, regulating cell signaling pathways in various tissues under physiological and pathological conditions. In the skeletal muscle of mammals, most of the six CCN family members are expressed during embryonic development or in adulthood. Their roles during the adult stage are related to the regulation of muscle mass and regeneration, maintaining vascularization, and the modulation of skeletal muscle fibrosis. This work reviews the CCNs proteins' role in skeletal muscle physiology and disease, focusing on skeletal muscle fibrosis and its regulation by Connective Tissue Growth factor (CCN2/CTGF). Furthermore, we review evidence on the modulation of fibrosis and CCN2/CTGF by the renin-angiotensin system and the kallikrein-kinin system of vasoactive peptides.
Collapse
Affiliation(s)
- Daniela L. Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6213515, Chile
| | - María José Acuña
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, Santiago 8370854, Chile
| | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Fundación Ciencia & Vida, Santiago 7810000, Chile
| |
Collapse
|
17
|
Jelinkova S, Sleiman Y, Fojtík P, Aimond F, Finan A, Hugon G, Scheuermann V, Beckerová D, Cazorla O, Vincenti M, Amedro P, Richard S, Jaros J, Dvorak P, Lacampagne A, Carnac G, Rotrekl V, Meli AC. Dystrophin Deficiency Causes Progressive Depletion of Cardiovascular Progenitor Cells in the Heart. Int J Mol Sci 2021; 22:ijms22095025. [PMID: 34068508 PMCID: PMC8125982 DOI: 10.3390/ijms22095025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 11/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating condition shortening the lifespan of young men. DMD patients suffer from age-related dilated cardiomyopathy (DCM) that leads to heart failure. Several molecular mechanisms leading to cardiomyocyte death in DMD have been described. However, the pathological progression of DMD-associated DCM remains unclear. In skeletal muscle, a dramatic decrease in stem cells, so-called satellite cells, has been shown in DMD patients. Whether similar dysfunction occurs with cardiac muscle cardiovascular progenitor cells (CVPCs) in DMD remains to be explored. We hypothesized that the number of CVPCs decreases in the dystrophin-deficient heart with age and disease state, contributing to DCM progression. We used the dystrophin-deficient mouse model (mdx) to investigate age-dependent CVPC properties. Using quantitative PCR, flow cytometry, speckle tracking echocardiography, and immunofluorescence, we revealed that young mdx mice exhibit elevated CVPCs. We observed a rapid age-related CVPC depletion, coinciding with the progressive onset of cardiac dysfunction. Moreover, mdx CVPCs displayed increased DNA damage, suggesting impaired cardiac muscle homeostasis. Overall, our results identify the early recruitment of CVPCs in dystrophic hearts and their fast depletion with ageing. This latter depletion may participate in the fibrosis development and the acceleration onset of the cardiomyopathy.
Collapse
MESH Headings
- Aging/genetics
- Aging/pathology
- Animals
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Cardiovascular System/metabolism
- Cardiovascular System/pathology
- DNA Damage/genetics
- Disease Models, Animal
- Dystrophin/deficiency
- Dystrophin/genetics
- Gene Expression Regulation/genetics
- Humans
- Mice
- Mice, Inbred mdx/genetics
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Proto-Oncogene Proteins c-kit/genetics
- Stem Cells/metabolism
- Stem Cells/pathology
Collapse
Affiliation(s)
- Sarka Jelinkova
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
| | - Yvonne Sleiman
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Petr Fojtík
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
| | - Franck Aimond
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Amanda Finan
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Gerald Hugon
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Valerie Scheuermann
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Deborah Beckerová
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
| | - Olivier Cazorla
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Marie Vincenti
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
- Pediatric and Adult Congenital Cardiology Department, M3C Regional Reference CHD Center, CHU Montpellier, 371 Avenue du Doyen Giraud, 34295 Montpellier, France
| | - Pascal Amedro
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
- Pediatric and Adult Congenital Cardiology Department, M3C Regional Reference CHD Center, CHU Montpellier, 371 Avenue du Doyen Giraud, 34295 Montpellier, France
| | - Sylvain Richard
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Josef Jaros
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5/A1, 62500 Brno, Czech Republic
| | - Petr Dvorak
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
| | - Alain Lacampagne
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Gilles Carnac
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Vladimir Rotrekl
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
- Correspondence: (V.R.); (A.C.M.); Tel.: +420-549-498-002 (V.R.); +33-4-67-41-52-44 (A.C.M.); Fax: +420-549-491-327 (V.R.); +33-4-67-41-52-42 (A.C.M.)
| | - Albano C. Meli
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
- Correspondence: (V.R.); (A.C.M.); Tel.: +420-549-498-002 (V.R.); +33-4-67-41-52-44 (A.C.M.); Fax: +420-549-491-327 (V.R.); +33-4-67-41-52-42 (A.C.M.)
| |
Collapse
|
18
|
Low human dystrophin levels prevent cardiac electrophysiological and structural remodelling in a Duchenne mouse model. Sci Rep 2021; 11:9779. [PMID: 33963238 PMCID: PMC8105358 DOI: 10.1038/s41598-021-89208-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 04/15/2021] [Indexed: 02/03/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive neuromuscular disorder caused by loss of dystrophin. This lack also affects cardiac structure and function, and cardiovascular complications are a major cause of death in DMD. Newly developed therapies partially restore dystrophin expression. It is unclear whether this will be sufficient to prevent or ameliorate cardiac involvement in DMD. We here establish the cardiac electrophysiological and structural phenotype in young (2-3 months) and aged (6-13 months) dystrophin-deficient mdx mice expressing 100% human dystrophin (hDMD), 0% human dystrophin (hDMDdel52-null) or low levels (~ 5%) of human dystrophin (hDMDdel52-low). Compared to hDMD, young and aged hDMDdel52-null mice displayed conduction slowing and repolarisation abnormalities, while only aged hDMDdel52-null mice displayed increased myocardial fibrosis. Moreover, ventricular cardiomyocytes from young hDMDdel52-null animals displayed decreased sodium current and action potential (AP) upstroke velocity, and prolonged AP duration at 20% and 50% of repolarisation. Hence, cardiac electrical remodelling in hDMDdel52-null mice preceded development of structural alterations. In contrast to hDMDdel52-null, hDMDdel52-low mice showed similar electrophysiological and structural characteristics as hDMD, indicating prevention of the cardiac DMD phenotype by low levels of human dystrophin. Our findings are potentially relevant for the development of therapeutic strategies aimed at restoring dystrophin expression in DMD.
Collapse
|
19
|
Maino E, Wojtal D, Evagelou SL, Farheen A, Wong TWY, Lindsay K, Scott O, Rizvi SZ, Hyatt E, Rok M, Visuvanathan S, Chiodo A, Schneeweiss M, Ivakine EA, Cohn RD. Targeted genome editing in vivo corrects a Dmd duplication restoring wild-type dystrophin expression. EMBO Mol Med 2021; 13:e13228. [PMID: 33724658 PMCID: PMC8103086 DOI: 10.15252/emmm.202013228] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 02/06/2021] [Accepted: 02/10/2021] [Indexed: 12/26/2022] Open
Abstract
Tandem duplication mutations are increasingly found to be the direct cause of many rare heritable diseases, accounting for up to 10% of cases. Unfortunately, animal models recapitulating such mutations are scarce, limiting our ability to study them and develop genome editing therapies. Here, we describe the generation of a novel duplication mouse model, harboring a multi-exonic tandem duplication in the Dmd gene which recapitulates a human mutation. Duplication correction of this mouse was achieved by implementing a single-guide RNA (sgRNA) CRISPR/Cas9 approach. This strategy precisely removed a duplication mutation in vivo, restored full-length dystrophin expression, and was accompanied by improvements in both histopathological and clinical phenotypes. We conclude that CRISPR/Cas9 represents a powerful tool to accurately model and treat tandem duplication mutations. Our findings will open new avenues of research for exploring the study and therapeutics of duplication disorders.
Collapse
Affiliation(s)
- Eleonora Maino
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Daria Wojtal
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Sonia L Evagelou
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Aiman Farheen
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Tatianna W Y Wong
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Kyle Lindsay
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Ori Scott
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
- Department of Pediatricsthe Hospital for Sick ChildrenTorontoONCanada
| | - Samar Z Rizvi
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Elzbieta Hyatt
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Matthew Rok
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Shagana Visuvanathan
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Amanda Chiodo
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Michelle Schneeweiss
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Evgueni A Ivakine
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of PhysiologyUniversity of TorontoTorontoONCanada
| | - Ronald D Cohn
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
- Department of Pediatricsthe Hospital for Sick ChildrenTorontoONCanada
| |
Collapse
|
20
|
Lindsay A, Kemp B, Larson AA, Baumann CW, McCourt PM, Holm J, Karachunski P, Lowe DA, Ervasti JM. Tetrahydrobiopterin synthesis and metabolism is impaired in dystrophin-deficient mdx mice and humans. Acta Physiol (Oxf) 2021; 231:e13627. [PMID: 33580591 DOI: 10.1111/apha.13627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022]
Abstract
AIM Loss of dystrophin causes oxidative stress and affects nitric oxide synthase-mediated vascular function in striated muscle. Because tetrahydrobiopterin is an antioxidant and co-factor for nitric oxide synthase, we tested the hypothesis that tetrahydrobiopterin would be low in mdx mice and humans deficient for dystrophin. METHODS Tetrahydrobiopterin and its metabolites were measured at rest and in response to exercise in Duchenne and Becker muscular dystrophy patients, age-matched male controls as well as wild-type, mdx and mdx mice transgenically overexpressing skeletal muscle-specific dystrophins. Mdx mice were also supplemented with tetrahydrobiopterin and pathophysiology was assessed. RESULTS Duchenne muscular dystrophy patients had lower urinary dihydrobiopterin + tetrahydrobiopterin/specific gravity1.020 compared to unaffected age-matched males and Becker muscular dystrophy patients. Mdx mice had low urinary and skeletal muscle dihydrobiopterin + tetrahydrobiopterin compared to wild-type mice. Overexpression of dystrophins that localize neuronal nitric oxide synthase restored dihydrobiopterin + tetrahydrobiopterin in mdx mice to wild-type levels while utrophin overexpression did not. Mdx mice and Duchenne muscular dystrophy patients did not increase tetrahydrobiopterin during exercise and in mdx mice tetrahydrobiopterin deficiency was likely because of lower levels of sepiapterin reductase in skeletal muscle. Tetrahydrobiopterin supplementation improved skeletal muscle strength, resistance to fatiguing and injurious contractions in vivo, increased utrophin and capillary density of skeletal muscle and lowered cardiac muscle fibrosis and left ventricular wall thickness in mdx mice. CONCLUSION These data demonstrate that impaired tetrahydrobiopterin synthesis is associated with dystrophin loss and treatment with tetrahydrobiopterin improves striated muscle histopathology and skeletal muscle function in mdx mice.
Collapse
Affiliation(s)
- Angus Lindsay
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Bailey Kemp
- Lillehei Heart Institute, Cancer and Cardiovascular Research Center, University of Minnesota, Minneapolis, MN, USA
| | - Alexie A Larson
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Cory W Baumann
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Preston M McCourt
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - John Holm
- Lillehei Heart Institute, Cancer and Cardiovascular Research Center, University of Minnesota, Minneapolis, MN, USA
| | - Peter Karachunski
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Dawn A Lowe
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
21
|
Rodriguez-Gonzalez M, Lubian-Gutierrez M, Cascales-Poyatos HM, Perez-Reviriego AA, Castellano-Martinez A. Role of the Renin-Angiotensin-Aldosterone System in Dystrophin-Deficient Cardiomyopathy. Int J Mol Sci 2020; 22:ijms22010356. [PMID: 33396334 PMCID: PMC7796305 DOI: 10.3390/ijms22010356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 12/27/2020] [Accepted: 12/28/2020] [Indexed: 12/16/2022] Open
Abstract
Dystrophin-deficient cardiomyopathy (DDC) is currently the leading cause of death in patients with dystrophinopathies. Targeting myocardial fibrosis (MF) has become a major therapeutic goal in order to prevent the occurrence of DDC. We aimed to review and summarize the current evidence about the role of the renin-angiotensin-aldosterone system (RAAS) in the development and perpetuation of MF in DCC. We conducted a comprehensive search of peer-reviewed English literature on PubMed about this subject. We found increasing preclinical evidence from studies in animal models during the last 20 years pointing out a central role of RAAS in the development of MF in DDC. Local tissue RAAS acts directly mainly through its main fibrotic component angiotensin II (ANG2) and its transducer receptor (AT1R) and downstream TGF-b pathway. Additionally, it modulates the actions of most of the remaining pro-fibrotic factors involved in DDC. Despite limited clinical evidence, RAAS blockade constitutes the most studied, available and promising therapeutic strategy against MF and DDC. Conclusion: Based on the evidence reviewed, it would be recommendable to start RAAS blockade therapy through angiotensin converter enzyme inhibitors (ACEI) or AT1R blockers (ARBs) alone or in combination with mineralocorticoid receptor antagonists (MRa) at the youngest age after the diagnosis of dystrophinopathies, in order to delay the occurrence or slow the progression of MF, even before the detection of any cardiovascular alteration.
Collapse
Affiliation(s)
- Moises Rodriguez-Gonzalez
- Pediatric Cardiology Division of Puerta del Mar University Hospital, University of Cadiz, 11009 Cadiz, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Research Unit, Puerta del Mar University Hospital, University of Cadiz, 11009 Cadiz, Spain;
- Correspondence: ; Tel.: +34-956002700
| | - Manuel Lubian-Gutierrez
- Pediatric Neurology Division of Puerta del Mar University Hospital, University of Cadiz, 11009 Cadiz, Spain;
- Pediatric Division of Doctor Cayetano Roldan Primary Care Center, 11100 San Fernando, Spain
| | | | | | - Ana Castellano-Martinez
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Research Unit, Puerta del Mar University Hospital, University of Cadiz, 11009 Cadiz, Spain;
- Pediatric Nephrology Division of Puerta del Mar University Hospital, University of Cadiz, 11009 Cadiz, Spain
| |
Collapse
|
22
|
Wong TWY, Ahmed A, Yang G, Maino E, Steiman S, Hyatt E, Chan P, Lindsay K, Wong N, Golebiowski D, Schneider J, Delgado-Olguín P, Ivakine EA, Cohn RD. A novel mouse model of Duchenne muscular dystrophy carrying a multi-exonic Dmd deletion exhibits progressive muscular dystrophy and early-onset cardiomyopathy. Dis Model Mech 2020; 13:13/9/dmm045369. [PMID: 32988972 PMCID: PMC7522028 DOI: 10.1242/dmm.045369] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/20/2020] [Indexed: 12/14/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a life-threatening neuromuscular disease caused by the lack of dystrophin, resulting in progressive muscle wasting and locomotor dysfunctions. By adulthood, almost all patients also develop cardiomyopathy, which is the primary cause of death in DMD. Although there has been extensive effort in creating animal models to study treatment strategies for DMD, most fail to recapitulate the complete skeletal and cardiac disease manifestations that are presented in affected patients. Here, we generated a mouse model mirroring a patient deletion mutation of exons 52-54 (Dmd Δ52-54). The Dmd Δ52-54 mutation led to the absence of dystrophin, resulting in progressive muscle deterioration with weakened muscle strength. Moreover, Dmd Δ52-54 mice present with early-onset hypertrophic cardiomyopathy, which is absent in current pre-clinical dystrophin-deficient mouse models. Therefore, Dmd Δ52-54 presents itself as an excellent pre-clinical model to evaluate the impact on skeletal and cardiac muscles for both mutation-dependent and -independent approaches.
Collapse
Affiliation(s)
- Tatianna Wai Ying Wong
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Abdalla Ahmed
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada.,Program in Translational Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Grace Yang
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Eleonora Maino
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sydney Steiman
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Elzbieta Hyatt
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Parry Chan
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Kyle Lindsay
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Nicole Wong
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | | | | | - Paul Delgado-Olguín
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada.,Program in Translational Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Evgueni A Ivakine
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada.,Department of Physiology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ronald D Cohn
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.,Department of Pediatrics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.,Department of Pediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
23
|
Valle-Tenney R, Rebolledo D, Acuña MJ, Brandan E. HIF-hypoxia signaling in skeletal muscle physiology and fibrosis. J Cell Commun Signal 2020; 14:147-158. [PMID: 32088838 DOI: 10.1007/s12079-020-00553-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
Hypoxia refers to the decrease in oxygen tension in the tissues, and the central effector of the hypoxic response is the transcription factor Hypoxia-Inducible Factor α (HIF1-α). Transient hypoxia in acute events, such as exercising or regeneration after damage, play an important role in skeletal muscle physiology and homeostasis. However, sustained activation of hypoxic signaling is a feature of skeletal muscle injury and disease, which can be a consequence of chronic damage but can also increase the severity of the pathology and worsen its outcome. Here, we review evidence that supports the idea that hypoxia and HIF-1α can contribute to the establishment of fibrosis in skeletal muscle through its crosstalk with other profibrotic factors, such as Transforming growth factor β (TGF-β), the induction of profibrotic cytokines expression, as is the case of Connective Tissue Growth Factor (CTGF/CCN2), or being the target of the Renin-angiotensin system (RAS).
Collapse
Affiliation(s)
- Roger Valle-Tenney
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile.,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.,Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, Santiago, Chile
| | - María José Acuña
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile.,Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, Santiago, Chile
| | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile. .,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Fundación Ciencia & Vida, Santiago, Chile. .,Department Cell and Molecular Biology, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
24
|
Estrada SM, Thagard AS, Dehart MJ, Damicis JR, Dornisch EM, Ippolito DL, Burd I, Napolitano PG, Ieronimakis N. The orphan nuclear receptor Nr4a1 mediates perinatal neuroinflammation in a murine model of preterm labor. Cell Death Dis 2020; 11:11. [PMID: 31907354 PMCID: PMC6944691 DOI: 10.1038/s41419-019-2196-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 12/25/2022]
Abstract
Prematurity is associated with perinatal neuroinflammation and injury. Screening for genetic modulators in an LPS murine model of preterm birth revealed the upregulation of Nr4a1, an orphan nuclear transcription factor that is normally absent or limited in embryonic brains. Concurrently, Nr4a1 was downregulated with magnesium sulfate (MgSO4) and betamethasone (BMTZ) treatments administered to LPS exposed dams. To understand the role of Nr4a1 in perinatal brain injury, we compared the preterm neuroinflammatory response in Nr4a1 knockout (KO) versus wild type (wt) mice. Key inflammatory factors Il1b, Il6 and Tnf, and Iba1+ microglia were significantly lower in Nr4a1 KO versus wt brains exposed to LPS in utero. Treatment with MgSO4/BMTZ mitigated the neuroinflammatory process in wt but not Nr4a1 KO brains. These results correspond with a reduction in cerebral hemorrhage in wt but not mutant embryos from dams given MgSO4/BMTZ. Further analysis with Nr4a1-GFP-Cre × tdTomato loxP reporter mice revealed that the upregulation of Nr4a1 with perinatal neuroinflammation occurs in the cerebral vasculature. Altogether, this study implicates Nr4a1 in the developing vasculature as a potent mediator of neuroinflammatory brain injury that occurs with preterm birth. It is also possible that MgSO4/BMTZ mitigates this process by direct or indirect inhibition of Nr4a1.
Collapse
Affiliation(s)
- Sarah M Estrada
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Madigan Army Medical Center, Tacoma, WA, USA
| | - Andrew S Thagard
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Madigan Army Medical Center, Tacoma, WA, USA
| | - Mary J Dehart
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, USA
| | - Jennifer R Damicis
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, USA
| | - Elisabeth M Dornisch
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, USA
| | | | - Irina Burd
- Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter G Napolitano
- Department of Obstetrics and Gynecology, University of Washington Medical Center, Seattle, WA, USA
| | - Nicholas Ieronimakis
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, USA.
| |
Collapse
|
25
|
Role of hypoxia in skeletal muscle fibrosis: Synergism between hypoxia and TGF-β signaling upregulates CCN2/CTGF expression specifically in muscle fibers. Matrix Biol 2019; 87:48-65. [PMID: 31669521 DOI: 10.1016/j.matbio.2019.09.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 02/06/2023]
Abstract
Several skeletal muscle diseases are characterized by fibrosis, the excessive accumulation of extracellular matrix. Transforming growth factor-β (TGF-β) and connective tissue growth factor (CCN2/CTGF) are two profibrotic factors augmented in fibrotic skeletal muscle, together with signs of reduced vasculature that implies a decrease in oxygen supply. We observed that fibrotic muscles are characterized by the presence of positive nuclei for hypoxia-inducible factor-1α (HIF-1α), a key mediator of the hypoxia response. However, it is not clear how a hypoxic environment could contribute to the fibrotic phenotype in skeletal muscle. We evaluated the role of hypoxia and TGF-β on CCN2 expression in vitro. Fibroblasts, myoblasts and differentiated myotubes were incubated with TGF-β1 under hypoxic conditions. Hypoxia and TGF-β1 induced CCN2 expression synergistically in myotubes but not in fibroblasts or undifferentiated muscle progenitors. This induction requires HIF-1α and the Smad-independent TGF-β signaling pathway. We performed in vivo experiments using pharmacological stabilization of HIF-1α or hypoxia-induced via hindlimb ischemia together with intramuscular injections of TGF-β1, and we found increased CCN2 expression. These observations suggest that hypoxic signaling together with TGF-β signaling, which are both characteristics of a fibrotic skeletal muscle environment, induce the expression of CCN2 in skeletal muscle fibers and myotubes.
Collapse
|
26
|
Rebolledo DL, González D, Faundez-Contreras J, Contreras O, Vio CP, Murphy-Ullrich JE, Lipson KE, Brandan E. Denervation-induced skeletal muscle fibrosis is mediated by CTGF/CCN2 independently of TGF-β. Matrix Biol 2019; 82:20-37. [DOI: 10.1016/j.matbio.2019.01.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/31/2019] [Accepted: 01/31/2019] [Indexed: 02/06/2023]
|
27
|
Rubi L, Todt H, Kubista H, Koenig X, Hilber K. Calcium current properties in dystrophin-deficient ventricular cardiomyocytes from aged mdx mice. Physiol Rep 2018; 6. [PMID: 29333726 PMCID: PMC5789658 DOI: 10.14814/phy2.13567] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 12/11/2017] [Indexed: 11/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), caused by mutations in the gene encoding for the cytoskeletal protein dystrophin, is linked with severe cardiac complications including cardiomyopathy development and cardiac arrhythmias. We and others recently reported that currents through L‐type calcium (Ca) channels were significantly increased, and channel inactivation was reduced in dystrophin‐deficient ventricular cardiomyocytes derived from the mdx mouse, the most commonly used animal model for human DMD. These gain‐of‐function Ca channel abnormalities may enhance the risk of Ca‐dependent arrhythmias and cellular Ca overload in the dystrophic heart. All studies, which have so far investigated L‐type Ca channel properties in dystrophic cardiomyocytes, have used hearts from either neonatal or young adult mdx mice as cell source. In consequence, the dimension of the Ca channel abnormalities present in the severely‐diseased aged dystrophic heart has remained unknown. Here, we have studied potential abnormalities in Ca currents and intracellular Ca transients in ventricular cardiomyocytes derived from aged dystrophic mdx mice. We found that both the L‐type and T‐type Ca current properties of mdx cardiomyocytes were similar to those of myocytes derived from aged wild‐type mice. Accordingly, Ca release from the sarcoplasmic reticulum was normal in cardiomyocytes from aged mdx mice. This suggests that, irrespective of the presence of a pronounced cardiomyopathy in aged mdx mice, Ca currents and Ca release in dystrophic cardiomyocytes are normal. Finally, our data imply that dystrophin‐ regulation of L‐type Ca channel function in the heart is lost during aging.
Collapse
Affiliation(s)
- Lena Rubi
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Hannes Todt
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Helmut Kubista
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Xaver Koenig
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Karlheinz Hilber
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
28
|
Murphy AP, Greally E, O'Hogain D, Blamire A, Caravan P, Straub V. Noninvasive quantification of fibrosis in skeletal and cardiac muscle in mdx mice using EP3533 enhanced magnetic resonance imaging. Magn Reson Med 2018; 81:2728-2735. [PMID: 30394578 DOI: 10.1002/mrm.27578] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/21/2018] [Accepted: 09/30/2018] [Indexed: 12/24/2022]
Abstract
PURPOSE Duchenne muscular dystrophy (DMD) is a genetic condition caused by mutations in the DMD gene leading to muscle degeneration, fatty replacement of muscle cells and fibrosis. A major obstacle to advancing therapeutic research into muscular dystrophies is development of sensitive, noninvasive outcome measures. To date, no validated method to noninvasively quantify fibrosis within skeletal muscle exists. EP3533 is a gadolinium-based MRI contrast agent with an affinity to collagen-1. The purpose of this study was to determine whether EP3533-enhanced MRI could quantify fibrosis in a murine model of DMD (mdx) in muscle. METHODS Mdx (n = 8) and control mice (BL10; n = 5) underwent contrast-enhanced MRI acquisitions with EP3533. T1 mapping pre- and postcontrast was performed in skeletal and cardiac muscle. Post-MRI the tibialis anterior (TA) and gastrocnemius (GCN) muscles and the heart were removed for fibrosis quantification by means of Masson's trichrome staining and the hydroxyproline assay. RESULTS Significant differences in postcontrast R1 were demonstrated between mdx and BL10 mice using EP3533 (cardiac P = 0.02, GCN P = 0.04, TA P = 0.04). Change in R1 from baseline following EP3533 administration correlated strongly to hydroxyproline levels (GCN: r = 0.83, P = 0.001; TA: r = 0.73, P = 0.01). CONCLUSIONS This study provides evidence for the suitability of EP3533 in the quantification of muscular fibrosis in mdx mice and demonstrated that EP3533-derived measurements correlated strongly to ex vivo fibrosis measurement.
Collapse
Affiliation(s)
- Alexander Peter Murphy
- The John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, The International Centre for Life, Newcastle University, Central Parkway, Newcastle Upon Tyne, United Kingdom
| | - Elizabeth Greally
- The John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, The International Centre for Life, Newcastle University, Central Parkway, Newcastle Upon Tyne, United Kingdom
| | - Dara O'Hogain
- Institute of Cellular Medicine, Newcastle Magnetic Resonance Centre, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, United Kingdom
| | - Andrew Blamire
- Institute of Cellular Medicine, Newcastle Magnetic Resonance Centre, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, United Kingdom
| | - Peter Caravan
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, The International Centre for Life, Newcastle University, Central Parkway, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
29
|
Voltage-Dependent Sarcolemmal Ion Channel Abnormalities in the Dystrophin-Deficient Heart. Int J Mol Sci 2018; 19:ijms19113296. [PMID: 30360568 PMCID: PMC6274787 DOI: 10.3390/ijms19113296] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/28/2022] Open
Abstract
Mutations in the gene encoding for the intracellular protein dystrophin cause severe forms of muscular dystrophy. These so-called dystrophinopathies are characterized by skeletal muscle weakness and degeneration. Dystrophin deficiency also gives rise to considerable complications in the heart, including cardiomyopathy development and arrhythmias. The current understanding of the pathomechanisms in the dystrophic heart is limited, but there is growing evidence that dysfunctional voltage-dependent ion channels in dystrophin-deficient cardiomyocytes play a significant role. Herein, we summarize the current knowledge about abnormalities in voltage-dependent sarcolemmal ion channel properties in the dystrophic heart, and discuss the potentially underlying mechanisms, as well as their pathophysiological relevance.
Collapse
|
30
|
Mihalko E, Huang K, Sproul E, Cheng K, Brown AC. Targeted Treatment of Ischemic and Fibrotic Complications of Myocardial Infarction Using a Dual-Delivery Microgel Therapeutic. ACS NANO 2018; 12:7826-7837. [PMID: 30016078 DOI: 10.1021/acsnano.8b01977] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Myocardial infarction (MI), commonly known as a heart attack, affects millions of people worldwide and results in significant death and disabilities. A major cause of MI is fibrin-rich thrombus formation that occludes the coronary arteries, blocking blood flow to the heart and causing fibrin deposition. In treating MI, re-establishing blood flow is critical. However, ischemia reperfusion (I/R) injury itself can also occur and contributes to cardiac fibrosis. Fibrin-specific poly( N-isopropylacrylamide) nanogels (FSNs) comprised of a core-shell colloidal hydrogel architecture are utilized in this study to design a dual-delivery system that simultaneously addresses the need to (1) re-establish blood flow and (2) inhibit cardiac fibrosis following I/R injury. These therapeutic needs are met by controlling the release of a fibrinolytic protein, tissue plasminogen activator (tPA), and a small molecule cell contractility inhibitor (Y-27632). In vitro, tPA and Y-27632-loaded FSNs rapidly degrade fibrin and decrease cardiac cell stress fiber formation and connective tissue growth factor expression, which are both upregulated in cardiac fibrosis. In vivo, FSNs localize to fibrin in injured heart tissue and, when loaded with tPA and Y-27632, showed significant improvement in left ventricular ejection fraction 2 and 4 weeks post-I/R as well as significantly decreased infarct size, α-smooth muscle actin expression, and connective tissue growth factor expression 4 weeks post-I/R. Together, these data demonstrate the feasibility of this targeted therapeutic strategy to improve cardiac function following MI.
Collapse
|
31
|
Hong L, Lai H, Fang Y, Tao Y, Qiu Y. Silencing CTGF/CCN2 inactivates the MAPK signaling pathway to alleviate myocardial fibrosis and left ventricular hypertrophy in rats with dilated cardiomyopathy. J Cell Biochem 2018; 119:9519-9531. [PMID: 30129221 DOI: 10.1002/jcb.27268] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 06/26/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Lang Hong
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| | - Heng‐Li Lai
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| | - Yan Fang
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| | - Yu Tao
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| | - Yun Qiu
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| |
Collapse
|
32
|
Long-term impact of intrauterine neuroinflammation and treatment with magnesium sulphate and betamethasone: Sex-specific differences in a preterm labor murine model. Sci Rep 2017; 7:17883. [PMID: 29263436 PMCID: PMC5738437 DOI: 10.1038/s41598-017-18197-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/06/2017] [Indexed: 11/17/2022] Open
Abstract
Preterm infants are at significantly increased risk for lifelong neurodevelopmental disability with male offspring disproportionately affected. Corticosteroids (such as betamethasone) and magnesium sulphate (MgSO4) are administered to women in preterm labor to reduce neurologic morbidity. Despite widespread use of MgSO4 in clinical practice, its effects on adult offspring are not well known nor have sex-specific differences in therapeutic response been explored. The objective of our study was to examine the long-term effects of perinatal neuroinflammation and the effectiveness of prenatal MgSO4/betamethasone treatments between males and females in a murine model via histologic and expression analyses. Our results demonstrate that male but not female offspring exposed to intrauterine inflammation demonstrated impaired performance in neurodevelopmental testing in early life assessed via negative geotaxis, while those exposed to injury plus treatment fared better. Histologic analysis of adult male brains identified a significant reduction in hippocampal neural density in the injured group compared to controls. Evaluation of key neural markers via qRT-PCR demonstrated more profound differences in gene expression in adult males exposed to injury and treatment compared to female offspring, which largely showed resistance to injury. Prenatal treatment with MgSO4/betamethasone confers long-term benefits beyond cerebral palsy prevention with sex-specific differences in response.
Collapse
|
33
|
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
|
34
|
Rutkovskiy A, Sagave J, Czibik G, Baysa A, Zihlavnikova Enayati K, Hillestad V, Dahl CP, Fiane A, Gullestad L, Gravning J, Ahmed S, Attramadal H, Valen G, Vaage J. Connective tissue growth factor and bone morphogenetic protein 2 are induced following myocardial ischemia in mice and humans. Scandinavian Journal of Clinical and Laboratory Investigation 2017; 77:321-331. [PMID: 28460577 DOI: 10.1080/00365513.2017.1318447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We aimed to study the cardiac expression of bone morphogenetic protein 2, its receptor 1 b, and connective tissue growth factor, factors implicated in cardiac embryogenesis, following ischemia/hypoxia, heart failure, and in remodeling hearts from humans and mice. Biopsies from the left ventricle of patients with end-stage heart failure due to dilated cardiomyopathy or coronary artery disease were compared with donor hearts and biopsies from patients with normal heart function undergoing coronary artery bypass grafting. Mouse model of post-infarction remodeling was made by permanent ligation of the left coronary artery. Hearts were analyzed by real-time polymerase chain reaction and Western blotting after 24 hours and after 2 and 4 weeks. Patients with dilated cardiomyopathy and mice post-infarction had increased cardiac expression of connective tissue growth factor. Bone morphogenetic protein 2 was increased in human hearts failing due to coronary artery disease and in mice post-infarction. Gene expression of bone morphogenetic protein receptor 1 beta was reduced in hearts of patients with failure, but increased two weeks following permanent ligation of the left coronary artery in mice. In conclusion, connective tissue growth factor is upregulated in hearts of humans with dilated cardiomyopathy, bone morphogenetic protein 2 is upregulated in remodeling due to myocardial infarction while its receptor 1 b in human failing hearts is downregulated. A potential explanation might be an attempt to engage regenerative processes, which should be addressed by further, mechanistic studies.
Collapse
Affiliation(s)
- Arkady Rutkovskiy
- a Division of Physiology, Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway.,b International Laboratory of Bioinformatics and Genomics, ITMO University , Saint-Petersburg , Russia
| | - Julia Sagave
- a Division of Physiology, Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway
| | - Gabor Czibik
- a Division of Physiology, Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway
| | - Anton Baysa
- a Division of Physiology, Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway
| | - Katarina Zihlavnikova Enayati
- a Division of Physiology, Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway
| | - Vigdis Hillestad
- c Institute for Experimental Medical Research, Oslo University Hospital, Ullevål , Oslo , Norway
| | | | - Arnt Fiane
- e Department of Cardiothoracic Surgery , Oslo University Hospital , Oslo , Norway
| | - Lars Gullestad
- d Department of Cardiology , Oslo University Hospital , Oslo , Norway
| | - Jørgen Gravning
- f Division of Medicine , Akershus University Hospital , Lørenskog , Norway
| | - Shakil Ahmed
- c Institute for Experimental Medical Research, Oslo University Hospital, Ullevål , Oslo , Norway
| | - Håvard Attramadal
- c Institute for Experimental Medical Research, Oslo University Hospital, Ullevål , Oslo , Norway
| | - Guro Valen
- a Division of Physiology, Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway
| | - Jarle Vaage
- g Department of Emergency Medicine and Intensive Care , Oslo University Hospital , Oslo , Norway
| |
Collapse
|
35
|
Abstract
Cardiac fibrosis is a significant global health problem that is closely associated with multiple forms of cardiovascular disease, including myocardial infarction, dilated cardiomyopathy, and diabetes. Fibrosis increases myocardial wall stiffness due to excessive extracellular matrix deposition, causing impaired systolic and diastolic function, and facilitating arrhythmogenesis. As a result, patient morbidity and mortality are often dramatically elevated compared with those with cardiovascular disease but without overt fibrosis, demonstrating that fibrosis itself is both a pathologic response to existing disease and a significant risk factor for exacerbation of the underlying condition. The lack of any specific treatment for cardiac fibrosis in patients suffering from cardiovascular disease is a critical gap in our ability to care for these individuals. Here we provide an overview of the development of cardiac fibrosis, and discuss new research directions that have recently emerged and that may lead to the creation of novel treatments for patients with cardiovascular diseases. Such treatments would, ideally, complement existing therapy by specifically focusing on amelioration of fibrosis.
Collapse
Affiliation(s)
- Danah Al Hattab
- a Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada.,b Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Michael P Czubryt
- a Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada.,b Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
36
|
Randhawa PK, Jaggi AS. TRPV1 channels in cardiovascular system: A double edged sword? Int J Cardiol 2017; 228:103-113. [DOI: 10.1016/j.ijcard.2016.11.205] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 11/06/2016] [Indexed: 02/08/2023]
|
37
|
Ieronimakis N, Hays A, Prasad A, Janebodin K, Duffield JS, Reyes M. PDGFRα signalling promotes fibrogenic responses in collagen-producing cells in Duchenne muscular dystrophy. J Pathol 2016; 240:410-424. [PMID: 27569721 PMCID: PMC5113675 DOI: 10.1002/path.4801] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 08/15/2016] [Accepted: 08/24/2016] [Indexed: 12/20/2022]
Abstract
Fibrosis is a characteristic of Duchenne muscular dystrophy (DMD), yet the cellular and molecular mechanisms responsible for DMD fibrosis are poorly understood. Utilizing the Collagen1a1-GFP transgene to identify cells producing Collagen-I matrix in wild-type mice exposed to toxic injury or those mutated at the dystrophin gene locus (mdx) as a model of DMD, we studied mechanisms of skeletal muscle injury/repair and fibrosis. PDGFRα is restricted to Sca1+, CD45- mesenchymal progenitors. Fate-mapping experiments using inducible CreER/LoxP somatic recombination indicate that these progenitors expand in injury or DMD to become PDGFRα+, Col1a1-GFP+ matrix-forming fibroblasts, whereas muscle fibres do not become fibroblasts but are an important source of the PDGFRα ligand, PDGF-AA. While in toxin injury/repair of muscle PDGFRα, signalling is transiently up-regulated during the regenerative phase in the DMD model and in human DMD it is chronically overactivated. Conditional expression of the constitutively active PDGFRα D842V mutation in Collagen-I+ fibroblasts, during injury/repair, hindered the repair phase and instead promoted fibrosis. In DMD, treatment of mdx mice with crenolanib, a highly selective PDGFRα/β tyrosine kinase inhibitor, reduced fibrosis, improved muscle strength, and was associated with decreased activity of Src, a downstream effector of PDGFRα signalling. These observations are consistent with a model in which PDGFRα activation of mesenchymal progenitors normally regulates repair of the injured muscle, but in DMD persistent and excessive activation of this pathway directly drives fibrosis and hinders repair. The PDGFRα pathway is a potential new target for treatment of progressive DMD. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
MESH Headings
- Animals
- Benzimidazoles/pharmacology
- Benzimidazoles/therapeutic use
- Cells, Cultured
- Collagen Type I/biosynthesis
- Disease Models, Animal
- Dystrophin/genetics
- Enzyme Inhibitors/pharmacology
- Fibroblasts/drug effects
- Fibroblasts/pathology
- Fibrosis
- Male
- Mice, Transgenic
- Muscle Strength/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Mutation
- Piperidines/pharmacology
- Piperidines/therapeutic use
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors
- Receptor, Platelet-Derived Growth Factor alpha/genetics
- Receptor, Platelet-Derived Growth Factor alpha/physiology
- Regeneration/drug effects
- Regeneration/physiology
- Signal Transduction/drug effects
- Signal Transduction/physiology
Collapse
Affiliation(s)
| | - Aislinn Hays
- Department of PathologyAlbert Einstein College of MedicineNYUSA
| | - Amalthiya Prasad
- Department of Pathology, School of MedicineUniversity of WashingtonWAUSA
| | | | - Jeremy S Duffield
- Department of Pathology, School of MedicineUniversity of WashingtonWAUSA
- Department of Medicine, School of MedicineUniversity of WashingtonWAUSA
- Discovery ResearchBiogen IncCambridgeMAUSA
| | - Morayma Reyes
- Department of PathologyAlbert Einstein College of MedicineNYUSA
- Montefiore Medical CenterBronxNYUSA
| |
Collapse
|
38
|
La Merrill MA, Sethi S, Benard L, Moshier E, Haraldsson B, Buettner C. Perinatal DDT Exposure Induces Hypertension and Cardiac Hypertrophy in Adult Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1722-1727. [PMID: 27325568 PMCID: PMC5089878 DOI: 10.1289/ehp164] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 03/21/2016] [Accepted: 05/18/2016] [Indexed: 05/08/2023]
Abstract
BACKGROUND Dichlorodiphenyltrichloroethane (DDT) was used extensively to control malaria, typhus, body lice, and bubonic plague worldwide, until countries began restricting its use in the 1970s. However, the use of DDT to control vector-borne diseases continues in developing countries. Prenatal DDT exposure is associated with elevated blood pressure in humans. OBJECTIVE We hypothesized that perinatal DDT exposure causes hypertension in adult mice. METHODS DDT was administered to C57BL/6J dams from gestational day 11.5 to postnatal day 5. Blood pressure (BP) and myocardial wall thickness were measured in male and female adult offspring. Adult mice were treated with an angiotensin converting enzyme (ACE) inhibitor, captopril, to evaluate sensitivity to amelioration of DDT-associated hypertension by ACE inhibition. We further assessed the influence of DDT exposure on the expression of mRNAs that regulate BP through renal ion transport. RESULTS Adult mice perinatally exposed to DDT exhibited chronically increased systolic BP, increased myocardial wall thickness, and elevated expression of mRNAs of several renal ion transporters. Captopril completely reversed hypertension in mice perinatally exposed to DDT. CONCLUSIONS These data demonstrate that perinatal exposure to DDT causes hypertension and cardiac hypertrophy in adult offspring. A key mechanism underpinning this hypertension is an overactivated renin angiotensin system because ACE inhibition reverses the hypertension induced by perinatal DDT exposure. Citation: La Merrill M, Sethi S, Benard L, Moshier E, Haraldsson B, Buettner C. 2016. Perinatal DDT exposure induces hypertension and cardiac hypertrophy in adult mice. Environ Health Perspect 124:1722-1727; http://dx.doi.org/10.1289/EHP164.
Collapse
Affiliation(s)
- Michele A. La Merrill
- Department of Environmental Toxicology, University of California, Davis, Davis, CA, USA
- Department of Preventive Medicine, and
- Address correspondence to M. La Merrill, Department of Environmental Toxicology, University of California at Davis, 1 Shields Ave., 4245 Meyer Hall, Davis, CA 95616-5270 USA. Telephone: (530) 752-1142. , or C. Buettner, Department of Medicine, Mount Sinai School of Medicine, Division of Endocrinology, Metabolism Institute, One Gustave L. Levy Place, Box 1055, New York, NY 10029-6574 USA. Telephone: (212) 241-3425.
| | - Sunjay Sethi
- Department of Environmental Toxicology, University of California, Davis, Davis, CA, USA
| | - Ludovic Benard
- Center for Science and Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Borje Haraldsson
- Department of Nephrology, University of Gothenburg, Gothenburg, Sweden
| | - Christoph Buettner
- Department of Medicine,
- Department of Neuroscience, and
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Address correspondence to M. La Merrill, Department of Environmental Toxicology, University of California at Davis, 1 Shields Ave., 4245 Meyer Hall, Davis, CA 95616-5270 USA. Telephone: (530) 752-1142. , or C. Buettner, Department of Medicine, Mount Sinai School of Medicine, Division of Endocrinology, Metabolism Institute, One Gustave L. Levy Place, Box 1055, New York, NY 10029-6574 USA. Telephone: (212) 241-3425.
| |
Collapse
|
39
|
Murphy S, Dowling P, Zweyer M, Mundegar RR, Henry M, Meleady P, Swandulla D, Ohlendieck K. Proteomic analysis of dystrophin deficiency and associated changes in the aged mdx-4cv heart model of dystrophinopathy-related cardiomyopathy. J Proteomics 2016; 145:24-36. [DOI: 10.1016/j.jprot.2016.03.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/19/2016] [Accepted: 03/02/2016] [Indexed: 12/27/2022]
|
40
|
Chatzifrangkeskou M, Le Dour C, Wu W, Morrow JP, Joseph LC, Beuvin M, Sera F, Homma S, Vignier N, Mougenot N, Bonne G, Lipson KE, Worman HJ, Muchir A. ERK1/2 directly acts on CTGF/CCN2 expression to mediate myocardial fibrosis in cardiomyopathy caused by mutations in the lamin A/C gene. Hum Mol Genet 2016; 25:2220-2233. [PMID: 27131347 DOI: 10.1093/hmg/ddw090] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/11/2016] [Indexed: 12/27/2022] Open
Abstract
Cardiomyopathy caused by lamin A/C gene mutations (LMNA cardiomyopathy) is characterized by increased myocardial fibrosis, which impairs left ventricular relaxation and predisposes to heart failure, and cardiac conduction abnormalities. While we previously discovered abnormally elevated extracellular signal-regulated kinase 1/2 (ERK1/2) activities in heart in LMNA cardiomyopathy, its role on the development of myocardial fibrosis remains unclear. We now showed that transforming growth factor (TGF)-β/Smad signaling participates in the activation of ERK1/2 signaling in LMNA cardiomyopathy. ERK1/2 acts on connective tissue growth factor (CTGF/CCN2) expression to mediate the myocardial fibrosis and left ventricular dysfunction. Studies in vivo demonstrate that inhibiting CTGF/CCN2 using a specific antibody decreases myocardial fibrosis and improves the left ventricular dysfunction. Together, these findings show that cardiac ERK1/2 activity is modulated in part by TGF-β/Smad signaling, leading to altered activation of CTGF/CCN2 to mediate fibrosis and alter cardiac function. This identifies a novel mechanism in the development of LMNA cardiomyopathy.
Collapse
Affiliation(s)
- Maria Chatzifrangkeskou
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Institut de Myologie, G.H. Pitié Salpêtrière, 75651 Paris Cedex 13, France
| | - Caroline Le Dour
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Wei Wu
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - John P Morrow
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Leroy C Joseph
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Maud Beuvin
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Institut de Myologie, G.H. Pitié Salpêtrière, 75651 Paris Cedex 13, France
| | - Fusako Sera
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Shunichi Homma
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Nicolas Vignier
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Institut de Myologie, G.H. Pitié Salpêtrière, 75651 Paris Cedex 13, France
| | - Nathalie Mougenot
- Sorbonne Universités, UPMC Paris 06, INSERM UMS28 Phénotypage du petit animal, Faculté de Médecine Pierre et Marie Curie, F-75013, Paris, France
| | - Gisèle Bonne
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Institut de Myologie, G.H. Pitié Salpêtrière, 75651 Paris Cedex 13, France
| | | | - Howard J Worman
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Antoine Muchir
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Institut de Myologie, G.H. Pitié Salpêtrière, 75651 Paris Cedex 13, France Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
41
|
Lv S, Wu M, Li M, Wang Q, Xu L, Wang X, Zhang J. Effect and Mechanism of QiShenYiQi Pill on Experimental Autoimmune Myocarditis Rats. Med Sci Monit 2016; 22:752-6. [PMID: 26946470 PMCID: PMC4784548 DOI: 10.12659/msm.895655] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background To observe the effect of QiShenYiQi pill (QSYQ) on experimental autoimmune myocarditis rats, and to explore its mechanism of action. Material/methods Lewis rats underwent the injection of myocardial myosin mixed with Freund’s complete adjuvant were randomized into 3 groups: model, valsartan, and QSYQ groups. Rats injected with phosphate-buffered saline (PBS) mixed with Freund’s complete adjuvant were used as the control group. Rats were euthanized at 4 and 8 weeks, and we weighed rat body mass, heart mass, and left ventricular mass. Myocardium sections were stained with hematoxylin and eosin (H&E) and Masson trichrome. Myocardial TGF-β1 and CTGF protein expression was detected by immunohistochemistry, and myocardial TGF-β1 and CTGF mRNA expression was detected by real-time qPCR. Results QSYQ reduced HMI and LVMI, as well as the histological score of hearts and CVF, which further decreased over time, and its effect was significantly greater than that of valsartan at 4 and 8 weeks. After 4 weeks, QSYQ inhibited the protein and mRNA expression of TGF-β1 and CTGF, and its effect on lowering CTGF was significantly greater than that of valsartan. In addition, after 8 weeks, QSYQ also inhibited the protein and mRNA expression of CTGF, whereas there was no significant difference in the expression of myocardial TGF-β1. Conclusions This study provides evidence that QSYQ can improve cardiac remodeling of experimental autoimmune myocarditis rats. It also effectively improved the degree of myocardial fibrosis, which is related to the mechanism of regulation of TGF-β1 CTGF.
Collapse
Affiliation(s)
- Shichao Lv
- Department of Geriatric Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China (mainland)
| | - Meifang Wu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China (mainland)
| | - Meng Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China (mainland)
| | - Qiang Wang
- Graduate School, Tianjin University of Traditional Chinese Medcine, Tianjin, China (mainland)
| | - Ling Xu
- Graduate School, Tianjin University of Traditional Chinese Medcine, Tianjin, China (mainland)
| | - Xiaojing Wang
- Graduate School, Tianjin University of Traditional Chinese Medcine, Tianjin, China (mainland)
| | - Junping Zhang
- Department of Geriatric Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medcine, Tianjin, China (mainland)
| |
Collapse
|
42
|
The Effects of Experimental Sleep Apnea on Cardiac and Respiratory Functions in 6 and 18 Month Old Dystrophic (mdx) Mice. PLoS One 2016; 11:e0147640. [PMID: 26808526 PMCID: PMC4726600 DOI: 10.1371/journal.pone.0147640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/05/2016] [Indexed: 11/19/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal disease where over 90% of patients succumb to respiratory or cardiac failure. Sleep apnea and sleep disordered breathing (SDB) are noted in a plurality of DMD patients, and the resulting nocturnal episodic hypoxia (EH) cannot be ruled out as a contributing factor to cardiac and respiratory dysfunction. In this study, we investigated the impact of long-term episodic hypoxia, which mimics the cyclic hypoxia seen in sleep apnea, on cardiac and respiratory function in a murine model of DMD (mdx mice). Since the severity and prevalence of sleep apnea in DMD increases with age, we studied the impact of EH on young (6-month) and on older (18-month) mdx mice. Mice were either exposed for 12 weeks to EH (8 hours/day, 5 days/week) or to room air. We noted a significant increase in left ventricular (LV) dilatation (transthoracic echocardiography) on EH exposure in both age groups, but reduced LV contractility was seen only in 6-month old mice. With EH exposure, an increased fibrosis (hydroxyproline) was noted in both cardiac and diaphragm muscle in 18-month but not 6-month old mice. No significant change in relative diaphragm strength (in-vitro) was noted on EH exposure in 18-month old mice. In contrast, EH exposed 6-month old mice showed a significant increase in relative diaphragm strength. EH exposure did not result in any significant change in ventilatory parameters (barometric plethysmography) in awake 6-month old mdx mice. In contrast, 18-month old mdx mice showed considerable ventilatory dysfunction, consistent with reduced ventilatory reserve. Our findings highlight that sleep apnea impacts respiratory and cardiac function in muscular dystrophy, and that EH can have divergent effects on both systems. To our knowledge, this is the first comprehensive study to investigate the impact of EH on cardiac and respiratory function in mdx mice.
Collapse
|
43
|
Koshman YE, Sternlicht MD, Kim T, O'Hara CP, Koczor CA, Lewis W, Seeley TW, Lipson KE, Samarel AM. Connective tissue growth factor regulates cardiac function and tissue remodeling in a mouse model of dilated cardiomyopathy. J Mol Cell Cardiol 2015; 89:214-22. [PMID: 26549358 DOI: 10.1016/j.yjmcc.2015.11.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/20/2015] [Accepted: 11/02/2015] [Indexed: 12/14/2022]
Abstract
Cardiac structural changes associated with dilated cardiomyopathy (DCM) include cardiomyocyte hypertrophy and myocardial fibrosis. Connective tissue growth factor (CTGF) has been associated with tissue remodeling and is highly expressed in failing hearts. Our aim was to test if inhibition of CTGF would alter the course of cardiac remodeling and preserve cardiac function in the protein kinase Cε (PKCε) mouse model of DCM. Transgenic mice expressing constitutively active PKCε in cardiomyocytes develop cardiac dysfunction that was evident by 3 months of age, and that progressed to cardiac fibrosis, heart failure, and increased mortality. Beginning at 3 months of age, PKCε mice were treated with a neutralizing monoclonal antibody to CTGF (FG-3149) for an additional 3 months. CTGF inhibition significantly improved left ventricular (LV) systolic and diastolic functions in PKCε mice, and slowed the progression of LV dilatation. Using gene arrays and quantitative PCR, the expression of many genes associated with tissue remodeling was elevated in PKCε mice, but significantly decreased by CTGF inhibition. However total collagen deposition was not attenuated. The observation of significantly improved LV function by CTGF inhibition in PKCε mice suggests that CTGF inhibition may benefit patients with DCM. Additional studies to explore this potential are warranted.
Collapse
Affiliation(s)
- Yevgeniya E Koshman
- The Cardiovascular Research Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, United States
| | | | - Taehoon Kim
- The Cardiovascular Research Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, United States
| | - Christopher P O'Hara
- The Cardiovascular Research Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, United States
| | - Christopher A Koczor
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - William Lewis
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Todd W Seeley
- FibroGen, Inc., San Francisco, CA 94158, United States
| | | | - Allen M Samarel
- The Cardiovascular Research Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, United States.
| |
Collapse
|
44
|
Lee Y, Kwak HB, Hord J, Kim JH, Lawler JM. Exercise training attenuates age-dependent elevation of angiotensin II type 1 receptor and Nox2 signaling in the rat heart. Exp Gerontol 2015; 70:163-73. [DOI: 10.1016/j.exger.2015.07.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 07/26/2015] [Accepted: 07/30/2015] [Indexed: 11/25/2022]
|
45
|
Bozzi M, Sciandra F, Brancaccio A. Role of gelatinases in pathological and physiological processes involving the dystrophin–glycoprotein complex. Matrix Biol 2015; 44-46:130-7. [DOI: 10.1016/j.matbio.2015.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 02/09/2015] [Accepted: 02/10/2015] [Indexed: 12/16/2022]
|
46
|
Vianello S, Bouyon S, Benoit E, Sebrié C, Boerio D, Herbin M, Roulot M, Fromes Y, de la Porte S. Arginine butyrate per os protects mdx mice against cardiomyopathy, kyphosis and changes in axonal excitability. Neurobiol Dis 2014; 71:325-33. [PMID: 25167832 DOI: 10.1016/j.nbd.2014.08.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 08/08/2014] [Accepted: 08/16/2014] [Indexed: 11/30/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive neuromuscular disease caused by lack of dystrophin, a sub-sarcolemmal protein, which leads to dramatic muscle deterioration. We studied in mdx mice, the effects of oral administration of arginine butyrate (AB), a compound currently used for the treatment of sickle cell anemia in children, on cardiomyopathy, vertebral column deformation and electromyographic abnormalities. Monthly follow-up by echocardiography from the 8th month to the 14th month showed that AB treatment protected the mdx mice against drastic reduction (20-23%) of ejection fraction and fractional shortening, and also against the ≈20% ventricular dilatation and 25% cardiac hypertrophy observed in saline-treated mdx mice. The phenotypic improvement was corroborated by the decrease in serum CK level and by better fatigue resistance. Moreover, AB treatment protected against the progressive spinal deformity observed in mdx mice, another similarity with DMD patients. The value of the kyphosis index in AB-treated mice reached 94% of the value in C57BL/10 mice. Finally, axonal excitability parameters such as the membrane resting potential, the threshold and amplitude of the action potential, the absolute and relative refractory periods and the supernormal and subnormal periods, recorded from caudal and plantar muscles in response to excitability tests, that were modified in saline-treated mdx mice were not significantly changed, compared with wild-type animals, in AB-treated mdx mice. All of these results suggest that AB could be a potential treatment for DMD patients.
Collapse
Affiliation(s)
- Sara Vianello
- CNRS, Institut de Neurobiologie Alfred Fessard, FRC2118, Neurobiologie & Développement, UPR 3294, Gif sur Yvette, F-91198, France.
| | - Sophie Bouyon
- UPMC, Université Paris 6, UMR 974, Institut de Myologie, F-75013 Paris, France.
| | - Evelyne Benoit
- CNRS, Institut de Neurobiologie Alfred Fessard, FRC2118, Neurobiologie & Développement, UPR 3294, Gif sur Yvette, F-91198, France.
| | | | - Delphine Boerio
- CNRS, Institut de Neurobiologie Alfred Fessard, FRC2118, Neurobiologie & Développement, UPR 3294, Gif sur Yvette, F-91198, France.
| | - Marc Herbin
- CNRS, Muséum National d'Histoire Naturelle, CNRS, UMR7179, Pavillon d'anatomie comparée, BP 55, 52 Rue Cuvier, 75231 Paris Cedex 05, France.
| | - Morgane Roulot
- CNRS, Institut de Neurobiologie Alfred Fessard, FRC2118, Neurobiologie & Développement, UPR 3294, Gif sur Yvette, F-91198, France.
| | - Yves Fromes
- UPMC, Université Paris 6, UMR 974, Institut de Myologie, F-75013 Paris, France; ONIRIS, Centre de Boisbonne, Nantes F-44307, France.
| | - Sabine de la Porte
- CNRS, Institut de Neurobiologie Alfred Fessard, FRC2118, Neurobiologie & Développement, UPR 3294, Gif sur Yvette, F-91198, France.
| |
Collapse
|
47
|
Proteomic profiling of the dystrophin-deficient mdx phenocopy of dystrophinopathy-associated cardiomyopathy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:246195. [PMID: 24772416 PMCID: PMC3977469 DOI: 10.1155/2014/246195] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/16/2014] [Indexed: 01/07/2023]
Abstract
Cardiorespiratory complications are frequent symptoms of Duchenne muscular dystrophy, a neuromuscular disorder caused by primary abnormalities in the dystrophin gene. Loss of cardiac dystrophin initially leads to changes in dystrophin-associated glycoproteins and subsequently triggers secondarily sarcolemmal disintegration, fibre necrosis, fibrosis, fatty tissue replacement, and interstitial inflammation. This results in progressive cardiac disease, which is the cause of death in a considerable number of patients afflicted with X-linked muscular dystrophy. In order to better define the molecular pathogenesis of this type of cardiomyopathy, several studies have applied mass spectrometry-based proteomics to determine proteome-wide alterations in dystrophinopathy-associated cardiomyopathy. Proteomic studies included both gel-based and label-free mass spectrometric surveys of dystrophin-deficient heart muscle from the established mdx animal model of dystrophinopathy. Comparative cardiac proteomics revealed novel changes in proteins associated with mitochondrial energy metabolism, glycolysis, signaling, iron binding, antibody response, fibre contraction, basal lamina stabilisation, and cytoskeletal organisation. This review summarizes the importance of studying cardiomyopathy within the field of muscular dystrophy research, outlines key features of the mdx heart and its suitability as a model system for studying cardiac pathogenesis, and discusses the impact of recent proteomic findings for exploring molecular and cellular aspects of cardiac abnormalities in inherited muscular dystrophies.
Collapse
|
48
|
Weng CM, Yu CC, Kuo ML, Chen BC, Lin CH. Endothelin-1 induces connective tissue growth factor expression in human lung fibroblasts by ETAR-dependent JNK/AP-1 pathway. Biochem Pharmacol 2014; 88:402-11. [PMID: 24486572 DOI: 10.1016/j.bcp.2014.01.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 01/18/2014] [Accepted: 01/22/2014] [Indexed: 01/22/2023]
Abstract
Endothelin-1 (ET-1) acts as a key mediator of vasoconstriction and tissue repair. Overproduction of connective tissue growth factor (CTGF) underlies the development of lung fibrosis. ET-1 induces expression of matrix-associated genes in lung fibroblasts, however, little is known about the signaling pathway of CTGF expression caused by ET-1. In this study, we found that ET-1 caused concentration- and time-dependently increases in CTGF expression in human embryonic lung fibroblast cell line (WI-38). ET-1-induced CTGF expression was inhibited by BQ123 (ETAR antagonist), but not BQ788 (ETBR antagonist). Moreover, ET-1-induced CTGF expression was significantly reduced by JNK inhibitor (SP600125), the dominant-negative mutants of JNK1/2 (JNK1/2 DN), and AP-1 inhibitor (curcumin). ET-1 induced phosphorylations of JNK and c-Jun in time-dependent manners. AP-1 luciferase activity was concentration-dependently increased by ET-1, and this effect was attenuated by SP600125. We also found that ET-1-induced CTGF expression was most controlled by the AP-1 binding region of CTGF promoter. ET-1-indiced CTGF luciferase activity was predominately controlled by the sequence -747 to -408 bp upstream of the transcription start site on the human CTGF promoter. Furthermore, ET-1 caused the formation of AP-1-specific DNA-protein complex and the recruitment of c-Jun to the CTGF promoter. Moreover, we found that ET-1 induced α-smooth muscle actin (α-SMA) expression, which was inhibited by BQ123, SP600125, curcumin, and anti-CTGF antibody. These results suggest that ET-1 stimulates expressions of CTGF and α-SMA through ETAR/JNK/AP-1 signaling pathway, and CTGF is required for ET-1-induced α-SMA expression in human lung fibroblasts.
Collapse
Affiliation(s)
- Chih-Ming Weng
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chung-Chi Yu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Min-Liang Kuo
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Graduate Institute of Biomedical Sciences, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Bing-Chang Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei 11031, Taiwan.
| | - Chien-Huang Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
49
|
Low dystrophin levels in heart can delay heart failure in mdx mice. J Mol Cell Cardiol 2014; 69:17-23. [PMID: 24486194 DOI: 10.1016/j.yjmcc.2014.01.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 12/25/2013] [Accepted: 01/21/2014] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy is caused by mutations that prevent synthesis of functional dystrophin. All patients develop dilated cardiomyopathy. Promising therapeutic approaches are underway that successfully restore dystrophin expression in skeletal muscle. However, their efficiency in the heart is limited. Improved quality and function of only skeletal muscle potentially accelerate the development of cardiomyopathy. Our study aimed to elucidate which dystrophin levels in the heart are required to prevent or delay cardiomyopathy in mice. Heart function and pathology assessed with magnetic resonance imaging and histopathological analysis were compared between 2, 6 and 10-month-old female mdx-Xist(Δhs) mice, expressing low dystrophin levels (3-15%) in a mosaic manner based on skewed X-inactivation, dystrophin-negative mdx mice, and wild type mice of corresponding genetic backgrounds and gender. With age mdx mice developed dilated cardiomyopathy and hypertrophy, whereas the onset of heart pathology was delayed and function improved in mdx-Xist(Δhs) mice. The ejection fraction, the most severely affected parameter for both ventricles, correlated to dystrophin expression and the percentage of fibrosis. Fibrosis was partly reduced from 9.8% in mdx to 5.4% in 10 month old mdx-Xist(Δhs) mice. These data suggest that mosaic expression of 4-15% dystrophin in the heart is sufficient to delay the onset and ameliorate cardiomyopathy in mice.
Collapse
|
50
|
SONG ZHIPING, LIU XU, ZHANG DADONG. Connective Tissue Growth Factor: A Predictor of Recurrence after Catheter Ablation in Patients with Nonparoxysmal Atrial Fibrillation. PACING AND CLINICAL ELECTROPHYSIOLOGY: PACE 2014; 37:630-7. [PMID: 24467520 DOI: 10.1111/pace.12345] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 12/06/2013] [Accepted: 12/09/2013] [Indexed: 01/22/2023]
Affiliation(s)
- ZHI-PING SONG
- Department of Cardiology; Minhang Hospital; Ruijin Hospital Group; Shanghai Jiaotong University School of Medicine; Shanghai People's Republic of China
| | - XU LIU
- Department of Cardiology; Shanghai Chest Hospital; Shanghai Jiaotong University School of Medicine; Shanghai People's Republic of China
| | - DA-DONG ZHANG
- Department of Cardiology; Minhang Hospital; Ruijin Hospital Group; Shanghai Jiaotong University School of Medicine; Shanghai People's Republic of China
| |
Collapse
|