1
|
Gao X, Cao Y, Li H, Yu F, Xi J, Zhang J, Zhuang R, Xu Y, Xu L. Mechanisms underlying altered ubiquitin-proteasome system activity during heart failure and pharmacological interventions. Eur J Med Chem 2025; 292:117725. [PMID: 40334506 DOI: 10.1016/j.ejmech.2025.117725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/28/2025] [Accepted: 05/04/2025] [Indexed: 05/09/2025]
Abstract
Heart failure (HF) is a refractory disease with a global prevalence that is continuously increasing. The mechanisms underlying the pathogenesis of HF are multi-faceted, intricate, and not yet fully elucidated. Appropriate levels of protein turnover are essential for maintaining cardiac homeostasis and, accordingly, compromised protein degradation systems can significantly contribute to heart disease. The ubiquitin-proteasome system (UPS) modulates the structure and function of cardiac cells by facilitating the degradation of signaling and structural proteins. Research in the preceding decade has focused on elucidating the role of the UPS in the context of cardiovascular physiology and pathophysiology. A comprehensive understanding of the UPS status and the underlying mechanisms contributing to its potential dysregulation in HF is imperative for developing targeted therapeutic interventions. Previous research has identified several novel interventions involving components of the UPS and several have been adapted for HF therapy. In this review, we summarize the mechanisms underlying altered UPS activity in HF and provide an outline of UPS regulators that affect the progression of HF. Additionally, the potential for small molecules to intervene in UPS function in HF is discussed.
Collapse
Affiliation(s)
- Xiaofei Gao
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 310006, Zhejiang, China
| | - Yu Cao
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China
| | - Hangyan Li
- Department of Cardiology, The Third People's Hospital of Yuhang District, Hangzhou, 311115, Zhejiang, China
| | - Faming Yu
- Department of Cardiology, The Third People's Hospital of Yuhang District, Hangzhou, 311115, Zhejiang, China
| | - Jianjun Xi
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China
| | - Jiankang Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Rangxiao Zhuang
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China.
| | - Yizhou Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 310006, Zhejiang, China.
| | - Linhao Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 310006, Zhejiang, China.
| |
Collapse
|
2
|
Barriault A, Iftikhar U, Stone JA. Cardiac Rehabilitation and Heart Failure with Reduced Ejection Fraction: Pathophysiology, Benefits, and Precautions. Can J Cardiol 2025; 41:443-455. [PMID: 39433254 DOI: 10.1016/j.cjca.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/23/2024] Open
Abstract
Heart failure (HF) is a highly comorbid condition associated with significant mortality, despite advances in current medical management. Patients who suffer from HF represent a high needs disease care population in whom structured, long-term chronic disease care delivery models, such as cardiac rehabilitation (CR), have been shown to be highly cost effective in reducing hospitalizations and improving quality of life. HF with reduced ejection fraction affects a growing number of Canadians and health care costs secondary to this condition are increasing, with further increases over the next decade to be expected. CR is a guideline-directed medical therapy for patients living with HF with reduced ejection fraction, and with increasing numbers of HF patients across the world, there is a prescient need to revisit the benefits, safety, and the prescription of this intervention for the health care professionals who treat this condition. Certainly, there is a clinical need for HF practitioners to better understand the pathophysiological benefits of CR with respect to exercise training, as well as the prudent precautions required to facilitate the safe delivery of this highly cost-effective patient intervention.
Collapse
Affiliation(s)
- Alexandra Barriault
- Total Cardiology, Calgary, Alberta, Canada; Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Umair Iftikhar
- Total Cardiology, Calgary, Alberta, Canada; Department of Cardiac Sciences, Division of Cardiology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - James A Stone
- Department of Cardiac Sciences, Division of Cardiology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Symphony of Health Connections, Calgary Alberta, Canada
| |
Collapse
|
3
|
Gouveia M, Schmidt C, Basilio PG, Aveiro SS, Domingues P, Xia K, Colón W, Vitorino R, Ferreira R, Santos M, Vieira SI, Ribeiro F. Exercise training decreases the load and changes the content of circulating SDS-resistant protein aggregates in patients with heart failure with reduced ejection fraction. Mol Cell Biochem 2024; 479:2711-2722. [PMID: 37902886 PMCID: PMC11455743 DOI: 10.1007/s11010-023-04884-z] [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: 07/25/2023] [Accepted: 10/15/2023] [Indexed: 11/01/2023]
Abstract
BACKGROUND Heart failure (HF) often disrupts the protein quality control (PQC) system leading to protein aggregate accumulation. Evidence from tissue biopsies showed that exercise restores PQC system in HF; however, little is known about its effects on plasma proteostasis. AIM To determine the effects of exercise training on the load and composition of plasma SDS-resistant protein aggregates (SRA) in patients with HF with reduced ejection fraction (HFrEF). METHODS Eighteen patients with HFrEF (age: 63.4 ± 6.5 years; LVEF: 33.4 ± 11.6%) participated in a 12-week combined (aerobic plus resistance) exercise program (60 min/session, twice per week). The load and content of circulating SRA were assessed using D2D SDS-PAGE and mass spectrometry. Cardiorespiratory fitness, quality of life, and circulating levels of high-sensitive C-reactive protein, N-terminal pro-B-type natriuretic peptide (NT-proBNP), haptoglobin and ficolin-3, were also evaluated at baseline and after the exercise program. RESULTS The exercise program decreased the plasma SRA load (% SRA/total protein: 38.0 ± 8.9 to 36.1 ± 9.7%, p = 0.018; % SRA/soluble fraction: 64.3 ± 27.1 to 59.8 ± 27.7%, p = 0.003). Plasma SRA of HFrEF patients comprised 31 proteins, with α-2-macroglobulin and haptoglobin as the most abundant ones. The exercise training significantly increased haptoglobin plasma levels (1.03 ± 0.40 to 1.11 ± 0.46, p = 0.031), while decreasing its abundance in SRA (1.83 ± 0.54 × 1011 to 1.51 ± 0.59 × 1011, p = 0.049). Cardiorespiratory fitness [16.4(5.9) to 19.0(5.2) ml/kg/min, p = 0.002], quality of life, and circulating NT-proBNP [720.0(850.0) to 587.0(847.3) pg/mL, p = 0.048] levels, also improved after the exercise program. CONCLUSION Exercise training reduced the plasma SRA load and enhanced PQC, potentially via haptoglobin-mediated action, while improving cardiorespiratory fitness and quality of life of patients with HFrEF.
Collapse
Affiliation(s)
- Marisol Gouveia
- Department of Medical Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Building 30, Agras do Crasto - Campus Universitário de Santiago, Aveiro, 3810-193, Portugal.
| | - Cristine Schmidt
- Surgery and Physiology Department, Faculty of Medicine, University of Porto, Porto, Portugal
- Research Centre in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
| | - Priscilla Gois Basilio
- Research Centre in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, Porto, Portugal
| | - Susana S Aveiro
- Mass Spectrometry Centre, Department of Chemistry, LAQV REQUIMTE, University of Aveiro, Aveiro, Portugal
- GreenCoLab - Green Ocean Association, University of Algarve, Faro, Portugal
| | - Pedro Domingues
- Mass Spectrometry Centre, Department of Chemistry, LAQV REQUIMTE, University of Aveiro, Aveiro, Portugal
| | - Ke Xia
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, NY, USA
- Centre for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Wilfredo Colón
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, NY, USA
- Centre for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Rui Vitorino
- Department of Medical Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Building 30, Agras do Crasto - Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
- Surgery and Physiology Department, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rita Ferreira
- Department of Chemistry, QOPNA & LAQV-REQUIMTE, University of Aveiro, Aveiro, Portugal
| | - Mário Santos
- Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
- Serviço de Cardiologia, Hospital Santo António, Centro Hospitalar Universitário do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, UMIB, University of Porto, Porto, Portugal
| | - Sandra I Vieira
- Department of Medical Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Building 30, Agras do Crasto - Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Fernando Ribeiro
- School of Health Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
4
|
Wernhart S, Rassaf T. Relevance of Cardiovascular Exercise in Cancer and Cancer Therapy-Related Cardiac Dysfunction. Curr Heart Fail Rep 2024; 21:238-251. [PMID: 38696059 PMCID: PMC11090948 DOI: 10.1007/s11897-024-00662-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/03/2024] [Indexed: 05/14/2024]
Abstract
PURPOSE OF THE REVIEW Cancer therapy-related cardiac dysfunction (CTRCD) has been identified as a threat to overall and cancer-related survival. Although aerobic exercise training (AET) has been shown to improve cardiorespiratory fitness (CRF), the relationship between specific exercise regimens and cancer survival, heart failure development, and reduction of CTRCD is unclear. In this review, we discuss the impact of AET on molecular pathways and the current literature of sports in the field of cardio-oncology. RECENT FINDINGS Cardio-oncological exercise trials have focused on variations of AET intensity by using moderate continuous and high intensity interval training, which are applicable, safe, and effective approaches to improve CRF. AET increases CRF, reduces cardiovascular morbidity and heart failure hospitalization and should thus be implemented as an adjunct to standard cancer therapy, although its long-term effect on CTRCD remains unknown. Despite modulating diverse molecular pathways, it remains unknown which exercise regimen, including variations of AET duration and frequency, is most suited to facilitate peripheral and central adaptations to exercise and improve survival in cancer patients.
Collapse
Affiliation(s)
- Simon Wernhart
- Department of Cardiology and Vascular Medicine, West German Heart- and Vascular Center, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart- and Vascular Center, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| |
Collapse
|
5
|
Wood N, Straw S, Cheng CW, Hirata Y, Pereira MG, Gallagher H, Egginton S, Ogawa W, Wheatcroft SB, Witte KK, Roberts LD, Bowen TS. Sodium-glucose cotransporter 2 inhibitors influence skeletal muscle pathology in patients with heart failure and reduced ejection fraction. Eur J Heart Fail 2024; 26:925-935. [PMID: 38468429 DOI: 10.1002/ejhf.3192] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/12/2024] [Accepted: 02/23/2024] [Indexed: 03/13/2024] Open
Abstract
AIMS Patients with heart failure and reduced ejection fraction (HFrEF) exhibit skeletal muscle pathology, which contributes to symptoms and decreased quality of life. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) improve clinical outcomes in HFrEF but their mechanism of action remains poorly understood. We aimed, therefore, to determine whether SGLT2i influence skeletal muscle pathology in patients with HFrEF. METHODS AND RESULTS Muscle biopsies from 28 male patients with HFrEF (New York Heart association class I-III) treated with SGLT2i (>12 months) or without SGLT2i were compared. Comprehensive analyses of muscle structure (immunohistochemistry), transcriptome (RNA sequencing), and metabolome (liquid chromatography-mass spectrometry) were performed, and serum inflammatory profiling (ELISA). Experiments in mice (n = 16) treated with SGLT2i were also performed. Myofiber atrophy was ~20% less in patients taking SGLT2i (p = 0.07). Transcriptomics and follow-up measures identified a unique signature in patients taking SGLT2i related to beneficial effects on atrophy, metabolism, and inflammation. Metabolomics identified influenced tryptophan metabolism in patients taking SGLT2i: kynurenic acid was 24% higher and kynurenine was 32% lower (p < 0.001). Serum profiling identified that SGLT2i treatment was associated with lower (p < 0.05) pro-inflammatory cytokines by 26-64% alongside downstream muscle interleukin (IL)-6-JAK/STAT3 signalling (p = 008 and 0.09). Serum IL-6 and muscle kynurenine were correlated (R = 0.65; p < 0.05). Muscle pathology was lower in mice treated with SGLT2i indicative of a conserved mammalian response to treatment. CONCLUSIONS Treatment with SGLT2i influenced skeletal muscle pathology in patients with HFrEF and was associated with anti-atrophic, anti-inflammatory, and pro-metabolic effects. These changes may be regulated via IL-6-kynurenine signalling. Together, clinical improvements following SGLT2i treatment in patients with HFrEF may be partly explained by their positive effects on skeletal muscle pathology.
Collapse
Affiliation(s)
- Nathanael Wood
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Sam Straw
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Chew W Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Yu Hirata
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Marcelo G Pereira
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Harrison Gallagher
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Stuart Egginton
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Klaus K Witte
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Clinic for Cardiology, Angiology and Internal Intensive Care Medicine, RWTH Aachen University, Aachen, Germany
| | - Lee D Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - T Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
6
|
Rutledge CA. Molecular mechanisms underlying sarcopenia in heart failure. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:7. [PMID: 38455513 PMCID: PMC10919908 DOI: 10.20517/jca.2023.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
The loss of skeletal muscle, also known as sarcopenia, is an aging-associated muscle disorder that is disproportionately present in heart failure (HF) patients. HF patients with sarcopenia have poor outcomes compared to the overall HF patient population. The prevalence of sarcopenia in HF is only expected to grow as the global population ages, and novel treatment strategies are needed to improve outcomes in this cohort. Multiple mechanistic pathways have emerged that may explain the increased prevalence of sarcopenia in the HF population, and a better understanding of these pathways may lead to the development of therapies to prevent muscle loss. This review article aims to explore the molecular mechanisms linking sarcopenia and HF, and to discuss treatment strategies aimed at addressing such molecular signals.
Collapse
Affiliation(s)
- Cody A. Rutledge
- Acute Medicine Section, Division of Medicine, Louis Stokes Cleveland Veteran Affairs Medical Center, Cleveland, OH 44106, USA
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
7
|
Alves PKN, Schauer A, Augstein A, Männel A, Barthel P, Joachim D, Friedrich J, Prieto ME, Moriscot AS, Linke A, Adams V. Leucine Supplementation Improves Diastolic Function in HFpEF by HDAC4 Inhibition. Cells 2023; 12:2561. [PMID: 37947639 PMCID: PMC10648219 DOI: 10.3390/cells12212561] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex syndrome associated with a high morbidity and mortality rate. Leucine supplementation has been demonstrated to attenuate cardiac dysfunction in animal models of cachexia and heart failure with reduced ejection fraction (HFrEF). So far, no data exist on leucine supplementation on cardiac function in HFpEF. Thus, the current study aimed to investigate the effect of leucine supplementation on myocardial function and key signaling pathways in an established HFpEF rat model. Female ZSF1 rats were randomized into three groups: Control (untreated lean rats), HFpEF (untreated obese rats), and HFpEF_Leu (obese rats receiving standard chow enriched with 3% leucine). Leucine supplementation started at 20 weeks of age after an established HFpEF was confirmed in obese rats. In all animals, cardiac function was assessed by echocardiography at baseline and throughout the experiment. At the age of 32 weeks, hemodynamics were measured invasively, and myocardial tissue was collected for assessment of mitochondrial function and for histological and molecular analyses. Leucine had already improved diastolic function after 4 weeks of treatment. This was accompanied by improved hemodynamics and reduced stiffness, as well as by reduced left ventricular fibrosis and hypertrophy. Cardiac mitochondrial respiratory function was improved by leucine without alteration of the cardiac mitochondrial content. Lastly, leucine supplementation suppressed the expression and nuclear localization of HDAC4 and was associated with Protein kinase A activation. Our data show that leucine supplementation improves diastolic function and decreases remodeling processes in a rat model of HFpEF. Beneficial effects were associated with HDAC4/TGF-β1/Collagenase downregulation and indicate a potential use in the treatment of HFpEF.
Collapse
Affiliation(s)
- Paula Ketilly Nascimento Alves
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508000, Brazil;
| | - Antje Schauer
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Antje Augstein
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Anita Männel
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Peggy Barthel
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Dirk Joachim
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Janet Friedrich
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Maria-Elisa Prieto
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Anselmo Sigari Moriscot
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508000, Brazil;
| | - Axel Linke
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Volker Adams
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| |
Collapse
|
8
|
Mangner N, Winzer EB, Linke A, Adams V. Locomotor and respiratory muscle abnormalities in HFrEF and HFpEF. Front Cardiovasc Med 2023; 10:1149065. [PMID: 37965088 PMCID: PMC10641491 DOI: 10.3389/fcvm.2023.1149065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 10/02/2023] [Indexed: 11/16/2023] Open
Abstract
Heart failure (HF) is a chronic and progressive syndrome affecting worldwide billions of patients. Exercise intolerance and early fatigue are hallmarks of HF patients either with a reduced (HFrEF) or a preserved (HFpEF) ejection fraction. Alterations of the skeletal muscle contribute to exercise intolerance in HF. This review will provide a contemporary summary of the clinical and molecular alterations currently known to occur in the skeletal muscles of both HFrEF and HFpEF, and thereby differentiate the effects on locomotor and respiratory muscles, in particular the diaphragm. Moreover, current and future therapeutic options to address skeletal muscle weakness will be discussed focusing mainly on the effects of exercise training.
Collapse
Affiliation(s)
- Norman Mangner
- Department of Internal Medicine and Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
| | - Ephraim B. Winzer
- Department of Internal Medicine and Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
| | - Axel Linke
- Department of Internal Medicine and Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
| | - Volker Adams
- Laboratory of Molecular and Experimental Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany
| |
Collapse
|
9
|
Kubat GB, Bouhamida E, Ulger O, Turkel I, Pedriali G, Ramaccini D, Ekinci O, Ozerklig B, Atalay O, Patergnani S, Nur Sahin B, Morciano G, Tuncer M, Tremoli E, Pinton P. Mitochondrial dysfunction and skeletal muscle atrophy: Causes, mechanisms, and treatment strategies. Mitochondrion 2023; 72:33-58. [PMID: 37451353 DOI: 10.1016/j.mito.2023.07.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Skeletal muscle, which accounts for approximately 40% of total body weight, is one of the most dynamic and plastic tissues in the human body and plays a vital role in movement, posture and force production. More than just a component of the locomotor system, skeletal muscle functions as an endocrine organ capable of producing and secreting hundreds of bioactive molecules. Therefore, maintaining healthy skeletal muscles is crucial for supporting overall body health. Various pathological conditions, such as prolonged immobilization, cachexia, aging, drug-induced toxicity, and cardiovascular diseases (CVDs), can disrupt the balance between muscle protein synthesis and degradation, leading to skeletal muscle atrophy. Mitochondrial dysfunction is a major contributing mechanism to skeletal muscle atrophy, as it plays crucial roles in various biological processes, including energy production, metabolic flexibility, maintenance of redox homeostasis, and regulation of apoptosis. In this review, we critically examine recent knowledge regarding the causes of muscle atrophy (disuse, cachexia, aging, etc.) and its contribution to CVDs. Additionally, we highlight the mitochondrial signaling pathways involvement to skeletal muscle atrophy, such as the ubiquitin-proteasome system, autophagy and mitophagy, mitochondrial fission-fusion, and mitochondrial biogenesis. Furthermore, we discuss current strategies, including exercise, mitochondria-targeted antioxidants, in vivo transfection of PGC-1α, and the potential use of mitochondrial transplantation as a possible therapeutic approach.
Collapse
Affiliation(s)
- Gokhan Burcin Kubat
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey.
| | - Esmaa Bouhamida
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Gaia Pedriali
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Daniela Ramaccini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Ozgur Ekinci
- Department of Pathology, Gazi University, 06500 Ankara, Turkey
| | - Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Ozbeyen Atalay
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Simone Patergnani
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Beyza Nur Sahin
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Giampaolo Morciano
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Elena Tremoli
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Paolo Pinton
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
10
|
Gallagher H, Hendrickse PW, Pereira MG, Bowen TS. Skeletal muscle atrophy, regeneration, and dysfunction in heart failure: Impact of exercise training. JOURNAL OF SPORT AND HEALTH SCIENCE 2023; 12:557-567. [PMID: 37040849 PMCID: PMC10466197 DOI: 10.1016/j.jshs.2023.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/30/2022] [Accepted: 02/20/2023] [Indexed: 05/31/2023]
Abstract
This review highlights some established and some more contemporary mechanisms responsible for heart failure (HF)-induced skeletal muscle wasting and weakness. We first describe the effects of HF on the relationship between protein synthesis and degradation rates, which determine muscle mass, the involvement of the satellite cells for continual muscle regeneration, and changes in myofiber calcium homeostasis linked to contractile dysfunction. We then highlight key mechanistic effects of both aerobic and resistance exercise training on skeletal muscle in HF and outline its application as a beneficial treatment. Overall, HF causes multiple impairments related to autophagy, anabolic-catabolic signaling, satellite cell proliferation, and calcium homeostasis, which together promote fiber atrophy, contractile dysfunction, and impaired regeneration. Although both wasting and weakness are partly rescued by aerobic and resistance exercise training in HF, the effects of satellite cell dynamics remain poorly explored.
Collapse
Affiliation(s)
- Harrison Gallagher
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Paul W Hendrickse
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Marcelo G Pereira
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - T Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
11
|
Malandish A, Gulati M. The impacts of exercise interventions on inflammaging markers in overweight/obesity patients with heart failure: A systematic review and meta-analysis of randomized controlled trials. IJC HEART & VASCULATURE 2023; 47:101234. [PMID: 37416483 PMCID: PMC10320319 DOI: 10.1016/j.ijcha.2023.101234] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/22/2023] [Accepted: 06/18/2023] [Indexed: 07/08/2023]
Abstract
Objectives The purpose of this meta-analysis was to investigate the association of aerobic, resistance and concurrent exercises vs. control group on inflammaging markers [tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), IL-1-beta, IL-8, and high sensitivity C-reactive protein (hs-CRP)] in overweight or obesity patients with heart failure (HF). Methods The databases of Scopus, PubMed, Web of Science and Google Scholar were searched until August 31, 2022 for exercise interventions vs. control group on circulating inflammaging markers in patients with HF. Only randomized controlled trial (RCT) articles were included. Standardized mean difference (SMD) and 95% confidence intervals (95%CIs) were calculated (registration code = CRD42022347164). Results Forty-six full-text articles (57 intervention arms and 3693 participants) were included. A significant reduction was occurred in inflammaging markers of IL-6 [SMD-0.205(95% CI:-0.332 to -0.078),p = 0.002] and hs-CRP [SMD -0.379 (95% CI:-0.556 to -0.202), p = 0.001] with exercise training in patients with HF. Analysis of subgroup by age, body mass index (BMI), type, intensity, duration of exercise and mean left ventricular ejection fraction (LVEF) revealed that there was a significant reduction in TNF-α for middle-aged (p = 0.031), concurrent training (p = 0.033), high intensity (p = 0.005), and heart failure with reduced ejection fraction (HFrEF) (p = 0.007) compared to the control group. There was a significant reduction in IL-6 for middle-aged (p = 0.006), overweight (p = 0.001), aerobic exercise (p = 0.001), both high and moderate intensities (p = 0.037 and p = 0.034), short-term follow-up (p = 0.001), and heart failure with preserved ejection fraction (HFpEF) (p = 0.001) compared to the control group. There was a significant reduction in hs-CRP for middle-aged (p = 0.004), elderly-aged (p = 0.001), overweight (p = 0.001), aerobic exercise (p = 0.001), concurrent training (p = 0.031), both high and moderate intensities (p = 0.017 and p = 0.001), short-term (p = 0.011), long-term (p = 0.049), and very long-term (p = 0.016) follow-ups, HFrEF (p = 0.003) and heart failure with mildly reduced ejection fraction (HFmrEF) (p = 0.048) compared to the control group. Conclusions The results confirmed that aerobic exercise and concurrent training interventions were effective to improve inflammaging markers of TNF-α, IL-6, and hs-CRP. These exercise-related anti-inflammaging responses were observed across ages (middle-aged and elderly-aged), exercise intensities, duration of follow-ups, and mean LVEFs (HFrEF, HFmrEF and HFpEF) in overweight patients with HF.
Collapse
Affiliation(s)
- Abbas Malandish
- Department of Exercise Physiology, Faculty of Sport Sciences, Urmia University, No. 19, Shams Tabrizi St., Velayat Ave., Keikhali Zone, Yamchi, East Azerbaijan, Urmia, Iran
| | - Martha Gulati
- Barbra Streisand Women’s Heart Center, Smidt Heart Institute, Cedars Sinai Medical Center, 127 S. San Vicente Blvd, Suite A3600, Los Angeles, CA 90048, USA
| |
Collapse
|
12
|
Ispoglou T, Wilson O, McCullough D, Aldrich L, Ferentinos P, Lyall G, Stavropoulos-Kalinoglou A, Duckworth L, Brown MA, Sutton L, Potts AJ, Archbold V, Hargreaves J, McKenna J. A Narrative Review of Non-Pharmacological Strategies for Managing Sarcopenia in Older Adults with Cardiovascular and Metabolic Diseases. BIOLOGY 2023; 12:892. [PMID: 37508325 PMCID: PMC10376679 DOI: 10.3390/biology12070892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 07/30/2023]
Abstract
This narrative review examines the mechanisms underlying the development of cardiovascular disease (CVD) and metabolic diseases (MDs), along with their association with sarcopenia. Furthermore, non-pharmacological interventions to address sarcopenia in patients with these conditions are suggested. The significance of combined training in managing metabolic disease and secondary sarcopenia in type II diabetes mellitus is emphasized. Additionally, the potential benefits of resistance and aerobic training are explored. This review emphasises the role of nutrition in addressing sarcopenia in patients with CVD or MDs, focusing on strategies such as optimising protein intake, promoting plant-based protein sources, incorporating antioxidant-rich foods and omega-3 fatty acids and ensuring sufficient vitamin D levels. Moreover, the potential benefits of targeting gut microbiota through probiotics and prebiotic fibres in sarcopenic individuals are considered. Multidisciplinary approaches that integrate behavioural science are explored to enhance the uptake and sustainability of behaviour-based sarcopenia interventions. Future research should prioritise high-quality randomized controlled trials to refine exercise and nutritional interventions and investigate the incorporation of behavioural science into routine practices. Ultimately, a comprehensive and multifaceted approach is essential to improve health outcomes, well-being and quality of life in older adults with sarcopenia and coexisting cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
| | - Oliver Wilson
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | | | - Luke Aldrich
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | | | - Gemma Lyall
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | | | - Lauren Duckworth
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | - Meghan A Brown
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | - Louise Sutton
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | - Alexandra J Potts
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | - Victoria Archbold
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | - Jackie Hargreaves
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | - Jim McKenna
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| |
Collapse
|
13
|
Malandish A, Karimi A, Naderi M, Ghadamyari N, Gulati M. Impacts of Exercise Interventions on Inflammatory Markers and Vascular Adhesion Molecules in Patients With Heart Failure: A Meta-analysis of RCTs. CJC Open 2023; 5:429-453. [PMID: 37397615 PMCID: PMC10314121 DOI: 10.1016/j.cjco.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Background The aim of this meta-analysis was to investigate the effects of concurrent, aerobic, and resistance exercise on markers of inflammation and vascular adhesion molecules (high-sensitivity C-reactive protein [hs-CRP], interleukin [IL]-6, tumour necrosis factor-alpha [TNF-α], soluble intercellular adhesion molecule-1 [sICAM-1], soluble vascular cell adhesion molecule-1 [sVCAM-1], fibrinogen, IL-1-β, IL-10, IL-18, and E-selectin) in patients with heart failure (HF). Methods The PubMed, Scopus, Web of Science, and Google Scholar databases were searched for dates up to August 31, 2022. Randomized controlled trial studies for exercise interventions on circulating inflammatory and vascular adhesion markers in patients with HF were included. Standardized mean difference (SMD) and 95% confidence interval (CI) were calculated. Results A total of 45 articles were included. Exercise training significantly reduced hs-CRP (SMD -0.441 [95% CI: -0.642 to -0.240], P = 0.001), IL-6 (SMD -0.158 (95% CI: -0.303 to -0.013], P = 0.032), and sICAM-1 (SMD -0.282 [95% CI: -0.477 to -0.086], P = 0.005) markers. Analysis of subgroups revealed that a significant reduction occurred in hs-CRP level for the following subgroups: middle-aged, elderly, overweight status, aerobic exercise, concurrent training, both high and moderate intensity, and short-term, long-term, and very long-term follow-up, compared to a control group (P < 0.05). A significant reduction occurred in IL-6 and sICAM-1 levels for those in the following subgroups, compared to a control group (P < 0.05): middle-aged, aerobic exercise, moderate-intensity exercise, and short-term follow-up. A reduction in TNF-α level occurred for middle-aged patients, compared to a control-group (P < 0.05). Conclusions These exercise-related changes (improved inflammation and vascular adhesion markers) as clinical benefits in general, and for exercise-based cardiac rehabilitation in a more-specific format, improve clinical evolution and survival in patients with HF of different etiologies (registration number = CRD42021271423).
Collapse
Affiliation(s)
- Abbas Malandish
- Department of Exercise Physiology, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| | - Asma Karimi
- Department of Exercise Physiology, Faculty of Sport Sciences, Hakim Sabzevari University, Sabzevar, Iran
| | - Mahdi Naderi
- Department of Exercise Physiology, Faculty of Sport Sciences, Kharazmi University, Tehran, Iran
| | - Niloufar Ghadamyari
- Department of Exercise Physiology, Faculty of Sport Health Sciences, Ankara University, Ankara, Turkey
| | - Martha Gulati
- Barbra Streisand Women’s Heart Center, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
14
|
Brock Symons T, Park J, Kim JH, Kwon EH, Delacruz J, Lee J, Park Y, Chung E, Lee S. Attenuation of skeletal muscle atrophy via acupuncture, electro-acupuncture, and electrical stimulation. Integr Med Res 2023; 12:100949. [PMID: 37214317 PMCID: PMC10192920 DOI: 10.1016/j.imr.2023.100949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/19/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Background Accelerated skeletal muscle wasting is a shared trait among many pathologies and aging. Acupuncture has been used as a therapeutic intervention to control pain; however, little is known about its effects on skeletal muscle atrophy and function. The study's purpose was to compare the effects of acupuncture, electro-acupuncture, and electrical stimulation on cast-induced skeletal muscle atrophy. Methods Forty female Sprague Dawley rats were randomly divided into groups: Control, casted (CAST), CAST+Acupuncture (CAST-A), 4) CAST+Electro-acupuncture (CAST-EA), and CAST+Electrical stimulation (CAST-ES) (n = 8). Plaster casting material was wrapped around the left hind limb. Acupuncture and electro-acupuncture (10 Hz, 6.4 mA) treatments were applied by needling acupoints (stomach-36 and gallbladder-34). Electrical stimulation (10 Hz, 6.4 mA) was conducted by needling the lateral and medial gastrocnemius muscles. Treatments were conducted for 15 min, three times/week for 14 days. Muscle atrophy F-box (MAFbx), muscle RING finger 1 (MuRF1), and contractile properties were assessed. Results Fourteen days of cast-immobilization decreased muscle fiber CSA by 56% in the CAST group (p = 0.00); whereas, all treatment groups demonstrated greater muscle fiber CSA than the CAST group (p = 0.00). Cast-immobilization increased MAFbx and MuRF1 protein expression in the CAST group (p<0.01) while the CAST-A, CAST-EA, and CAST-ES groups demonstrated lower levels of MAFbx and MuRF1 protein expression (p<0.02) compared to the CAST group. Following fourteen days of cast-immobilization, peak twitch tension did not differ between the CAST-A and CON groups (p = 0.12). Conclusion Skeletal muscle atrophy, induced by 14 days of cast-immobilization, was significantly attenuated by acupuncture, electro-acupuncture, or electrical stimulation.
Collapse
Affiliation(s)
- T. Brock Symons
- Department of Counselling, Health, and Kinesiology, Texas A&M University-San Antonio, San Antonio, TX, U.S.A
| | - Jinho Park
- Department of Counselling, Health, and Kinesiology, Texas A&M University-San Antonio, San Antonio, TX, U.S.A
| | - Joo Hyun Kim
- Department of Counselling, Health, and Kinesiology, Texas A&M University-San Antonio, San Antonio, TX, U.S.A
| | - Eun Hye Kwon
- Department of Counselling, Health, and Kinesiology, Texas A&M University-San Antonio, San Antonio, TX, U.S.A
| | - Jesse Delacruz
- Department of Counselling, Health, and Kinesiology, Texas A&M University-San Antonio, San Antonio, TX, U.S.A
| | - Junghoon Lee
- Department of Health and Human Performance, University of Houston, Houston, TX, U.S.A
| | - Yoonjung Park
- Department of Health and Human Performance, University of Houston, Houston, TX, U.S.A
| | - Eunhee Chung
- Department of Kinesiology, The University of Texas at San Antonio, San Antonio, TX, U.S.A
| | - Sukho Lee
- Department of Counselling, Health, and Kinesiology, Texas A&M University-San Antonio, San Antonio, TX, U.S.A
| |
Collapse
|
15
|
Zuo X, Li X, Tang K, Zhao R, Wu M, Wang Y, Li T. Sarcopenia and cardiovascular diseases: A systematic review and meta-analysis. J Cachexia Sarcopenia Muscle 2023; 14:1183-1198. [PMID: 37002802 PMCID: PMC10235887 DOI: 10.1002/jcsm.13221] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 12/23/2022] [Accepted: 03/06/2023] [Indexed: 06/03/2023] Open
Abstract
Sarcopenia is an age-related disease and is often accompanied by other diseases. Now, many studies have shown that cardiovascular diseases (CVDs) may raise the incidence rate of sarcopenia. Therefore, the purpose of this study was to conduct a systematic review and meta-analysis to investigate the prevalence of sarcopenia in patients with CVDs compared with the general population, defined as relatively healthy non-hospitalized subjects. The databases of PubMed, Embase, Medline and Web of Science were searched for eligible studies published up to 12 November 2022. Two assessment tools were used to evaluate study quality and the risk of bias. Statistical analysis was conducted using STATA 14.0 and R Version 4.1.2. Thirty-eight out of the 89 629 articles retrieved were included in our review. The prevalence of sarcopenia ranged from 10.1% to 68.9% in patients with CVDs, and the pooled prevalence was 35% (95% confidence interval [95% CI]: 28-42%). The pooled prevalence of sarcopenia was 32% (95% CI: 23-41%) in patients with chronic heart failure (CHF), 61% (95% CI: 49-72%) in patients with acute decompensated heart failure (ADHF), 43% (95% CI: 2-85%) in patients with coronary artery disease, 30% (95% CI: 25-35%) in patients with cardiac arrhythmia (CA), 35% (95% CI: 10-59%) in patients with congenital heart disease and 12% (95% CI: 7-17%) in patients with unclassed CVDs. However, in the general population, the prevalence of sarcopenia varied from 2.9% to 28.6% and the pooled prevalence was 13% (95% CI: 9-17%), suggesting that the prevalence of sarcopenia in patients with CVDs was about twice compared with the general population. The prevalence of sarcopenia was significantly higher only in patients with ADHF, CHF and CA compared with the general population. There is a positive correlation between CVDs and sarcopenia. The prevalence of sarcopenia is higher in patients with CVDs than that in the general population. With global aging, sarcopenia has brought a heavy burden to individuals and society. Therefore, it is important to identify the populations with high-risk or probable sarcopenia in order to do an early intervention, such as exercise, to counteract or slow down the progress of sarcopenia.
Collapse
Affiliation(s)
- Xinrong Zuo
- Department of AnesthesiologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanChina
| | - Xuehong Li
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Kuo Tang
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Rui Zhao
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Minming Wu
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Yang Wang
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Tao Li
- Department of AnesthesiologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanChina
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengduSichuanChina
| |
Collapse
|
16
|
Setiawan T, Sari IN, Wijaya YT, Julianto NM, Muhammad JA, Lee H, Chae JH, Kwon HY. Cancer cachexia: molecular mechanisms and treatment strategies. J Hematol Oncol 2023; 16:54. [PMID: 37217930 DOI: 10.1186/s13045-023-01454-0] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/13/2023] [Indexed: 05/24/2023] Open
Abstract
Muscle wasting is a consequence of physiological changes or a pathology characterized by increased catabolic activity that leads to progressive loss of skeletal muscle mass and strength. Numerous diseases, including cancer, organ failure, infection, and aging-associated diseases, are associated with muscle wasting. Cancer cachexia is a multifactorial syndrome characterized by loss of skeletal muscle mass, with or without the loss of fat mass, resulting in functional impairment and reduced quality of life. It is caused by the upregulation of systemic inflammation and catabolic stimuli, leading to inhibition of protein synthesis and enhancement of muscle catabolism. Here, we summarize the complex molecular networks that regulate muscle mass and function. Moreover, we describe complex multi-organ roles in cancer cachexia. Although cachexia is one of the main causes of cancer-related deaths, there are still no approved drugs for cancer cachexia. Thus, we compiled recent ongoing pre-clinical and clinical trials and further discussed potential therapeutic approaches for cancer cachexia.
Collapse
Affiliation(s)
- Tania Setiawan
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
| | - Ita Novita Sari
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Yoseph Toni Wijaya
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
| | - Nadya Marcelina Julianto
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
| | - Jabir Aliyu Muhammad
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
| | - Hyeok Lee
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
| | - Ji Heon Chae
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
| | - Hyog Young Kwon
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea.
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea.
| |
Collapse
|
17
|
The complex pathophysiology of cardiac cachexia: A review of current pathophysiology and implications for clinical practice. Am J Med Sci 2023; 365:9-18. [PMID: 36055378 DOI: 10.1016/j.amjms.2022.08.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 04/09/2022] [Accepted: 08/24/2022] [Indexed: 01/04/2023]
Abstract
Cardiac cachexia is a muscle wasting process that often develops in those with chronic heart failure resulting in weight loss, low levels of physical activity, reduced quality of life, and is associated with a poor prognosis. The pathology of cardiac cachexia is complex with new evidence emerging that implicates several body systems. This review describes the pathophysiology associated with cardiac cachexia and addresses: 1) hormonal changes- neurohormonal abnormalities and metabolic hormone imbalance; 2) mechanisms of muscle wasting in cardiac cachexia, and the integral mechanisms between changed hormones due to cardiac cachexia and muscle wasting processes, and 3) associated abnormalities of gastrointestinal system that contribute to cardiac cachexia. These pleiotropic mechanisms demonstrate the intricate interplay between the affected systems and account for why cardiac cachexia is difficult to manage clinically. This review summarises current pathophysiology of cardiac cachexia and highlights symptoms of cardiac cachexia, implications for clinical practice and research gaps.
Collapse
|
18
|
Cai Y, Song W, Li J, Jing Y, Liang C, Zhang L, Zhang X, Zhang W, Liu B, An Y, Li J, Tang B, Pei S, Wu X, Liu Y, Zhuang CL, Ying Y, Dou X, Chen Y, Xiao FH, Li D, Yang R, Zhao Y, Wang Y, Wang L, Li Y, Ma S, Wang S, Song X, Ren J, Zhang L, Wang J, Zhang W, Xie Z, Qu J, Wang J, Xiao Y, Tian Y, Wang G, Hu P, Ye J, Sun Y, Mao Z, Kong QP, Liu Q, Zou W, Tian XL, Xiao ZX, Liu Y, Liu JP, Song M, Han JDJ, Liu GH. The landscape of aging. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2354-2454. [PMID: 36066811 PMCID: PMC9446657 DOI: 10.1007/s11427-022-2161-3] [Citation(s) in RCA: 193] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023]
Abstract
Aging is characterized by a progressive deterioration of physiological integrity, leading to impaired functional ability and ultimately increased susceptibility to death. It is a major risk factor for chronic human diseases, including cardiovascular disease, diabetes, neurological degeneration, and cancer. Therefore, the growing emphasis on "healthy aging" raises a series of important questions in life and social sciences. In recent years, there has been unprecedented progress in aging research, particularly the discovery that the rate of aging is at least partly controlled by evolutionarily conserved genetic pathways and biological processes. In an attempt to bring full-fledged understanding to both the aging process and age-associated diseases, we review the descriptive, conceptual, and interventive aspects of the landscape of aging composed of a number of layers at the cellular, tissue, organ, organ system, and organismal levels.
Collapse
Affiliation(s)
- Yusheng Cai
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Wei Song
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430071, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Jing
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chuqian Liang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Liyuan Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Xia Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wenhui Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Beibei Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Yongpan An
- Peking University International Cancer Institute, Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Jingyi Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Baixue Tang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Siyu Pei
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xueying Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuxuan Liu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Cheng-Le Zhuang
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, China
| | - Yilin Ying
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
- International Laboratory in Hematology and Cancer, Shanghai Jiaotong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China
| | - Xuefeng Dou
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Fu-Hui Xiao
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Dingfeng Li
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ruici Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ya Zhao
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China
| | - Yang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Lihui Wang
- Institute of Ageing Research, Hangzhou Normal University, School of Basic Medical Sciences, Hangzhou, 311121, China
| | - Yujing Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- The Fifth People's Hospital of Chongqing, Chongqing, 400062, China.
| | - Xiaoyuan Song
- MOE Key Laboratory of Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
| | - Jie Ren
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Liang Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Jun Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Weiqi Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| | - Zhengwei Xie
- Peking University International Cancer Institute, Peking University Health Science Center, Peking University, Beijing, 100191, China.
| | - Jing Qu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jianwei Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Ye Tian
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Gelin Wang
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China.
| | - Ping Hu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, China.
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, 510005, China.
| | - Jing Ye
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
- International Laboratory in Hematology and Cancer, Shanghai Jiaotong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China.
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Medicine and VAPSHCS, University of Washington, Seattle, 98195, USA.
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Qing-Peng Kong
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Qiang Liu
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China.
| | - Zhi-Xiong Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China.
| | - Yong Liu
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430071, China.
| | - Jun-Ping Liu
- Institute of Ageing Research, Hangzhou Normal University, School of Basic Medical Sciences, Hangzhou, 311121, China.
- Department of Immunology and Pathology, Monash University Faculty of Medicine, Prahran, Victoria, 3181, Australia.
- Hudson Institute of Medical Research, and Monash University Department of Molecular and Translational Science, Clayton, Victoria, 3168, Australia.
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology, Peking University, Beijing, 100871, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
19
|
Bouredji Z, Argaw A, Frenette J. The inflammatory response, a mixed blessing for muscle homeostasis and plasticity. Front Physiol 2022; 13:1032450. [PMID: 36505042 PMCID: PMC9726740 DOI: 10.3389/fphys.2022.1032450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/10/2022] [Indexed: 11/24/2022] Open
Abstract
Skeletal muscle makes up almost half the body weight of heathy individuals and is involved in several vital functions, including breathing, thermogenesis, metabolism, and locomotion. Skeletal muscle exhibits enormous plasticity with its capacity to adapt to stimuli such as changes in mechanical loading, nutritional interventions, or environmental factors (oxidative stress, inflammation, and endocrine changes). Satellite cells and timely recruited inflammatory cells are key actors in muscle homeostasis, injury, and repair processes. Conversely, uncontrolled recruitment of inflammatory cells or chronic inflammatory processes leads to muscle atrophy, fibrosis and, ultimately, impairment of muscle function. Muscle atrophy and loss of function are reported to occur either in physiological situations such as aging, cast immobilization, and prolonged bed rest, as well as in many pathological situations, including cancers, muscular dystrophies, and several other chronic illnesses. In this review, we highlight recent discoveries with respect to the molecular mechanisms leading to muscle atrophy caused by modified mechanical loading, aging, and diseases. We also summarize current perspectives suggesting that the inflammatory process in muscle homeostasis and repair is a double-edged sword. Lastly, we review recent therapeutic approaches for treating muscle wasting disorders, with a focus on the RANK/RANKL/OPG pathway and its involvement in muscle inflammation, protection and regeneration processes.
Collapse
Affiliation(s)
- Zineb Bouredji
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CRCHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC, Canada
| | - Anteneh Argaw
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CRCHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC, Canada
| | - Jérôme Frenette
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CRCHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC, Canada,Département de Réadaptation, Faculté de Médecine, Université Laval, Quebec City, QC, Canada,*Correspondence: Jérôme Frenette,
| |
Collapse
|
20
|
Reed CH, Buhr TJ, Tystahl AC, Bauer EE, Clark PJ, Valentine RJ. The effects of voluntary binge-patterned ethanol ingestion and daily wheel running on signaling of muscle protein synthesis and degradation in female mice. Alcohol 2022; 104:45-52. [PMID: 35926812 DOI: 10.1016/j.alcohol.2022.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 01/26/2023]
Abstract
Excessive ethanol ingestion can reduce skeletal muscle protein synthesis (MPS) through the disruption of signaling along the Akt-mTOR pathway and increase muscle protein degradation (MPD) through the Ubiquitin Proteasome Pathway (UPP) and autophagy. Identification of interventions that curb the disrupting effects of alcohol misuse on MPS and MPD are of central importance for the prevention of chronic health complications that arise from muscle loss. Physical activity is one potential strategy to combat the deleterious effects of alcohol on skeletal muscle. Therefore, the purpose of this study was to investigate the interaction between daily wheel running and binge-patterned ethanol consumption, through episodes of voluntary binge-patterned ethanol drinking, on signaling factors along the Akt-mTOR, Ubiquitin-Proteasome, and autophagy pathways. Adult female C57BL/6J mice received daily access to cages with or without running wheels for 2.5 h/day for five weeks. During the final five days of the study, mice received 2-4 h of daily access to sipper tubes containing water (n = 14 sedentary; n = 15 running) or 20% ethanol (n = 14 sedentary; n = 16 running) 30 min after running wheel access, using the "Drinking in the Dark" (DID) model of binge-patterned ethanol consumption. Immediately after the final episode of DID, gastrocnemius muscle was extracted. Western blotting was performed to measure proteins along Akt-mTOR, Ubiquitin-Proteasome, and autophagy pathways, and PCR was used to assess mRNA expression of atrogenes. Ethanol access increased expression of MAFbx by 82% (p = 0.048), but did not robustly influence Akt-mTOR or UPP signaling. Daily wheel access did not prevent alcohol-induced MAFbx expression; however, ethanol access decreased the phosphorylation of p70S6K by 45% in running mice (p = 0.020). These results suggest that physical activity may be insufficient to prevent alcohol-induced changes to signaling factors along pathways involved in muscle loss. Instead, binge-patterned ethanol ingestion may impair the benefits of physical activity on factors involved in MPS.
Collapse
Affiliation(s)
- Carter H Reed
- Department of Kinesiology, Forker Building, 534 Wallace Road, Iowa State University, Ames, IA, 50011, United States; Interdepartmental Graduate Program of Nutritional Sciences, MacKay Hall, 2302 Osborn Drive, Iowa State University, Ames, IA, 50011, United States
| | - Trevor J Buhr
- Neuroscience Program, MacKay Hall, 2302 Osborn Drive, Iowa State University, Ames, IA, 50011, United States; Department of Food Science and Human Nutrition, MacKay Hall, 2302 Osborn Drive, Iowa State University, Ames, IA, 50011, United States
| | - Anna C Tystahl
- Department of Kinesiology, Forker Building, 534 Wallace Road, Iowa State University, Ames, IA, 50011, United States
| | - Ella E Bauer
- Interdepartmental Graduate Program of Nutritional Sciences, MacKay Hall, 2302 Osborn Drive, Iowa State University, Ames, IA, 50011, United States; Neuroscience Program, MacKay Hall, 2302 Osborn Drive, Iowa State University, Ames, IA, 50011, United States; Department of Food Science and Human Nutrition, MacKay Hall, 2302 Osborn Drive, Iowa State University, Ames, IA, 50011, United States
| | - Peter J Clark
- Interdepartmental Graduate Program of Nutritional Sciences, MacKay Hall, 2302 Osborn Drive, Iowa State University, Ames, IA, 50011, United States; Neuroscience Program, MacKay Hall, 2302 Osborn Drive, Iowa State University, Ames, IA, 50011, United States; Department of Food Science and Human Nutrition, MacKay Hall, 2302 Osborn Drive, Iowa State University, Ames, IA, 50011, United States.
| | - Rudy J Valentine
- Department of Kinesiology, Forker Building, 534 Wallace Road, Iowa State University, Ames, IA, 50011, United States; Interdepartmental Graduate Program of Nutritional Sciences, MacKay Hall, 2302 Osborn Drive, Iowa State University, Ames, IA, 50011, United States; Neuroscience Program, MacKay Hall, 2302 Osborn Drive, Iowa State University, Ames, IA, 50011, United States.
| |
Collapse
|
21
|
Sato R, Vatic M, da Fonseca GWP, von Haehling S. Sarcopenia and Frailty in Heart Failure: Is There a Biomarker Signature? Curr Heart Fail Rep 2022; 19:400-411. [PMID: 36261756 DOI: 10.1007/s11897-022-00575-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/10/2022] [Indexed: 10/24/2022]
Abstract
PURPOSE OF REVIEW Sarcopenia and frailty are common in patients with heart failure (HF) and are strongly associated with prognosis. This review aims to examine promising biomarkers that can guide physicians in identifying sarcopenia and frailty in HF. RECENT FINDINGS Traditional biomarkers including C-reactive protein, aminotransaminase, myostatin, and urinary creatinine as well as novel biomarkers including microRNAs, suppression of tumorigenicity 2 (ST2), galectin-3, and procollagen type III N-terminal peptide may help in predicting the development of sarcopenia and frailty in HF patients. Among those biomarkers, aminotransferase, urinary creatinine, and ST2 predicted the prognosis in HF patients with sarcopenia and frailty. This review outlines the current knowledge of biomarkers that are considered promising for diagnosing sarcopenia and frailty in HF. The listed biomarkers might support the diagnosis, prognosis, and therapeutic decisions for sarcopenia and frailty in HF patients.
Collapse
Affiliation(s)
- Ryosuke Sato
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075, Gottingen, Germany
| | - Mirela Vatic
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075, Gottingen, Germany
| | | | - Stephan von Haehling
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Gottingen, Germany.
| |
Collapse
|
22
|
Winzer EB, Augstein A, Schauer A, Mueller S, Fischer-Schaepmann T, Goto K, Hommel J, van Craenenbroeck EM, Wisløff U, Pieske B, Halle M, Linke A, Adams V. Impact of Different Training Modalities on Molecular Alterations in Skeletal Muscle of Patients With Heart Failure With Preserved Ejection Fraction: A Substudy of the OptimEx Trial. Circ Heart Fail 2022; 15:e009124. [DOI: 10.1161/circheartfailure.121.009124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background:
Exercise intolerance is a cardinal feature of heart failure with preserved ejection fraction and so far exercise training (ET) is the most effective treatment. Since the improvement in exercise capacity is only weakly associated with changes in diastolic function other mechanisms, like changes in the skeletal muscle, contribute to improvement in peak oxygen consumption. The aim of the present study was to analyze molecular changes in skeletal muscle of patients with heart failure with preserved ejection fraction performing different ET modalities.
Methods:
Skeletal muscle biopsies were taken at study begin and after 3 and 12 months from patients with heart failure with preserved ejection fraction randomized either into a control group (guideline based advice for ET), a high-intensity interval training group (HIIT) or a moderate continuous training group. The first 3 months of ET were supervised in-hospital followed by 9 months home-based ET. Protein and mRNA expression of atrophy-related proteins, enzyme activities of enzymes linked to energy metabolism and satellite cells (SCs) were quantified.
Results:
Exercise capacity improved 3 months after moderate continuous exercise training and HIIT. This beneficial effect was lost after 12 months. HIIT mainly improved markers of energy metabolism and the amount and function of SC, with minor changes in markers for muscle atrophy. Only slight changes were observed after moderate continuous exercise training. The molecular changes were no longer detectable after 12 months.
Conclusions:
Despite similar improvements in exercise capacity by HIIT and moderate continuous exercise training after 3 months, only HIIT altered proteins related to energy metabolism and amount/function of SC. These effects were lost after switching from in-hospital to at-home-based ET.
Registration:
URL:
https://www.clinicaltrials.gov
; Unique identifier: NCT02078947.
Collapse
Affiliation(s)
- Ephraim B. Winzer
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Antje Augstein
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Antje Schauer
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Stephan Mueller
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany (S.M., M.H.)
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (S.M., M.H.)
| | - Tina Fischer-Schaepmann
- Department of Internal Medicine/Cardiology, Heart Center Leipzig – University Hospital, Helios Stiftungsprofessur, Germany (T.F.-S.)
| | - Keita Goto
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Jennifer Hommel
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Emeline M. van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Belgium (E.M.v.C.)
- Department of Cardiology, Antwerp University Hospital, Belgium (E.M.v.C.)
| | - Ulrik Wisløff
- Cardiac Exercise Research Group at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway (U.W.)
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Germany (B.P.)
| | - Martin Halle
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany (S.M., M.H.)
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (S.M., M.H.)
| | - Axel Linke
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Volker Adams
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Germany (V.A.)
| |
Collapse
|
23
|
Abstract
Cachexia is a complex wasting syndrome, accompanying a variety of end-stage chronic diseases, such as cancer, heart failure and human immunodeficiency virus (HIV) infection/acquired immunodeficiency syndrome (AIDS). It significantly affects patients' quality of life and survival. Multiple therapeutic approaches have been studied over time. However, despite promising results, no drug has been approved to date. In this review, we examine and discuss the available data on the therapeutic effects of androgens and selective androgen receptor modulators (SARMs) for cachexia.
Collapse
Affiliation(s)
- Luca Giovanelli
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20100, Milan, Italy; Department of Endocrine and Metabolic Medicine, IRCCS Istituto Auxologico Italiano, 20100, Milan, Italy; Department of Endocrinology, Diabetes & Metabolism, Newcastle-upon-Tyne Hospitals, NE1 4LP, UK.
| | - Richard Quinton
- Department of Endocrinology, Diabetes & Metabolism, Newcastle-upon-Tyne Hospitals, NE1 4LP, UK; Translational & Clinical Research Institute, University of Newcastle-upon-Tyne, NE1 3BZ, UK.
| |
Collapse
|
24
|
da Silva Soares DB, Shinjo SK, Santos AS, de Cassia Rosa de Jesus J, Schenk S, de Castro GS, Zanoteli E, Krustrup P, da Silva MER, de Sousa MV. Skeletal muscle gene expression in older adults with type 2 diabetes mellitus undergoing calorie-restricted diet and recreational sports training - a randomized clinical trial. Exp Gerontol 2022; 164:111831. [PMID: 35525396 DOI: 10.1016/j.exger.2022.111831] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/10/2022] [Accepted: 05/02/2022] [Indexed: 11/27/2022]
Abstract
AIMS This study aimed to evaluate the impact of a 12-week calorie-restricted diet and recreational sports training on gene expressions IL15, ATROGIN1 and MURF-1 in skeletal muscle of T2D patients. METHODS Older adults with T2D (n = 39, 60 ± 6.0 years, BMI 33.5 ± 0.6 kg/m2) were randomly allocated to Diet+Soccer (DS), Diet+Running (DR) or Diet (D). The training sessions were moderate-to-high-intensity and performed 3 × 40 min/week for 12-weeks. Gene expression from vastus lateralis muscle obtained by qRT-PCR, dual-energy X-ray and fasting blood testing measurements were performed before and after 12-weeks. Statistical analysis adopted were two-way ANOVA and Paired t-test for gene expression, and RM-ANOVA test for the remainder variables. RESULTS Total body weight was reduced in ~4 kg representing body fat mass in all groups after 12-weeks (P < 0.05). HbA1c values decreased in all groups post-intervention. Lipids profile improved in the training groups (P < 0.05) after 12-weeks. ATROGIN1 and MURF-1 mRNA reduced in the DS (1.084 ± 0.14 vs. 0.754 ± 1.14 and (1.175 ± 0.34 vs. 0.693 ± 0.12, respectively; P < 0.05), while IL15 mRNA increased in the DR (1056 ± 0,12 vs. 1308 ± 0,13; P < 0.05) after 12-weeks intervention. CONCLUSION Recreational training with a moderate calorie-restricted diet can downregulates the expression of atrophy-associated myokines and increases the expression of anti-inflammatory gene IL15.
Collapse
Affiliation(s)
- Diana Bento da Silva Soares
- Laboratory of Medical Investigation, LIM-18, Endocrinology Division, School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Aritânia Sousa Santos
- Laboratory of Medical Investigation, LIM-18, Endocrinology Division, School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Simon Schenk
- Department of Orthopaedic Surgery, University of California San Diego, La Jolla, CA, USA
| | - Gabriela Salim de Castro
- Institute of Biomedical Sciences, Cancer Metabolism Research Group, University of São Paulo, São Paulo, Brazil
| | - Edmar Zanoteli
- Department of Neurology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Peter Krustrup
- Department of Sports Science and Clinical Biomechanics, SDU Sport and Health Sciences Cluster (SHSC), University of Southern Denmark, Odense, Denmark
| | - Maria Elizabeth Rossi da Silva
- Laboratory of Medical Investigation, LIM-18, Endocrinology Division, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Maysa Vieira de Sousa
- Laboratory of Medical Investigation, LIM-18, Endocrinology Division, School of Medicine, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
25
|
Valentova M, Anker SD, von Haehling S. Cardiac Cachexia Revisited. Cardiol Clin 2022; 40:199-207. [DOI: 10.1016/j.ccl.2021.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
26
|
Guan Y, Yan Z. Molecular Mechanisms of Exercise and Healthspan. Cells 2022; 11:872. [PMID: 35269492 PMCID: PMC8909156 DOI: 10.3390/cells11050872] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 12/16/2022] Open
Abstract
Healthspan is the period of our life without major debilitating diseases. In the modern world where unhealthy lifestyle choices and chronic diseases taper the healthspan, which lead to an enormous economic burden, finding ways to promote healthspan becomes a pressing goal of the scientific community. Exercise, one of humanity's most ancient and effective lifestyle interventions, appears to be at the center of the solution since it can both treat and prevent the occurrence of many chronic diseases. Here, we will review the current evidence and opinions about regular exercise promoting healthspan through enhancing the functionality of our organ systems and preventing diseases.
Collapse
Affiliation(s)
- Yuntian Guan
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA;
- Center for Skeletal Muscle Research at the Robert M. Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Zhen Yan
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA;
- Center for Skeletal Muscle Research at the Robert M. Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
- Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
- Department of Molecular Physiology and Biological Biophysics, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
27
|
Skeletal muscle derived Musclin protects the heart during pathological overload. Nat Commun 2022; 13:149. [PMID: 35013221 PMCID: PMC8748430 DOI: 10.1038/s41467-021-27634-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 12/02/2021] [Indexed: 12/14/2022] Open
Abstract
Cachexia is associated with poor prognosis in chronic heart failure patients, but the underlying mechanisms of cachexia triggered disease progression remain poorly understood. Here, we investigate whether the dysregulation of myokine expression from wasting skeletal muscle exaggerates heart failure. RNA sequencing from wasting skeletal muscles of mice with heart failure reveals a reduced expression of Ostn, which encodes the secreted myokine Musclin, previously implicated in the enhancement of natriuretic peptide signaling. By generating skeletal muscle specific Ostn knock-out and overexpressing mice, we demonstrate that reduced skeletal muscle Musclin levels exaggerate, while its overexpression in muscle attenuates cardiac dysfunction and myocardial fibrosis during pressure overload. Mechanistically, Musclin enhances the abundance of C-type natriuretic peptide (CNP), thereby promoting cardiomyocyte contractility through protein kinase A and inhibiting fibroblast activation through protein kinase G signaling. Because we also find reduced OSTN expression in skeletal muscle of heart failure patients, augmentation of Musclin might serve as therapeutic strategy.
Collapse
|
28
|
Supriya R, Singh KP, Gao Y, Gu Y, Baker JS. Effect of Exercise on Secondary Sarcopenia: A Comprehensive Literature Review. BIOLOGY 2021; 11:biology11010051. [PMID: 35053049 PMCID: PMC8773430 DOI: 10.3390/biology11010051] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 01/01/2023]
Abstract
Simple Summary Sarcopenia is an inevitable component of aging. It is officially recognized as a muscle disease with an ICD-10-MC diagnosis code that can be used to bill for care in some countries. Sarcopenia can be classified into primary or age-related sarcopenia and secondary sarcopenia. The condition is referred to as secondary sarcopenia when any other comorbidities are present in conjunction with aging. Secondary sarcopenia is more prevalent than primary sarcopenia and requires special attention. Exercise interventions may help in our understanding and prevention of sarcopenia with a specific morbidity Glomerular filtration rate that exercise improves muscle mass, quality or physical function in elderly subjects with cancer, type 2 diabetes, kidney diseases and lung diseases. In this review, we summarize recent research that has studied the impact of exercise on patients with secondary sarcopenia, specifically those with one comorbid condition. We did not discover any exercise intervention specifically for subjects with secondary sarcopenia (with one comorbidity). Even though there is a strong argument for using exercise to improve muscle mass, quality or physical function in subjects with cancer, type 2 diabetes, kidney diseases, lung diseases and many more, very few studies have reported baseline sarcopenia assessments. Based on the trials summarized in this review, we may propose but not conclude that resistance, aerobic, balance training or even walking can be useful in subjects with secondary sarcopenia with only one comorbidity due to the limited number of trials. This review is significant because it reveals the need for broad-ranging research initiatives involving secondary sarcopenic patients and highlights a large secondary sarcopenia research gap. Abstract Background: Sarcopenia has been recognized as an inevitable part of aging. However, its severity and the age at which it begins cannot be predicted by age alone. The condition can be categorized into primary or age-related sarcopenia and secondary sarcopenia. Sarcopenia is diagnosed as primary when there are no other specific causes. However, secondary sarcopenia occurs if other factors, including malignancy or organ failure, are evident in addition to aging. The prevalence of secondary sarcopenia is far greater than that of primary sarcopenia and requires special attention. To date, nutrition and exercise have proven to be the best methods to combat this disease. The impact of exercise on subjects suffering from sarcopenia with a specific morbidity is worthy of examination for understanding and prevention. The purpose of this review, therefore, is to summarize recent research that has investigated the impact of exercise in patients with secondary sarcopenia, specifically with one comorbidity. Methods: Pubmed, Web of Science, Embase and Medline databases were searched comprehensively with no date limit for randomized controlled trials. The literature was specifically searched for clinical trials in which subjects were sarcopenic with only one comorbidity participating in an exercise intervention. The most visible comorbidities identified and used in the search were lung disease, kidney disease, heart disease, type 2 diabetes, cancer, neurological diseases, osteoporosis and arthritis. Results: A total of 1752 studies were identified that matched the keywords. After removing duplicates, there were 1317 articles remaining. We extracted 98 articles for full screening. Finally, we included 21 relevant papers that were used in this review. Conclusion: Despite a strong rationale for using exercise to improve muscle mass, quality or physical function in subjects with cancer, type 2 diabetes, kidney disease, lung disease and many more, baseline sarcopenia evaluation has been reported in very few trials. The limited number of studies does not allow us to conclude that exercise can improve sarcopenia in patients with other comorbidities. This review highlights the necessity for wide-ranging research initiatives involving secondary sarcopenic patients.
Collapse
Affiliation(s)
- Rashmi Supriya
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China; (Y.G.); (Y.G.); (J.S.B.)
- Centre for Health and Exercise Science Research, Sarcopenia Research Unit, Department of Sport, Physical Education and Health, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong
- Correspondence:
| | - Kumar Purnendu Singh
- FEBT, School of Environment, Resources and Development, Asian Institute of Technology, Klong Luang, Pathum Thani 12120, Thailand;
| | - Yang Gao
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China; (Y.G.); (Y.G.); (J.S.B.)
- Centre for Health and Exercise Science Research, Sarcopenia Research Unit, Department of Sport, Physical Education and Health, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong
| | - Yaodong Gu
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China; (Y.G.); (Y.G.); (J.S.B.)
| | - Julien S. Baker
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China; (Y.G.); (Y.G.); (J.S.B.)
- Centre for Health and Exercise Science Research, Sarcopenia Research Unit, Department of Sport, Physical Education and Health, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong
| |
Collapse
|
29
|
Sasaki KI, Fukumoto Y. Sarcopenia as a comorbidity of cardiovascular disease. J Cardiol 2021; 79:596-604. [PMID: 34906433 DOI: 10.1016/j.jjcc.2021.10.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/27/2022]
Abstract
Sarcopenia, the lowered skeletal muscle mass, weakened skeletal muscle strength, and reduced physical performance with aging, is a component of frailty and high-risk factor for falls, resulting in an increase in mortality. In cardiovascular disease (CVD) patients, systemic inflammation, oxidative stress, overactivation of ubiquitin-proteasome system, endothelial dysfunction, lowering muscle blood flow, impaired glucose tolerance, hormonal changes, and physical inactivity possibly contribute to CVD-related sarcopenia. Prevalence of sarcopenia and osteosarcopenia, which is osteopenia and sarcopenia coexisting together, seems to be higher in CVD patients than in community-dwelling adults, suggesting the necessity of early diagnosis and prevention of CVD-related sarcopenia. Atrial stiffness, coronary artery calcification score, and serum vitamin D levels may be of help as the biomarkers to suspect sarcopenia, and renin-angiotensin-aldosterone system inhibitors may play a role in the medical prevention and treatment of CVD-related sarcopenia. There are few reports to convince the efficacies of dietary and antioxidant supplementation on sarcopenia at present, whereas aerobic and resistance training exercises have been recognized as an effective strategy to prevent and treat sarcopenia.
Collapse
Affiliation(s)
- Ken-Ichiro Sasaki
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan.
| | - Yoshihiro Fukumoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
| |
Collapse
|
30
|
Gene expression changes in vastus lateralis muscle after different strength training regimes during rehabilitation following anterior cruciate ligament reconstruction. PLoS One 2021; 16:e0258635. [PMID: 34648569 PMCID: PMC8516190 DOI: 10.1371/journal.pone.0258635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 10/03/2021] [Indexed: 11/19/2022] Open
Abstract
Impaired muscle regeneration has repeatedly been described after anterior cruciate ligament reconstruction (ACL-R). The results of recent studies provided some evidence for negative alterations in knee extensor muscles after ACL-R causing persisting strength deficits in spite of the regain of muscle mass. Accordingly, we observed that 12 weeks of concentric/eccentric quadriceps strength training with eccentric overload (CON/ECC+) induced a significantly greater hypertrophy of the atrophied quadriceps muscle after ACL-R than conventional concentric/eccentric quadriceps strength training (CON/ECC). However, strength deficits persisted and there was an unexpected increase in the proportion of slow type I fibers instead of the expected shift towards a faster muscle phenotype after CON/ECC+. In order to shed further light on muscle recovery after ACL-R, the steady-state levels of 84 marker mRNAs were analyzed in biopsies obtained from the vastus lateralis muscle of 31 subjects before and after 12 weeks of CON/ECC+ (n = 18) or CON/ECC strength training (n = 13) during rehabilitation after ACL-R using a custom RT2 Profiler PCR array. Significant (p < 0.05) changes were detected in the expression of 26 mRNAs, several of them involved in muscle wasting/atrophy. A different pattern with regard to the strength training mode was observed for 16 mRNAs, indicating an enhanced hypertrophic stimulus, mechanical sensing or fast contractility after CON/ECC+. The effects of the type of autograft (quadriceps, QUAD, n = 19, or semitendinosus tendon, SEMI, n = 12) were reflected in the lower expression of 6 mRNAs involved in skeletal muscle hypertrophy or contractility in QUAD. In conclusion, the greater hypertrophic stimulus and mechanical stress induced by CON/ECC+ and a beginning shift towards a faster muscle phenotype after CON/ECC+ might be indicated by significant gene expression changes as well as still ongoing muscle wasting processes and a negative impact of QUAD autograft.
Collapse
|
31
|
Koh FH, Chua JMW, Tan JLJ, Foo FJ, Tan WJ, Sivarajah SS, Ho LML, Teh BT, Chew MH. Paradigm shift in gastrointestinal surgery − combating sarcopenia with prehabilitation: Multimodal review of clinical and scientific data. World J Gastrointest Surg 2021; 13:734-755. [PMID: 34512898 PMCID: PMC8394378 DOI: 10.4240/wjgs.v13.i8.734] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/08/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023] Open
Abstract
A growing body of evidence has demonstrated the prognostic significance of sarcopenia in surgical patients as an independent predictor of postoperative complications and outcomes. These included an increased risk of total complications, major complications, re-admissions, infections, severe infections, 30 d mortality, longer hospital stay and increased hospitalization expenditures. A program to enhance recovery after surgery was meant to address these complications; however, compliance to the program since its introduction has been less than ideal. Over the last decade, the concept of prehabilitation, or “pre-surgery rehabilitation”, has been discussed. The presurgical period represents a window of opportunity to boost and optimize the health of an individual, providing a compensatory “buffer” for the imminent reduction in physiological reserve post-surgery. Initial results have been promising. We review the literature to critically review the utility of prehabilitation, not just in the clinical realm, but also in the scientific realm, with a resource management point-of-view.
Collapse
Affiliation(s)
- Frederick H Koh
- Division of Surgery, Sengkang General Hospital, Singapore 544886, Singapore
| | - Jason MW Chua
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Singapore 138673, Singapore
| | - Joselyn LJ Tan
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Singapore 138673, Singapore
| | - Fung-Joon Foo
- Division of Surgery, Sengkang General Hospital, Singapore 544886, Singapore
| | - Winson J Tan
- Division of Surgery, Sengkang General Hospital, Singapore 544886, Singapore
| | | | - Leonard Ming Li Ho
- Division of Surgery, Sengkang General Hospital, Singapore 544886, Singapore
| | - Bin-Tean Teh
- Duke-NUS Graduate Medical School, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Min-Hoe Chew
- Division of Surgery, Sengkang General Hospital, Singapore 544886, Singapore
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Heart failure (HF) is a structural or functional cardiac abnormality which leads to failure of the heart to deliver oxygen commensurately with the requirements of the tissues and it may progress to a generalized wasting of skeletal muscle, fat tissue, and bone tissue (cardiac cachexia). Clinically, dyspnea, fatigue, and exercise intolerance are some typical signs and symptoms that characterize HF patients. This review focused on the phenotypic characteristics of HF-induced skeletal myopathy as well as the mechanisms of muscle wasting due to HF and highlighted possible therapeutic strategies for skeletal muscle wasting in HF. RECENT FINDINGS The impaired exercise capacity of those patients is not attributed to the reduced blood flow in the exercising muscles, but rather to abnormal metabolic responses, myocyte apoptosis and atrophy of skeletal muscle. Specifically, the development of skeletal muscle wasting in chronic HF is characterized by structural, metabolic, and functional abnormalities in skeletal muscle and may be a result not only of reduced physical activity, but also of metabolic or hormonal derangements that favour catabolism over anabolism. In particular, abnormal energy metabolism, mitochondrial dysfunction, transition of myofibers from type I to type II, muscle atrophy, and reduction in muscular strength are included in skeletal muscle abnormalities which play a central role in the decreased exercise capacity of HF patients. Skeletal muscle alterations and exercise intolerance observed in HF are reversible by exercise training, since it is the only demonstrated intervention able to improve skeletal muscle metabolism, growth factor activity, and functional capacity and to reverse peripheral abnormalities.
Collapse
|
33
|
Adams V, Wunderlich S, Mangner N, Hommel J, Esefeld K, Gielen S, Halle M, Ellingsen Ø, Van Craenenbroeck EM, Wisløff U, Pieske B, Linke A, Winzer EB. Ubiquitin-proteasome-system and enzymes of energy metabolism in skeletal muscle of patients with HFpEF and HFrEF. ESC Heart Fail 2021; 8:2556-2568. [PMID: 33955206 PMCID: PMC8318515 DOI: 10.1002/ehf2.13405] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/17/2021] [Accepted: 04/22/2021] [Indexed: 01/14/2023] Open
Abstract
Background Skeletal muscle (SM) alterations contribute to exercise intolerance in heart failure patients with preserved (HFpEF) or reduced (HFrEF) left ventricular ejection fraction (LVEF). Protein degradation via the ubiquitin‐proteasome‐system (UPS), nuclear apoptosis, and reduced mitochondrial energy supply is associated with SM weakness in HFrEF. These mechanisms are incompletely studied in HFpEF, and a direct comparison between these groups is missing. Methods and results Patients with HFpEF (LVEF ≥ 50%, septal E/e′ > 15 or >8 and NT‐proBNP > 220 pg/mL, n = 20), HFrEF (LVEF ≤ 35%, n = 20) and sedentary control subjects (Con, n = 12) were studied. Inflammatory markers were measured in serum, and markers of the UPS, nuclear apoptosis, and energy metabolism were determined in percutaneous SM biopsies. Both HFpEF and HFrEF showed increased proteolysis (MuRF‐1 protein expression, ubiquitination, and proteasome activity) with proteasome activity significantly related to interleukin‐6. Proteolysis was more pronounced in patients with lower exercise capacity as indicated by peak oxygen uptake in per cent predicted below the median. Markers of apoptosis did not differ between groups. Mitochondrial energy supply was reduced in HFpEF and HFrEF (complex‐I activity: −31% and −53%; malate dehydrogenase activity: −20% and −29%; both P < 0.05 vs. Con). In contrast, short‐term energy supply via creatine kinase was increased in HFpEF but decreased in HFrEF (47% and −45%; P < 0.05 vs. Con). Conclusions Similarly to HFrEF, skeletal muscle in HFpEF is characterized by increased proteolysis linked to systemic inflammation and reduced exercise capacity. Energy metabolism is disturbed in both groups; however, its regulation seems to be severity‐dependent.
Collapse
Affiliation(s)
- Volker Adams
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany.,Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany
| | - Sebastian Wunderlich
- Department of Internal Medicine/Cardiology, Heart Center Leipzig - University Hospital, Leipzig, Germany
| | - Norman Mangner
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany
| | - Jennifer Hommel
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany
| | - Katrin Esefeld
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Prevention and Sports Medicine, Technische Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Stephan Gielen
- Department of Cardiology, Angiology and Intensive Care, Klinikum Lippe, Detmold, Germany
| | - Martin Halle
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Prevention and Sports Medicine, Technische Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Øyvind Ellingsen
- Department of Cardiology, St. Olavs University Hospital, Trondheim, Norway.,The Cardiac Exercise Research Group at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Emeline M Van Craenenbroeck
- Department of Cardiology, Antwerp University Hospital, Edegem, Belgium.,Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Ulrik Wisløff
- The Cardiac Exercise Research Group at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Burkert Pieske
- Department Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Axel Linke
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany.,Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany
| | - Ephraim B Winzer
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany
| |
Collapse
|
34
|
Mangner N, Garbade J, Heyne E, van den Berg M, Winzer EB, Hommel J, Sandri M, Jozwiak-Nozdrzykowska J, Meyer AL, Lehmann S, Schmitz C, Malfatti E, Schwarzer M, Ottenheijm CAC, Bowen TS, Linke A, Adams V. Molecular Mechanisms of Diaphragm Myopathy in Humans With Severe Heart Failure. Circ Res 2021; 128:706-719. [PMID: 33535772 DOI: 10.1161/circresaha.120.318060] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Norman Mangner
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
| | - Jens Garbade
- Department of Cardiac Surgery (J.G., S.L.), Heart Center Leipzig - University Hospital, Germany
| | - Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Germany (E.H., M.S.)
| | | | - Ephraim B Winzer
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
| | - Jennifer Hommel
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
| | - Marcus Sandri
- Department of Cardiology (M.S., J.J.-N.), Heart Center Leipzig - University Hospital, Germany
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Germany (E.H., M.S.)
| | | | - Anna L Meyer
- Cardiac Surgery, Heart and Marfan Center, University of Heidelberg, Germany (A.L.M.)
| | - Sven Lehmann
- Department of Cardiac Surgery (J.G., S.L.), Heart Center Leipzig - University Hospital, Germany
| | - Clara Schmitz
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
| | - Edoardo Malfatti
- Neurology, Centre de Référence Maladies Neuromusculaires Nord-Est-Ile-de-France, CHU Raymond-Poincaré, Garches, France (E.M.). U1179 UVSQ-INSERM, Université Versailles-Saint-Quentin-en-Yvelines, France
| | | | - Coen A C Ottenheijm
- Physiology, Amsterdam UMC (location VUmc), the Netherlands (M.v.d.B., C.A.C.O.)
| | - T Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, United Kingdom (T.S.B.)
| | - Axel Linke
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany (A.L., V.A.)
| | - Volker Adams
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany (A.L., V.A.)
| |
Collapse
|
35
|
Abstract
Although muscle loss is part of the natural course of human aging, sarcopenia has been associated with an increased risk of physical disability and mortality in older patients. Many heart failure patients concomitantly develop deficits in muscle mass and strength, resulting in decreased quality of life and exercise capacity. An underlying state of inflammation is central to the development of sarcopenia and muscle wasting in heart failure; however, additional research in human models is needed to further delineate the pathophysiology of muscle wasting in these patients. Previous studies have shed light on many of the potential targets for therapeutic intervention of sarcopenia in heart failure; however, physical exercise remains the prominent beneficial intervention. Future research must explore other therapeutic interventions in randomized, double-blind clinical trials, which may help to supplement exercise regimens. Sarcopenia shows promise as an easily measured predictor of outcomes after transcatheter aortic valve replacement.
Collapse
|
36
|
Chen L, Wang R, Liu H, Wei S, Jing M, Wang M, Zhao Y. Clinical Efficacy and Safety of Qishen Yiqi Dropping Pill Combined with Conventional Western Medicine in the Treatment of Chronic Heart Failure: A Systematic Review and Meta-Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6612653. [PMID: 33603818 PMCID: PMC7872761 DOI: 10.1155/2021/6612653] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 12/26/2020] [Accepted: 01/16/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVE The systematic review was designed to evaluate the safety and efficacy of Qishen Yiqi dropping pill combined with conventional Western medicine in the treatment of chronic heart failure (CHF). METHODS Relevant randomized controlled trials (RCTs) investigating the clinical efficacy of Qishen Yiqi dropping pill combined with conventional Western medicine in treating CHF were widely searched in electronic databases, including PubMed, Cochrane Library, EMBASE, CBM, CNKI, Read-show database, VIP database, and WanFang up to December 26, 2020. The methodological quality of each trial was assessed according to the Cochrane Reviewers' Handbook 5.0. Meta-analysis was performed by using Review Manager 5.3. RESULTS Twenty-one RCTs (N = 2162) that met the criteria were included in the review for the assessment of methodological quality. Meta-analysis showed that compared with the conventional Western medicine (control group), Qishen Yiqi dropping pill combined with conventional Western medicine (experience group) significantly improved clinical efficiency, left ventricular end-diastolic diameter (LVEDD), left ventricular end-systolic diameter (LVESD), left ventricular ejection fraction (LVEF), brain natriuretic peptide level (BNP), 6 min-walk distance (6-MWD), and adverse reactions. CONCLUSION Qishen Yiqi dropping pill combined with conventional Western medicine are better than conventional Western medicine alone to improve the indicators of patients with CHF, which provides a certain basis for the treatment of CHF.
Collapse
Affiliation(s)
- Lisheng Chen
- Department of Pharmacy, Fifth Medical Center, General Hospital of Chinese PLA, Beijing, China
- Department of Pharmacy, Hebei North University, Zhangjiakou, China
| | - Ruilin Wang
- Department of Traditional Chinese Medicine, Fifth Medical Center, General Hospital of Chinese PLA, Beijing, China
| | - Honghong Liu
- Integrative Medical Center, Fifth Medical Center, General Hospital of Chinese PLA, Beijing, China
| | - Shizhang Wei
- Department of Pharmacy, Fifth Medical Center, General Hospital of Chinese PLA, Beijing, China
| | - Manyi Jing
- Department of Pharmacy, Fifth Medical Center, General Hospital of Chinese PLA, Beijing, China
| | - Min Wang
- Department of Pharmacy, Fifth Medical Center, General Hospital of Chinese PLA, Beijing, China
| | - Yanling Zhao
- Department of Pharmacy, Fifth Medical Center, General Hospital of Chinese PLA, Beijing, China
| |
Collapse
|
37
|
Wood N, Straw S, Scalabrin M, Roberts LD, Witte KK, Bowen TS. Skeletal muscle atrophy in heart failure with diabetes: from molecular mechanisms to clinical evidence. ESC Heart Fail 2021; 8:3-15. [PMID: 33225593 PMCID: PMC7835554 DOI: 10.1002/ehf2.13121] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 12/25/2022] Open
Abstract
Two highly prevalent and growing global diseases impacted by skeletal muscle atrophy are chronic heart failure (HF) and type 2 diabetes mellitus (DM). The presence of either condition increases the likelihood of developing the other, with recent studies revealing a large and relatively poorly characterized clinical population of patients with coexistent HF and DM (HFDM). HFDM results in worse symptoms and poorer clinical outcomes compared with DM or HF alone, and cardiovascular-focused disease-modifying agents have proven less effective in HFDM indicating a key role of the periphery. This review combines current clinical knowledge and basic biological mechanisms to address the critical emergence of skeletal muscle atrophy in patients with HFDM as a key driver of symptoms. We discuss how the degree of skeletal muscle wasting in patients with HFDM is likely underpinned by a variety of mechanisms that include mitochondrial dysfunction, insulin resistance, inflammation, and lipotoxicity. Given many atrophic triggers (e.g. ubiquitin proteasome/autophagy/calpain activity and supressed IGF1-Akt-mTORC1 signalling) are linked to increased production of reactive oxygen species, we speculate that a higher pro-oxidative state in HFDM could be a unifying mechanism that promotes accelerated fibre atrophy. Overall, our proposal is that patients with HFDM represent a unique clinical population, prompting a review of treatment strategies including further focus on elucidating potential mechanisms and therapeutic targets of muscle atrophy in these distinct patients.
Collapse
Affiliation(s)
- Nathanael Wood
- Faculty of Biomedical SciencesUniversity of LeedsLeedsLS2 9JTUK
| | - Sam Straw
- Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | | | - Lee D. Roberts
- Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Klaus K. Witte
- Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | | |
Collapse
|
38
|
Goto K, Schauer A, Augstein A, Methawasin M, Granzier H, Halle M, Craenenbroeck EMV, Rolim N, Gielen S, Pieske B, Winzer EB, Linke A, Adams V. Muscular changes in animal models of heart failure with preserved ejection fraction: what comes closest to the patient? ESC Heart Fail 2020; 8:139-150. [PMID: 33350094 PMCID: PMC7835579 DOI: 10.1002/ehf2.13142] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 10/31/2020] [Accepted: 11/15/2020] [Indexed: 12/16/2022] Open
Abstract
AIMS Heart failure with preserved ejection fraction (HFpEF) is associated with reduced exercise capacity elicited by skeletal muscle (SM) alterations. Up to now, no clear medical treatment advice for HFpEF is available. Identification of the ideal animal model mimicking the human condition is a critical step in developing and testing treatment strategies. Several HFpEF animals have been described, but the most suitable in terms of comparability with SM alterations in HFpEF patients is unclear. The aim of the present study was to investigate molecular changes in SM of three different animal models and to compare them with alterations of muscle biopsies obtained from human HFpEF patients. METHODS AND RESULTS Skeletal muscle tissue was obtained from HFpEF and control patients and from three different animal models including the respective controls-ZSF1 rat, Dahl salt-sensitive rat, and transverse aortic constriction surgery/deoxycorticosterone mouse. The development of HFpEF was verified by echocardiography. Protein expression and enzyme activity of selected markers were assessed in SM tissue homogenates. Protein expression between SM tissue obtained from HFpEF patients and the ZSF1 rats revealed similarities for protein markers involved in muscle atrophy (MuRF1 expression, protein ubiquitinylation, and LC3) and mitochondrial metabolism (succinate dehydrogenase and malate dehydrogenase activity, porin expression). The other two animal models exhibited far less similarities to the human samples. CONCLUSIONS None of the three tested animal models mimics the condition in HFpEF patients completely, but among the animal models tested, the ZSF1 rat (ZSF1-lean vs. ZSF1-obese) shows the highest overlap to the human condition. Therefore, when studying therapeutic interventions to treat HFpEF and especially alterations in the SM, we suggest that the ZSF1 rat is a suitable model.
Collapse
Affiliation(s)
- Keita Goto
- Laboratory for Experimental and Molecular Cardiology, Department of Internal Medicine and Cardiology, TU Dresden, Heart Center Dresden University Hospital, Fetscherstrasse 76, Dresden, 01307, Germany
| | - Antje Schauer
- Laboratory for Experimental and Molecular Cardiology, Department of Internal Medicine and Cardiology, TU Dresden, Heart Center Dresden University Hospital, Fetscherstrasse 76, Dresden, 01307, Germany
| | - Antje Augstein
- Laboratory for Experimental and Molecular Cardiology, Department of Internal Medicine and Cardiology, TU Dresden, Heart Center Dresden University Hospital, Fetscherstrasse 76, Dresden, 01307, Germany
| | - Mei Methawasin
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, USA
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, USA
| | - Martin Halle
- Prevention and Sports Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | | | - Natale Rolim
- Department of Circulation and Medical Imaging, Faculty of Medicine, NTNU, K.G. Jebsen Center of Exercise in Medicine, Trondheim, Norway
| | - Stephan Gielen
- Department of Cardiology, Angiology and Intensive Care, Klinikum Lippe, Detmold, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ephraim B Winzer
- Laboratory for Experimental and Molecular Cardiology, Department of Internal Medicine and Cardiology, TU Dresden, Heart Center Dresden University Hospital, Fetscherstrasse 76, Dresden, 01307, Germany
| | - Axel Linke
- Laboratory for Experimental and Molecular Cardiology, Department of Internal Medicine and Cardiology, TU Dresden, Heart Center Dresden University Hospital, Fetscherstrasse 76, Dresden, 01307, Germany.,Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany
| | - Volker Adams
- Laboratory for Experimental and Molecular Cardiology, Department of Internal Medicine and Cardiology, TU Dresden, Heart Center Dresden University Hospital, Fetscherstrasse 76, Dresden, 01307, Germany.,Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany
| |
Collapse
|
39
|
Carbone S, Billingsley HE, Rodriguez-Miguelez P, Kirkman DL, Garten R, Franco RL, Lee DC, Lavie CJ. Lean Mass Abnormalities in Heart Failure: The Role of Sarcopenia, Sarcopenic Obesity, and Cachexia. Curr Probl Cardiol 2020; 45:100417. [PMID: 31036371 PMCID: PMC11146283 DOI: 10.1016/j.cpcardiol.2019.03.006] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 03/24/2019] [Indexed: 02/07/2023]
Abstract
The role of body composition in patients with heart failure (HF) has been receiving much attention in the last few years. Particularly, reduced lean mass (LM), the best surrogate for skeletal muscle mass, is independently associated with abnormal cardiorespiratory fitness (CRF) and muscle strength, ultimately leading to reduced quality of life and worse prognosis. While in the past, reduced CRF in patients with HF was thought to result exclusively from cardiac dysfunction leading to reduced cardiac output at peak exercise, current evidence supports the concept that abnormalities in LM may also play a critical role. Abnormalities in the LM body composition compartment are associated with the development of sarcopenia, sarcopenic obesity, and cachexia. Such conditions have been implicated in the pathophysiology and progression of HF. However, identification of such conditions remains challenging, as universal definitions for sarcopenia, sarcopenic obesity, and cachexia are lacking. In this review article, we describe the most common body composition abnormalities related to the LM compartment, including skeletal and respiratory muscle mass abnormalities, and the consequences of such anomalies on CRF and muscle strength in patients with HF. Finally, we discuss the potential nonpharmacologic therapeutic strategies such as exercise training (ie, aerobic exercise and resistance exercise) and dietary interventions (ie, dietary supplementation and dietary patterns) that have been implemented to target body composition, with a focus on HF.
Collapse
|
40
|
Liu Q, Gao J, Deng J, Xiao J. Current Studies and Future Directions of Exercise Therapy for Muscle Atrophy Induced by Heart Failure. Front Cardiovasc Med 2020; 7:593429. [PMID: 33195482 PMCID: PMC7644508 DOI: 10.3389/fcvm.2020.593429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Muscle atrophy is a common complication of heart failure. At present, there is no specific treatment to reverse the course of muscle atrophy. Exercise training, due to the safety and easy operation, is a recommended therapy for muscle atrophy induced by heart failure. However, the patients with muscle atrophy are weak in mobility and may not be able to train for a long time. Therefore, it is necessary to explore novel targets of exercise protection for muscle atrophy, so as to improve the quality of life and survival rate of patients with muscular atrophy induced by heart failure. This article aims to review latest studies, summarize the evidence and limitations, and provide a glimpse into the future of exercise for the treatment of muscle atrophy induced by heart failure. We wish to highlight some important findings about the essential roles of exercise sensors in muscle atrophy induced by heart failure, which might be helpful for searching potential therapeutic targets for muscle wasting induced by heart failure.
Collapse
Affiliation(s)
- Qi Liu
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Juan Gao
- School of Medicine, Shanghai University, Shanghai, China
| | - Jiali Deng
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China.,School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
41
|
Exercise as a Therapeutic Strategy for Sarcopenia in Heart Failure: Insights into Underlying Mechanisms. Cells 2020; 9:cells9102284. [PMID: 33066240 PMCID: PMC7602002 DOI: 10.3390/cells9102284] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/06/2020] [Accepted: 10/10/2020] [Indexed: 12/16/2022] Open
Abstract
Sarcopenia, a syndrome commonly seen in elderly populations, is often characterized by a gradual loss of skeletal muscle, leading to the decline of muscle strength and physical performance. Growing evidence suggests that the prevalence of sarcopenia increases in patients with heart failure (HF), which is a dominant pathogenesis in the aging heart. HF causes diverse metabolic complications that may result in sarcopenia. Therefore, sarcopenia may act as a strong predictor of frailty, disability, and mortality associated with HF. Currently, standard treatments for slowing muscle loss in patients with HF are not available. Therefore, here, we review the pathophysiological mechanisms underlying sarcopenia in HF as well as current knowledge regarding the beneficial effects of exercise on sarcopenia in HF and related mechanisms, including hormonal changes, myostatin, oxidative stress, inflammation, apoptosis, autophagy, the ubiquitin-proteasome system, and insulin resistance.
Collapse
|
42
|
Gielen S. Domain-Management der Herzinsuffizienz beim geriatrischen Patienten. AKTUELLE KARDIOLOGIE 2020. [DOI: 10.1055/a-1238-2576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Geriatrische Patienten mit chronischer Herzinsuffizienz (CHI) sind aufgrund von Multimorbität, Einschränkungen der Mobilität und Kognition sowie reduzierter sozialer Netzwerke Hochrisikopatienten. Eine einfache kardiologisch fokussierte medikamentöse/interventionelle Behandlung führt daher oftmals nicht allein zum Ziel. Das vom American College of Cardiology vorgeschlagene System des Domain-Managements bei geriatrischen Patienten mit CHI ergänzt daher sinnvoll die strukturellen Versorgungskonzepte und gliedert die Problembereiche der Patienten in 4 große Blöcke:Durch systematische Bearbeitung der 4 Blöcke reduziert sich das Risiko, prognoserelevante Probleme zu übersehen, erheblich.Insgesamt bietet das Domain-Management einen Ansatz, die Komplexität prognoserelevanter Probleme des geriatrischen Patienten mit CHI zuverlässiger zu erkennen und in Therapieentscheidungen einzubeziehen.
Collapse
Affiliation(s)
- Stephan Gielen
- Klinik für Kardiologie, Angiologie und Internistische Intensivmedizin, Klinikum Lippe GmbH, Detmold, Deutschland
- Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Deutschland
| |
Collapse
|
43
|
Effect of exercise training on cardiovascular autonomic and muscular function in subclinical Chagas cardiomyopathy: a randomized controlled trial. Clin Auton Res 2020; 31:239-251. [PMID: 32875456 DOI: 10.1007/s10286-020-00721-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 08/18/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Patients with chronic chagasic cardiomyopathy with preserved ventricular function present with autonomic imbalance. This study evaluated the effects of exercise training (ET) in restoring peripheral and cardiac autonomic control and skeletal muscle phenotype in patients with subclinical chronic chagasic cardiomyopathy. METHODS This controlled trial (NCT02295215) included 24 chronic chagasic cardiomyopathy patients who were randomized www.random.org/lists/ into two groups: those who underwent exercise training (n = 12) and those who continued their usual activities (n = 12). Eight patients completed the exercise training protocol, and 10 patients were clinically followed up for 4 months. Muscular sympathetic nerve activity was measured by microneurography and muscle blood flow (MBF) using venous occlusion plethysmography. The low-frequency component of heart rate variability in normalized units (LFnuHR) reflects sympathetic activity in the heart, and the low-frequency component of systolic blood pressure variability in normalized units reflects sympathetic activity in the vessels. The infusion of vasoactive drugs (phenylephrine and sodium nitroprusside) was used to evaluate cardiac baroreflex sensitivity, and a vastus lateralis muscle biopsy was performed to evaluate atrogin-1 and MuRF-1 gene expression. RESULTS The baroreflex sensitivity for increases (p = 0.002) and decreases (p = 0.02) in systolic blood pressure increased in the ET group. Muscle blood flow also increased only in the ET group (p = 0.004). Only the ET group had reduced resting muscular sympathetic nerve activity levels (p = 0.008) and sympathetic activity in the heart (LFnu; p = 0.004) and vessels (p = 0.04) after 4 months. Regarding skeletal muscle, after 4 months, participants in the exercise training group presented with lower atrogin-1 gene expression than participants who continued their activities as usual (p = 0.001). The reduction in muscular sympathetic nerve activity was positively associated with reduced atrogin-1 (r = 0.86; p = 0.02) and MuRF-1 gene expression (r = 0.64; p = 0.06); it was negatively associated with improved baroreflex sensitivity both for increases (r = -0.72; p = 0.020) and decreases (r = -0.82; p = 0.001) in blood pressure. CONCLUSIONS ET improved cardiac and peripheral autonomic function in patients with subclinical chagasic cardiomyopathy. ET reduced MSNA and sympathetic activity in the heart and vessels and increased cardiac parasympathetic tone and baroreflex sensitivity. Regarding peripheral muscle, after 4 months, patients who underwent exercise training had an increased cross-sectional area of type I fibers and oxidative metabolism of muscle fibers, and decreased atrogin-1 gene expression, compared to participants who continued their activities as usual. In addition, the reduction in MSNA was associated with improved cardiac baroreflex sensitivity, reduced sympathetic cardiovascular tone, and reduced atrogin-1 and MuRF-1 gene expression. TRIAL REGISTRATION ID: NCT02295215. Registered in June 2013.
Collapse
|
44
|
Duan K, Gao X, Zhu D. The clinical relevance and mechanism of skeletal muscle wasting. Clin Nutr 2020; 40:27-37. [PMID: 32788088 DOI: 10.1016/j.clnu.2020.07.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/15/2020] [Accepted: 07/21/2020] [Indexed: 02/08/2023]
Abstract
Skeletal muscle wasting occurs in both chronic and acute diseases. Increasing evidence has shown this debilitating process is associated with short- and long-term outcomes in critical, cancer and surgical patients. Both muscle quantity and quality, as reflected by the area and density of a given range of attenuation in CT scan, impact the patient prognosis. In addition, ultrasound and bioelectrical impedance analysis (BIA) are also widely used in the assessment of body composition due to their bedside viability and no radioactivity. Mechanism researches have revealed complicated pathways are involved in muscle wasting, which include altered IGF1-Akt-FoxO signaling, elevated levels of myostatin and activin A, activation of NF-κB pathway and glucocorticoid effects. Particularly, central nervous system (CNS) has been proven to participate in regulating muscle wasting in various conditions, such as infection and tumor. Several promising therapeutic agents have been under developing in the treatment of muscle atrophy, such as myostatin antagonist, ghrelin analog, non-steroidal selective androgen receptor modulators (SARMs). Notably, nutritional therapy is still the fundamental support in combating muscle wasting. However, the optimizing and tailored nutrition regimen relies on accurate metabolism measurement and large clinical trials in the future. Here, we will discuss the current understanding of muscle wasting and potential treatment in clinical practice.
Collapse
Affiliation(s)
- Kaipeng Duan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| | - Xin Gao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| | - Dongming Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China.
| |
Collapse
|
45
|
Lu F, Lu B, Zhang L, Wen J, Wang M, Zhang S, Li Q, Shu F, Sun Y, Liu N, Peng S, Zhao Y, Dong S, Zhao D, Lu F, Zhang W. Hydrogen sulphide ameliorating skeletal muscle atrophy in db/db mice via Muscle RING finger 1 S-sulfhydration. J Cell Mol Med 2020; 24:9362-9377. [PMID: 32633463 PMCID: PMC7417732 DOI: 10.1111/jcmm.15587] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 12/25/2022] Open
Abstract
Muscle atrophy occurs in many pathological states, including cancer, diabetes and sepsis, whose results primarily from accelerated protein degradation and activation of the ubiquitin‐proteasome pathway. Expression of Muscle RING finger 1 (MuRF1), an E3 ubiquitin ligase, was increased to induce the loss of muscle mass in diabetic condition. However, hydrogen sulphide (H2S) plays a crucial role in the variety of physiological functions, including antihypertension, antiproliferation and antioxidant. In this study, db/db mice and C2C12 myoblasts treated by high glucose and palmitate and oleate were chose as animal and cellular models. We explored how exogenous H2S attenuated the degradation of skeletal muscle via the modification of MuRF1 S‐sulfhydration in db/db mice. Our results show cystathionine‐r‐lyase expression, and H2S level in skeletal muscle of db/db mice was reduced. Simultaneously, exogenous H2S could alleviate ROS production and reverse expression of ER stress protein markers. Exogenous H2S could decrease the ubiquitination level of MYOM1 and MYH4 in db/db mice. In addition, exogenous H2S reduced the interaction between MuRF1 with MYOM1 and MYH4 via MuRF1 S‐sulfhydration. Based on these results, we establish that H2S prevented the degradation of skeletal muscle via MuRF1 S‐sulfhydration at the site of Cys44 in db/db mice.
Collapse
Affiliation(s)
- Fangping Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Baoling Lu
- Department of Infectious, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Linxue Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - JingChen Wen
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Mengyi Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shiwu Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Qianzhu Li
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Feng Shu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yu Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Ning Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shuo Peng
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yajun Zhao
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shiyun Dong
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Dechao Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fanghao Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| |
Collapse
|
46
|
Scalabrin M, Adams V, Labeit S, Bowen TS. Emerging Strategies Targeting Catabolic Muscle Stress Relief. Int J Mol Sci 2020; 21:E4681. [PMID: 32630118 PMCID: PMC7369951 DOI: 10.3390/ijms21134681] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle wasting represents a common trait in many conditions, including aging, cancer, heart failure, immobilization, and critical illness. Loss of muscle mass leads to impaired functional mobility and severely impedes the quality of life. At present, exercise training remains the only proven treatment for muscle atrophy, yet many patients are too ill, frail, bedridden, or neurologically impaired to perform physical exertion. The development of novel therapeutic strategies that can be applied to an in vivo context and attenuate secondary myopathies represents an unmet medical need. This review discusses recent progress in understanding the molecular pathways involved in regulating skeletal muscle wasting with a focus on pro-catabolic factors, in particular, the ubiquitin-proteasome system and its activating muscle-specific E3 ligase RING-finger protein 1 (MuRF1). Mechanistic progress has provided the opportunity to design experimental therapeutic concepts that may affect the ubiquitin-proteasome system and prevent subsequent muscle wasting, with novel advances made in regards to nutritional supplements, nuclear factor kappa-light-chain-enhancer of activated B cells (NFB) inhibitors, myostatin antibodies, β2 adrenergic agonists, and small-molecules interfering with MuRF1, which all emerge as a novel in vivo treatment strategies for muscle wasting.
Collapse
Affiliation(s)
- Mattia Scalabrin
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK;
| | - Volker Adams
- Department of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany;
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, 01067 Dresden, Germany
| | - Siegfried Labeit
- Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany;
- Myomedix GmbH, Im Biengarten 36, 69151 Neckargemünd, Germany
| | - T. Scott Bowen
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK;
| |
Collapse
|
47
|
|
48
|
Zhang JR, Li XX, Hu WN, Li CY. Emerging Role of TRIM Family Proteins in Cardiovascular Disease. Cardiology 2020; 145:390-400. [PMID: 32305978 DOI: 10.1159/000506150] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/23/2020] [Indexed: 11/19/2022]
Abstract
Ubiquitination is one of the basic mechanisms of cell protein homeostasis and degradation and is accomplished by 3 enzymes, E1, E2, and E3. Tripartite motif-containing proteins (TRIMs) constitute the largest subfamily of RING E3 ligases, with >70 current members in humans and mice. These members are involved in multiple biological processes, including growth, differentiation, and apoptosis as well as disease and tumorigenesis. Accumulating evidence has shown that many TRIM proteins are associated with various cardiac processes and pathologies, such as heart development, signal transduction, protein degradation, autophagy mediation, ion channel regulation, congenital heart disease, and cardiomyopathies. In this review, we provide an overview of the TRIM family and discuss its involvement in the regulation of cardiac proteostasis and pathophysiology and its potential therapeutic implications.
Collapse
Affiliation(s)
- Jing-Rui Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xin-Xin Li
- Department of Respiratory Medicine, Tangshan People's Hospital, Tangshan, China
| | - Wan-Ning Hu
- Department of Cardiology, Laboratory of Molecular Biology, Tangshan Gongren Hospital, Tangshan, China,
| | - Chang-Yi Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Department of Cardiology, Laboratory of Molecular Biology, Tangshan Gongren Hospital, Tangshan, China
| |
Collapse
|
49
|
Sun D, Zhang F, Ma T, Zhang Y, Liang Z. Atorvastatin alleviates left ventricular remodeling in isoproterenol-induced chronic heart failure in rats by regulating the RhoA/Rho kinase signaling pathway. Pharmacol Rep 2020; 72:903-911. [PMID: 32144744 DOI: 10.1007/s43440-020-00085-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 02/13/2020] [Accepted: 02/21/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND Chronic heart failure (CHF) is characterized by left ventricular dysfunction and altered autonomic control of cardiac function. This study aimed to investigate the effects of atorvastatin on left ventricular remodeling (LVR) and cardiac function in rats with isoproterenol-induced CHF and the possible mechanism. METHODS An isoproterenol-induced CHF model was established in rata, which were subsequently treated with atorvastatin. Echocardiography, hemodynamic, and left ventricular mass indexes were assessed. The mRNA expression of RhoA, Rho kinase, and endothelial nitric oxide synthase (eNOS) was determined by RT-qPCR. The protein expression of myosin-binding subunit (MBS), MBS-P, eNOS, phosphorylated-eNOS, RhoA, and Rho kinase was measured by Western blot analysis. The relative activity of NADPH oxidase, ROS, and NO was assessed by ELISA. RESULTS Isoproterenol-induced CHF rats treated with atorvastatin exhibited decreased left ventricular end-systolic dimension, left ventricular end-diastolic dimension, left ventricular end-diastolic pressure, left ventricular mass index, maximum fall rate of change in left ventricular pressure, heart rate (p < 0.001), expression of RhoA, Rho kinase, MBS and MBS-P (p < 0.01), and relative activity of NADPH oxidase, ROS and NO (p < 0.05) and increased left ventricular short axis fractional shortening, left ventricular end-systolic pressure, maximum rise rate of change in left ventricular pressure (p < 0.001) and expression of eNOS, and phosphorylated-eNOS ser1177 (all p < 0.05) compared with those of rats with isoproterenol-induced CHF. CONCLUSION We demonstrated that atorvastatin inhibits LVR and improves cardiac function in rats with isoproterenol-induced CHF through inhibition of the RhoA/Rho kinase signaling pathway.
Collapse
Affiliation(s)
- Dingjun Sun
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital (Haikou People's Hospital), Haikou, 570208, People's Republic of China
| | - Fuwei Zhang
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital (Haikou People's Hospital), Haikou, 570208, People's Republic of China
| | - Tianyi Ma
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital (Haikou People's Hospital), Haikou, 570208, People's Republic of China
| | - Yixue Zhang
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital (Haikou People's Hospital), Haikou, 570208, People's Republic of China
| | - Zhongshu Liang
- Department of Cardiology, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, People's Republic of China.
| |
Collapse
|
50
|
Duraes AR, da Silva WAP, Filho CRH, de Souza Lima Bitar Y, Neto MG. Heart Failure and Comorbidities—Part 2. CURRENT EMERGENCY AND HOSPITAL MEDICINE REPORTS 2020. [DOI: 10.1007/s40138-020-00211-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|