1
|
Kaneguchi A, Sakitani N, Umehara T. Histological changes in skeletal muscle induced by heart failure in human patients and animal models: A scoping review. Acta Histochem 2024; 126:152210. [PMID: 39442432 DOI: 10.1016/j.acthis.2024.152210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE This scoping review aimed to characterize the histological changes in skeletal muscle after heart failure (HF) and to identify gaps in knowledge. METHODS On April 03, 2024, systematic searches were performed for papers in which histological analyses were conducted on skeletal muscle sampled from patients with HF or animal models of HF. Screening and data extraction were conducted by two independent authors. RESULTS AND CONCLUSION A total of 118 papers were selected, including 33 human and 85 animal studies. Despite some disagreements among studies, some trends were observed. These trends included a slow-to-fast transition, a decrease in muscle fiber size, capillary to muscle fiber ratio, and mitochondrial activity and content, and an increase in apoptosis. These changes may contribute to the fatigability and decrease in muscle strength observed after HF. Although there were some disagreements between the results of human and animal studies, the results were generally similar. Animal models of HF will therefore be useful in elucidating the histological changes in skeletal muscle that occur in human patients with HF. Because the muscles subjected to histological analysis were mostly thigh muscles in humans and mostly lower leg muscles in animals, it remains uncertain whether changes similar to those seen in lower limb (hindlimb) muscles after HF also occur in upper limb (forelimb) muscles. The results of this review will consolidate the current knowledge on HF-induced histological changes in skeletal muscle and consequently aid in the rehabilitation of patients with HF and future studies.
Collapse
Affiliation(s)
- Akinori Kaneguchi
- Department of Rehabilitation, Faculty of Rehabilitation, Hiroshima International University, Kurose-Gakuendai 555-36, Higashi-Hiroshima, Hiroshima, 739-2695, Japan.
| | - Naoyoshi Sakitani
- Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Hayashi-cho 2217-4, Takamatsu, Kagawa, 761-0395, Japan
| | - Takuya Umehara
- Department of Rehabilitation, Faculty of Rehabilitation, Hiroshima International University, Kurose-Gakuendai 555-36, Higashi-Hiroshima, Hiroshima, 739-2695, Japan
| |
Collapse
|
2
|
Chaudhari M, Zelko I, Lorkiewicz P, Hoetker D, Nong Y, Doelling B, Brittian K, Bhatnagar A, Srivastava S, Baba SP. Metabolic pathways for removing reactive aldehydes are diminished in the skeletal muscle during heart failure. Skelet Muscle 2024; 14:24. [PMID: 39425168 PMCID: PMC11488087 DOI: 10.1186/s13395-024-00354-2] [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: 11/16/2023] [Accepted: 09/25/2024] [Indexed: 10/21/2024] Open
Abstract
Muscle wasting is a serious complication in heart failure patients. Oxidative stress and inflammation are implicated in the pathogenesis of muscle wasting. Oxidative stress leads to the formation of toxic lipid peroxidation products, such as 4-hydroxy-2-nonenal (HNE), which covalently bind with proteins and DNA and activate atrophic pathways. Whether the formation of lipid peroxidation products and metabolic pathways that remove these toxic products are affected during heart failure-associated skeletal muscle wasting has never been studied. Male C57BL/6J mice were subjected to sham and transverse aortic constriction (TAC) surgeries for 4, 8 or 14 weeks. Different skeletal muscle beds were weighed, and the total cross-sectional area of the gastrocnemius muscle was measured via immunohistochemistry. Muscle function and muscle stiffness were measured by a grip strength meter and atomic force microscope, respectively. Atrophic and inflammatory marker levels were measured via qRT‒PCR. The levels of acrolein and HNE-protein adducts, aldehyde-removing enzymes, the histidyl dipeptide-synthesizing enzyme carnosine synthase (CARNS), and amino acid transporters in the gastrocnemius muscle were measured via Western blotting and qRT‒PCR. Histidyl dipeptides and histidyl dipeptide aldehyde conjugates in the Gastrocnemius and soleus muscles were analyzed by LC/MS-MS. Body weight, gastrocnemius muscle and soleus muscle weights and the total cross-sectional area of the gastrocnemius muscle were decreased after 14 weeks of TAC. Heart weight, cardiac function, grip strength and muscle stiffness were decreased in the TAC-operated mice. Expression of the atrophic and inflammatory markers Atrogin1 and TNF-α, respectively, was increased ~ 1.5-2fold in the gastrocnemius muscle after 14 weeks of TAC (p < 0.05 and p = 0.004 vs sham). The formation of HNE and acrolein protein adducts was increased, and the expression of the aldehyde-removing enzyme aldehyde dehydrogenase (ALDH2) was decreased in the gastrocnemius muscle of TAC mice. Carnosine (sham: 5.76 ± 1.3 vs TAC: 4.72 ± 0.7 nmol/mg tissue, p = 0.04) and total histidyl dipeptide levels (carnosine and anserine; sham: 11.97 ± 1.5 vs TAC: 10.13 ± 1.4 nmol/mg tissue, p < 0.05) were decreased in the gastrocnemius muscle of TAC mice. Depletion of histidyl dipeptides diminished the aldehyde removal capacity of the atrophic gastrocnemius muscle. Furthermore, CARNS and TAUT protein expression were decreased in the atrophic gastrocnemius muscle. Our data reveals that reduced expression of ALDH2 and depletion of histidyl dipeptides in the gastrocnemius muscle during heart failure leads to the accumulation of toxic aldehydes and might contribute to muscle wasting.
Collapse
Affiliation(s)
- Mamata Chaudhari
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - Igor Zelko
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - Pawel Lorkiewicz
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - David Hoetker
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - Yibing Nong
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - Benjamin Doelling
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - Kenneth Brittian
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - Aruni Bhatnagar
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | | | - Shahid P Baba
- Center for Cardiometabolic Science, Louisville, KY, USA.
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA.
| |
Collapse
|
3
|
Lord SO, Dawson PW, Chunthorng-Orn J, Ng J, Baehr LM, Hughes DC, Sridhar P, Knowles T, Bodine SC, Lai YC. Uncovering the mechanisms of MuRF1-induced ubiquitylation and revealing similarities with MuRF2 and MuRF3. Biochem Biophys Rep 2024; 37:101636. [PMID: 38283190 PMCID: PMC10818185 DOI: 10.1016/j.bbrep.2023.101636] [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: 11/06/2023] [Revised: 12/12/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
MuRF1 (Muscle-specific RING finger protein 1; gene name TRIM63) is a ubiquitin E3 ligase, associated with the progression of muscle atrophy. As a RING (Really Interesting New Gene) type E3 ligase, its unique activity of ubiquitylation is driven by a specific interaction with a UBE2 (ubiquitin conjugating enzyme). Our understanding of MuRF1 function remains unclear as candidate UBE2s have not been fully elucidated. In the present study, we screened human ubiquitin dependent UBE2s in vitro and found that MuRF1 engages in ubiquitylation with UBE2D, UBE2E, UBE2N/V families and UBE2W. MuRF1 can cause mono-ubiquitylation, K48- and K63-linked polyubiquitin chains in a UBE2 dependent manner. Moreover, we identified a two-step UBE2 dependent mechanism whereby MuRF1 is monoubiquitylated by UBE2W which acts as an anchor for UBE2N/V to generate polyubiquitin chains. With the in vitro ubiquitylation assay, we also found that MuRF2 and MuRF3 not only share the same UBE2 partners as MuRF1 but can also directly ubiquitylate the same substrates: Titin (A168-A170), Desmin, and MYLPF (Myosin Light Chain, Phosphorylatable, Fast Skeletal Muscle; also called Myosin Light Regulatory Chain 2). In summary, our work presents new insights into the mechanisms that underpin MuRF1 activity and reveals overlap in MuRF-induced ubiquitylation which could explain their partial redundancy in vivo.
Collapse
Affiliation(s)
- Samuel O. Lord
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Peter W.J. Dawson
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| | | | - Jimi Ng
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Leslie M. Baehr
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - David C. Hughes
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Pooja Sridhar
- School of Biosciences, University of Birmingham, Birmingham, UK
| | - Timothy Knowles
- School of Biosciences, University of Birmingham, Birmingham, UK
| | - Sue C. Bodine
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Yu-Chiang Lai
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Janini Gomes M, Sabela AK, Ferreira ITP, de Souza SLB, Mota GAF, da Silva VL, de Campos DHS, Lima ARR, Carvalho MR, Bazan SGZ, Corrêa CR, Cicogna AC, Okoshi MP, Pacagnelli FL. Effects of aerobic exercise on cardiac function and gene expression of NADPH oxidases in diaphragm muscle of rats with aortic stenosis-induced heart failure. Front Physiol 2023; 14:1182303. [PMID: 37362442 PMCID: PMC10285051 DOI: 10.3389/fphys.2023.1182303] [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: 03/08/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
We evaluated the influence of aerobic physical exercise (EX) on gene-encoding proteins associated with oxidative stress in diaphragm muscle of rats with aortic stenosis-induced heart failure (HF). Wistar male rats were divided into four groups: Control sedentary (C); Control exercise (C-Ex); Sedentary aortic stenosis (AS); Aortic stenosis exercise (AS-Ex). Exercised rats trained 5 times a week for 10 weeks on a treadmill. Statistical analysis was performed by ANOVA or Kruskal-Wallis test. In the final echocardiogram, animals with aortic stenosis subjected to exercise demonstrated improvement in systolic function compared to the sedentary aortic stenosis group. In diaphragm muscle, the activity of antioxidant enzymes, malondialdehyde malondialdehyde concentration, protein carbonylation, and protein expression of p65 and its inhibitor IκB did not differ between groups. Alterations in gene expression of sources that generate reactive species of oxygen were observed in AS-Ex group, which showed decreased mRNA abundance of NOX2 and NOX4 compared to the aortic stenosis group (p < 0.05). We concluded that aerobic exercise has a positive impact during heart failure, ameliorating systolic dysfunction and biomarkers of oxidative stress in diaphragm muscle of rats with aortic stenosis-induced heart failure.
Collapse
Affiliation(s)
- Mariana Janini Gomes
- Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX, United States
| | - Ana Karenina Sabela
- Physiotherapy Department, UNOESTE, Presidente Prudente, Brazil
- Post-graduate Program, Animal Science, UNOESTE, Presidente Prudente, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | - Francis Lopes Pacagnelli
- Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX, United States
- Physiotherapy Department, UNOESTE, Presidente Prudente, Brazil
| |
Collapse
|
7
|
Liu Q, Chen L, Liang X, Cao Y, Zhu X, Wang S, Li J, Gao J, Xiao J. Exercise attenuates angiotensinⅡ-induced muscle atrophy by targeting PPARγ/miR-29b. JOURNAL OF SPORT AND HEALTH SCIENCE 2022; 11:696-707. [PMID: 34116237 PMCID: PMC9729927 DOI: 10.1016/j.jshs.2021.06.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/11/2021] [Accepted: 05/07/2021] [Indexed: 05/10/2023]
Abstract
BACKGROUND Exercise is beneficial for muscle atrophy. Peroxisome proliferator-activated receptor gamma (PPARγ) and microRNA-29b (miR-29b) have been reported to be responsible for angiotensinⅡ (AngⅡ)-induced muscle atrophy. However, it is unclear whether exercise can protect AngⅡ-induced muscle atrophy by targeting PPARγ/miR-29b. METHODS Skeletal muscle atrophy in both the control group and the run group was established by AngⅡ infusion; after 1 week of exercise training, the mice were sacrificed, and muscle weight was determined. Myofiber size was measured by hematoxylin-eosin and wheat-germ agglutinin staining. Apoptosis was evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling staining. The expression level of muscle atrogenes, including F-box only protein 32 (FBXO32, also called Atrogin-1) and muscle-specific RING-finger 1 (MuRF-1), the phosphorylation level of protein kinase B (PKB, also called AKT)/forkhead box O3A (FOXO3A)/mammalian target of rapamycin (mTOR) pathway proteins, the expression level of PPARγ and apoptosis-related proteins, including B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X (Bax), cysteine-aspartic acid protease 3 (caspase-3), and cleaved-caspase-3, were determined by western blot. The expression level of miR-29b was checked by reverse-transcription quantitative polymerase chain reaction. A PPARγ inhibitor (T0070907) or adeno-associated virus serotype-8 (AAV8)-mediated miR-29b overexpression was used to demonstrate whether PPARγ activation or miR-29b inhibition mediates the beneficial effects of exercise in AngⅡ-induced muscle atrophy. RESULTS Exercise can significantly attenuate AngⅡ-induced muscle atrophy, which is demonstrated by increased skeletal muscle weight, cross-sectional area of myofiber, and activation of AKT/mTOR signaling and by decreased atrogenes expressions and apoptosis. In AngⅡ-induced muscle atrophy mice models, PPARγ was elevated whereas miR-29b was decreased by exercise. The protective effects of exercise in AngⅡ-induced muscle atrophy were inhibited by a PPARγ inhibitor (T0070907) or adeno-associated virus serotype-8 (AAV8)-mediated miR-29b overexpression. CONCLUSION Exercise attenuates AngⅡ-induced muscle atrophy by activation of PPARγ and suppression of miR-29b.
Collapse
Affiliation(s)
- Qi Liu
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Liyang Chen
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xuchun Liang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Yuqing Cao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xinyue Zhu
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Siqi Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Jin Li
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Juan Gao
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China; Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
8
|
Molecular and cellular effects of gold nanoparticles treatment in experimental diabetic myopathy. Heliyon 2022; 8:e10358. [PMID: 36060470 PMCID: PMC9437799 DOI: 10.1016/j.heliyon.2022.e10358] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/28/2022] [Accepted: 08/15/2022] [Indexed: 12/25/2022] Open
Abstract
Background This study aims to address the effects of gold nanoparticles (AuNPs) on diabetic myopathy in streptozotocin (STZ)-induced diabetic rats. Materials and methods Adult male rats were separated into three groups (n = 15): non-diabetic control (ND), diabetic (D), and diabetic treated with AuNPs (2.5 mg/kg, D + AuNPs) intraperitoneally for 4 weeks. A single injection of 50 mg/kg STZ was used to induce diabetes. Results Treatment with AuNPs lowered blood glucose levels. Skeletal muscle mRNA expression of two muscle-specific E3 ubiquitin-ligases enzymes, F-box-only protein 32 (FBXO32) and muscle RING-finger protein-1 (MuRF1) were upregulated in the D group. Diabetic rats showed significant increases in the skeletal muscle expression levels of plasminogen activator inhibitor-1 (PAI-1), tumor necrosis factor-α (TNF-α), transforming growth factor-β1 (TGF-β1), and a decrease in glucose transporter 4 (GLUT4) expression. Superoxide dismutase (SOD) activity decreased and malondialdehyde (MDA) level increased in skeletal muscles of D group. Compared to the D group, expression levels of FBXO32, MuRF1, PAI-1 TNF-α, and TGF-β1 were decreased in the D + AuNPs group, and mRNA of GLUT4 increased. Furthermore, in D + AuNPs group, skeletal muscle MDA levels decreased while SOD activity increased. Conclusion In experimental models, AuNPs can ameliorate muscle atrophy by reducing hyperglycemia, inflammation, and oxidative stress, and by suppressing the ubiquitin-proteasome proteolytic process.
Collapse
|
9
|
Growth differentiation factor 11 induces skeletal muscle atrophy via a STAT3-dependent mechanism in pulmonary arterial hypertension. Skelet Muscle 2022; 12:10. [PMID: 35524286 PMCID: PMC9074369 DOI: 10.1186/s13395-022-00292-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/05/2022] [Indexed: 11/10/2022] Open
Abstract
Skeletal muscle wasting is a clinically remarkable phenotypic feature of pulmonary arterial hypertension (PAH) that increases the risk of mortality. Growth differentiation factor 11 (GDF11), centrally involved in PAH pathogenesis, has an inhibitory effect on skeletal muscle growth in other conditions. However, whether GDF11 is involved in the pathogenesis of skeletal muscle wasting in PAH remains unknown. We showed that serum GDF11 levels in patients were increased following PAH. Skeletal muscle wasting in the MCT-treated PAH model is accompanied by an increase in circulating GDF11 levels and local catabolic markers (Fbx32, Trim63, Foxo1, and protease activity). In vitro GDF11 activated phosphorylation of STAT3. Antagonizing STAT3, with Stattic, in vitro and in vivo, could partially reverse proteolytic pathways including STAT3/socs3 and iNOS/NO in GDF11-meditated muscle wasting. Our findings demonstrate that GDF11 contributes to muscle wasting and the inhibition of its downstream molecule STAT3 shows promise as a therapeutic intervention by which muscle atrophy may be directly prevented in PAH.
Collapse
|
10
|
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: 0.8] [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
|
11
|
Deminice R, Hyatt H, Yoshihara T, Ozdemir M, Nguyen B, Levine S, Powers S. Human and Rodent Skeletal Muscles Express Angiotensin II Type 1 Receptors. Cells 2020; 9:cells9071688. [PMID: 32674346 PMCID: PMC7407103 DOI: 10.3390/cells9071688] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/02/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023] Open
Abstract
Abundant evidence reveals that activation of the renin-angiotensin system promotes skeletal muscle atrophy in several conditions including congestive heart failure, chronic kidney disease, and prolonged mechanical ventilation. However, controversy exists about whether circulating angiotensin II (AngII) promotes skeletal muscle atrophy by direct or indirect effects; the centerpiece of this debate is the issue of whether skeletal muscle fibers express AngII type 1 receptors (AT1Rs). While some investigators assert that skeletal muscle expresses AT1Rs, others argue that skeletal muscle fibers do not contain AT1Rs. These discordant findings in the literature are likely the result of study design flaws and additional research using a rigorous experimental approach is required to resolve this issue. We tested the hypothesis that AT1Rs are expressed in both human and rat skeletal muscle fibers. Our premise was tested using a rigorous, multi-technique experimental design. First, we established both the location and abundance of AT1Rs on human and rat skeletal muscle fibers by means of an AngII ligand-binding assay. Second, using a new and highly selective AT1R antibody, we carried out Western blotting and determined the abundance of AT1R protein within isolated single muscle fibers from humans and rats. Finally, we confirmed the presence of AT1R mRNA in isolated single muscle fibers from rats. Our results support the hypothesis that AT1Rs are present in both human and rat skeletal muscle fibers. Moreover, our experiments provide the first evidence that AT1Rs are more abundant in fast, type II muscle fibers as compared with slow, type I fibers. Together, these discoveries provide the foundation for an improved understanding of the mechanism(s) responsible for AngII-induced skeletal muscle atrophy.
Collapse
Affiliation(s)
- Rafael Deminice
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32608 USA; (R.D.); (T.Y.); (M.O.); (B.N.); (S.P.)
- Department of Physical Education, State University of Londrina, Londrina 860570-970, Brazil
| | - Hayden Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32608 USA; (R.D.); (T.Y.); (M.O.); (B.N.); (S.P.)
- Correspondence: ; Tel.: +1-352-294-1713; Fax: +1-352-392-0316
| | - Toshinori Yoshihara
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32608 USA; (R.D.); (T.Y.); (M.O.); (B.N.); (S.P.)
- Department of Exercise Physiology, Juntendo University, Chiba 270-1695, Japan
| | - Mustafa Ozdemir
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32608 USA; (R.D.); (T.Y.); (M.O.); (B.N.); (S.P.)
| | - Branden Nguyen
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32608 USA; (R.D.); (T.Y.); (M.O.); (B.N.); (S.P.)
| | - Sanford Levine
- Department of Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Scott Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32608 USA; (R.D.); (T.Y.); (M.O.); (B.N.); (S.P.)
| |
Collapse
|
12
|
Adams V, Bowen TS, Werner S, Barthel P, Amberger C, Konzer A, Graumann J, Sehr P, Lewis J, Provaznik J, Benes V, Büttner P, Gasch A, Mangner N, Witt CC, Labeit D, Linke A, Labeit S. Small-molecule-mediated chemical knock-down of MuRF1/MuRF2 and attenuation of diaphragm dysfunction in chronic heart failure. J Cachexia Sarcopenia Muscle 2019; 10:1102-1115. [PMID: 31140761 PMCID: PMC6818456 DOI: 10.1002/jcsm.12448] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Chronic heart failure (CHF) leads to diaphragm myopathy that significantly impairs quality of life and worsens prognosis. In this study, we aimed to assess the efficacy of a recently discovered small-molecule inhibitor of MuRF1 in treating CHF-induced diaphragm myopathy and loss of contractile function. METHODS Myocardial infarction was induced in mice by ligation of the left anterior descending coronary artery. Sham-operated animals (sham) served as controls. One week post-left anterior descending coronary artery ligation animals were randomized into two groups-one group was fed control rodent chow, whereas the other group was fed a diet containing 0.1% of the compound ID#704946-a recently described MuRF1-interfering small molecule. Echocardiography confirmed development of CHF after 10 weeks. Functional and molecular analysis of the diaphragm was subsequently performed. RESULTS Chronic heart failure induced diaphragm fibre atrophy and contractile dysfunction by ~20%, as well as decreased activity of enzymes involved in mitochondrial energy production (P < 0.05). Treatment with compound ID#704946 in CHF mice had beneficial effects on the diaphragm: contractile function was protected, while mitochondrial enzyme activity and up-regulation of the MuRF1 and MuRF2 was attenuated after infarct. CONCLUSIONS Our murine CHF model presented with diaphragm fibre atrophy, impaired contractile function, and reduced mitochondrial enzyme activities. Compound ID#704946 rescued from this partially, possibly by targeting MuRF1/MuRF2. However, at this stage of our study, we refrain to claim specific mechanism(s) and targets of compound ID#704946, because the nature of changes after 12 weeks of feeding is likely to be complex and is not necessarily caused by direct mechanistic effects.
Collapse
Affiliation(s)
- Volker Adams
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, Dresden, Germany
| | - T Scott Bowen
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Sarah Werner
- University Clinic of Cardiology, Heart Center Leipzig, Leipzig, Germany
| | - Peggy Barthel
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, Dresden, Germany
| | | | - Anne Konzer
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Rhine-Main, Germany
| | - Johannes Graumann
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Rhine-Main, Germany
| | - Peter Sehr
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Joe Lewis
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jan Provaznik
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Vladimir Benes
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Petra Büttner
- University Clinic of Cardiology, Heart Center Leipzig, Leipzig, Germany
| | - Alexander Gasch
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Norman Mangner
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, Dresden, Germany
| | - Christian C Witt
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Dittmar Labeit
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.,Myomedix GmbH, Neckargemünd, Germany
| | - Axel Linke
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, Dresden, Germany
| | - Siegfried Labeit
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.,Myomedix GmbH, Neckargemünd, Germany
| |
Collapse
|
13
|
Dumitru A, Radu BM, Radu M, Cretoiu SM. Muscle Changes During Atrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1088:73-92. [PMID: 30390248 DOI: 10.1007/978-981-13-1435-3_4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Muscle atrophy typically is a direct effect of protein degradation induced by a diversity of pathophysiologic states such as disuse, immobilization, denervation, aging, sepsis, cachexia, glucocorticoid treatment, hereditary muscular disorders, cancer, diabetes and obesity, kidney and heart failure, and others. Muscle atrophy is defined by changes in the muscles, consisting in shrinkage of myofibers, changes in the types of fiber and myosin isoforms, and a net loss of cytoplasm, organelles and overall a protein loss. Although in the literature there are extensive studies in a range of animal models, the paucity of human data is a reality. This chapter is focused on various aspects of muscle wasting and describes the transitions of myofiber types during the progression of muscle atrophy in several pathological states. Clinical conditions associated with muscle atrophy have been grouped based on the fast-to-slow or slow-to-fast fiber-type shifts. We have also summarized the ultrastructural and histochemical features characteristic for muscle atrophy in clinical and experimental models for aging, cancer, diabetes and obesity, and heart failure and arrhythmia.
Collapse
Affiliation(s)
- Adrian Dumitru
- Department of Pathology, Emergency University Hospital, Bucharest, Romania
| | - Beatrice Mihaela Radu
- Faculty of Biology, Department of Anatomy, Animal Physiology and Biophysics, University of Bucharest, Bucharest, Romania.,Life, Environmental and Earth Sciences Division, Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
| | - Mihai Radu
- Department of Life & Environmental Physics, 'Horia Hulubei' National Institute for Physics & Nuclear Engineering, Magurele, Romania
| | - Sanda Maria Cretoiu
- Division of Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.
| |
Collapse
|
14
|
Abstract
Skeletal muscle atrophy is a common side effect of most human diseases. Muscle loss is not only detrimental for the quality of life but it also dramatically impairs physiological processes of the organism and decreases the efficiency of medical treatments. While hypothesized for years, the existence of an atrophying programme common to all pathologies is still incompletely solved despite the discovery of several actors and key regulators of muscle atrophy. More than a decade ago, the discovery of a set of genes, whose expression at the mRNA levels were similarly altered in different catabolic situations, opened the way of a new concept: the presence of atrogenes, i.e. atrophy-related genes. Importantly, the atrogenes are referred as such on the basis of their mRNA content in atrophying muscles, the regulation at the protein level being sometimes more complicate to elucidate. It should be noticed that the atrogenes are markers of atrophy and that their implication as active inducers of atrophy is still an open question for most of them. While the atrogene family has grown over the years, it has mostly been incremented based on data coming from rodent models. Whether the rodent atrogenes are valid for humans still remain to be established. An "atrogene" was originally defined as a gene systematically up- or down-regulated in several catabolic situations. Even if recent works often restrict this notion to the up-regulation of a limited number of proteolytic enzymes, it is important to keep in mind the big picture view. In this review, we provide an update of the validated and potential rodent atrogenes and the metabolic pathways they belong, and based on recent work, their relevance in human physio-pathological situations. We also propose a more precise definition of the atrogenes that integrates rapid recovery when catabolic stimuli are stopped or replaced by anabolic ones.
Collapse
Affiliation(s)
- Daniel Taillandier
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000, Clermont-Ferrand, France.
| | - Cécile Polge
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000, Clermont-Ferrand, France
| |
Collapse
|
15
|
Wang X, Zhao H, Ni J, Pan J, Hua H, Wang Y. Identification of suitable reference genes for gene expression studies in rat skeletal muscle following sciatic nerve crush injury. Mol Med Rep 2019; 19:4377-4387. [PMID: 30942461 PMCID: PMC6472138 DOI: 10.3892/mmr.2019.10102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 02/28/2019] [Indexed: 12/14/2022] Open
Abstract
Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) is a molecular biological method used to assess gene expression characterized by high simplicity, effectiveness, specificity and sensitivity. The selection of a suitable reference gene for normalization is critical for the accuracy of quantitative results. Peripheral nerve injury is a common clinical disorder that affects multiple tissues and organs, including peripheral nerves, neurons and the innervated muscles. Numerous genes are differentially expressed in skeletal muscles during muscle denervation and reinnervation following peripheral nerve injury. The identification of a suitable reference gene in innervated muscles following nerve injury may improve the understanding of the alterations in gene expression in the processes of peripheral nerve repair and regeneration. Therefore, in the present study, by using a rat sciatic nerve crush model, the expression levels of various housekeeping genes were examined. In particular, the expression levels of 13 housekeeping genes, including 18S ribosomal RNA, actin β, ankyrin repeat domain 27, cyclophilin A, GAPDH, hypoxanthine phosphoribosyltransferase 1 (HPRT1), mitochondrial ribosomal protein L10, phosphoglycerate kinase 1, RPTOR independent companion of mammalian target of rapamycin complex 2, TATA-box binding protein, ubiquitin C, UBX domain protein 11 and tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein ζ, were investigated in gastrocnemius muscles. The geNorm and NormFinder analyses suggested that the expression level of HPRT1 was particularly stable in gastrocnemius muscles following rat sciatic nerve crush injury. Therefore, HPRT1 may be used as a reference gene for the normalization of gene expression data generated by RT-qPCR.
Collapse
Affiliation(s)
- Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‑innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Hualong Zhao
- Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, Yancheng Teachers' University, Yancheng, Jiangsu 224051, P.R. China
| | - Jun Ni
- Department of Rehabilitation, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jiacheng Pan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‑innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Hao Hua
- Department of Medicine, Xinglin College, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yaxian Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‑innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
16
|
Garfield BE, Crosby A, Shao D, Yang P, Read C, Sawiak S, Moore S, Parfitt L, Harries C, Rice M, Paul R, Ormiston ML, Morrell NW, Polkey MI, Wort SJ, Kemp PR. Growth/differentiation factor 15 causes TGFβ-activated kinase 1-dependent muscle atrophy in pulmonary arterial hypertension. Thorax 2019; 74:164-176. [PMID: 30554141 PMCID: PMC6467240 DOI: 10.1136/thoraxjnl-2017-211440] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 09/24/2018] [Accepted: 10/01/2018] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Skeletal muscle dysfunction is a clinically important complication of pulmonary arterial hypertension (PAH). Growth/differentiation factor 15 (GDF-15), a prognostic marker in PAH, has been associated with muscle loss in other conditions. We aimed to define the associations of GDF-15 and muscle wasting in PAH, to assess its utility as a biomarker of muscle loss and to investigate its downstream signalling pathway as a therapeutic target. METHODS GDF-15 levels and measures of muscle size and strength were analysed in the monocrotaline (MCT) rat, Sugen/hypoxia mouse and in 30 patients with PAH. In C2C12 myotubes the downstream targets of GDF-15 were identified. The pathway elucidated was then antagonised in vivo. RESULTS Circulating GDF-15 levels correlated with tibialis anterior (TA) muscle fibre diameter in the MCT rat (Pearson r=-0.61, p=0.003). In patients with PAH, plasma GDF-15 levels of <564 pg/L predicted those with preserved muscle strength with a sensitivity and specificity of ≥80%. In vitro GDF-15 stimulated an increase in phosphorylation of TGFβ-activated kinase 1 (TAK1). Antagonising TAK1, with 5(Z)-7-oxozeaenol, in vitro and in vivo led to an increase in fibre diameter and a reduction in mRNA expression of atrogin-1 in both C2C12 cells and in the TA of animals who continued to grow. Circulating GDF-15 levels were also reduced in those animals which responded to treatment. CONCLUSIONS Circulating GDF-15 is a biomarker of muscle loss in PAH that is responsive to treatment. TAK1 inhibition shows promise as a method by which muscle atrophy may be directly prevented in PAH. TRIAL REGISTRATION NUMBER NCT01847716; Results.
Collapse
Affiliation(s)
- Benjamin E Garfield
- National Heart and Lung Institute, Imperial College London, London, UK
- National Pulmonary Hypertension Service, Royal Brompton Hospital, London, UK
| | - Alexi Crosby
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Dongmin Shao
- National Heart and Lung Institute, Imperial College London, London, UK
- National Pulmonary Hypertension Service, Royal Brompton Hospital, London, UK
| | - Peiran Yang
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Cai Read
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Steven Sawiak
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Stephen Moore
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Lisa Parfitt
- National Pulmonary Hypertension Service, Royal Brompton Hospital, London, UK
| | - Carl Harries
- National Pulmonary Hypertension Service, Royal Brompton Hospital, London, UK
| | - Martin Rice
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Richard Paul
- NIHR Respiratory Biomedical Research Unit at the Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, UK
| | - Mark L Ormiston
- Departments of Biomedical and Molecular Sciences, Medicine and Surgery, Queen's University, Kingston, Ontario, Canada
| | - Nicholas W Morrell
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Michael I Polkey
- NIHR Respiratory Biomedical Research Unit at the Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, UK
| | - Stephen John Wort
- National Heart and Lung Institute, Imperial College London, London, UK
- National Pulmonary Hypertension Service, Royal Brompton Hospital, London, UK
| | - Paul R Kemp
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
17
|
Wafi AM, Hong J, Rudebush TL, Yu L, Hackfort B, Wang H, Schultz HD, Zucker IH, Gao L. Curcumin improves exercise performance of mice with coronary artery ligation-induced HFrEF: Nrf2 and antioxidant mechanisms in skeletal muscle. J Appl Physiol (1985) 2018; 126:477-486. [PMID: 30462567 DOI: 10.1152/japplphysiol.00654.2018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A hallmark of chronic heart failure (HF) with low ejection fraction (HFrEF) is exercise intolerance. We hypothesized that reduced expression of nuclear factor E2-related factor 2 (Nrf2) in skeletal muscle contributes to impaired exercise performance. We further hypothesized that curcumin, a Nrf2 activator, would preserve or increase exercise capacity in HF. Experiments were carried out in mice with coronary artery ligation-induced HFrEF. Curcumin was deliveried by a subcutaneous osmotic minipump at a dose of 50 mg·kg-1·day-1 for 8 weeks. In vivo, in situ, and in vitro experiments were employed to evaluate exercise capacity, muscle function, and molecular mechanisms. We found that: 1) the maximal speed, running distance to exhaustion, and limb grip force were significantly lower in HFrEF mice compared with sham. Curcumin-treated HF mice displayed enhanced exercise performance compared with vehicle-treated HF mice; 2) both soleus (Sol) and extensor digitorum longus (EDL) muscles of HFrEF mice exhibited reduced force and rapid fatigue, which were ameliorated by curcumin; and 3) protein expression of Nrf2, hemeoxygenase-1, SOD2, myogenin, and MyoD were significantly lower, but total ubiquitinated proteins, MURF1, and atrogen-1 were higher in Sol and EDL of HFrEF compared with sham mice, whereas these alterations in Nrf2 signaling and antioxidant defenses in HFrEF were attenuated by curcumin, which had no effect on cardiac function per se in mice with severe HFrEF. These data suggest that impaired Nrf2 signaling intrinsic to skeletal muscle contributes to exercise intolerance in HFrEF. Skeletal muscle Nrf2 should be considered as a novel therapeutic target in severe HF. NEW & NOTEWORTHY These studies suggest that impaired nuclear factor E2-related factor 2 (Nrf2) signaling is a critical mechanism underlying the enhanced oxidative stress in skeletal muscle in heart failure with low ejection fraction (HFrEF). Curcumin prevents the decline in running performance in HFrEF mice by upregulating antioxidant defenses in skeletal muscle, likely mediated by activating Nrf2 signaling. These findings suggest a novel therapeutic target for the improvement of exercise capacity and quality of life in HFrEF patients.
Collapse
Affiliation(s)
- Ahmed M Wafi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Nebraska Medical Center , Omaha, Nebraska
| | - Juan Hong
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Nebraska Medical Center , Omaha, Nebraska
| | - Tara L Rudebush
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Nebraska Medical Center , Omaha, Nebraska
| | - Li Yu
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Nebraska Medical Center , Omaha, Nebraska
| | - Bryan Hackfort
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Nebraska Medical Center , Omaha, Nebraska
| | - Hanjun Wang
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Nebraska Medical Center , Omaha, Nebraska
| | - Harold D Schultz
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Nebraska Medical Center , Omaha, Nebraska
| | - Irving H Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Nebraska Medical Center , Omaha, Nebraska
| | - Lie Gao
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Nebraska Medical Center , Omaha, Nebraska
| |
Collapse
|
18
|
Pinotti MF, Matias AM, Sugizaki MM, Nascimento AFD, Pai MD, Leopoldo APL, Cicogna AC, Leopoldo AS. Fasting/Refeeding Cycles Prevent Myocardial Dysfunction and Morphology Damage in the Spontaneously Hypertensive Rats. Arq Bras Cardiol 2018; 111:400-409. [PMID: 30133552 PMCID: PMC6173345 DOI: 10.5935/abc.20180152] [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: 11/10/2017] [Accepted: 05/09/2018] [Indexed: 11/23/2022] Open
Abstract
Background Caloric restriction is known to impair the cardiac function and morphology in
hypertrophied hearts of spontaneously hypertensive rats (SHR); however, the
influence of fasting/refeeding (RF) is unknown. Objective To investigate the fasting/refeeding approach on myocardial remodeling and
function. In addition, the current study was designed to bring information
regarding the mechanisms underlying the participation of Ca2+
handling and β-adrenergic system. Methods Sixty-day-old male SHR rats were submitted to food ad
libitum (C), 50% food restriction (R50) or RF cycles
for 90 days. Cardiac remodeling was assessed by ultrastructure analysis and
isolated papillary muscle function. The level of significance considered was
5% (α = 0.05). Results The RF rats presented lower cardiac atrophy than R50 in relation
to C rats. The C rats increased weight gain, R50 maintained their
initial body weight and RF rats increased and decreased weight during RF.
The RF did not cause functional impairment because the isotonic and
isometric parameters showed similar behavior to those of C. The isotonic and
isometric cardiac parameters were significantly elevated in RF rats compared
to R50 rats. In addition, the R50 rats had cardiac
damage in relation to C for isotonic and isometric variables. While the
R50 rats showed focal changes in many muscle fibers, the RF
rats displayed mild alterations, such as loss or disorganization of
myofibrils. Conclusion Fasting/refeeding promotes cardiac beneficial effects and attenuates
myocardial injury caused by caloric restriction in SHR rats, contributing to
reduce the cardiovascular risk profile and morphological injuries.
Furthermore, RF promotes mild improvement in Ca2+ handling and
β-adrenergic system.
Collapse
Affiliation(s)
- Matheus Fécchio Pinotti
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Estadual Paulista (UNESP), Botucatu, SP - Brazil
| | - Amanda Martins Matias
- Departamento de Desportos, Centro de Educação Física e Desportos, Universidade Federal do Espírito Santo (UFES), Vitória, ES - Brazil
| | | | | | - Maeli Dal Pai
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Estadual Paulista (UNESP), Botucatu, SP - Brazil.,Departamento de Morfologia, Instituto de Biosciências da Universidade Estadual Paulista UNESP, Botucatu, SP - Brazil
| | - Ana Paula Lima Leopoldo
- Departamento de Desportos, Centro de Educação Física e Desportos, Universidade Federal do Espírito Santo (UFES), Vitória, ES - Brazil
| | - Antônio Carlos Cicogna
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Estadual Paulista (UNESP), Botucatu, SP - Brazil
| | - André Soares Leopoldo
- Departamento de Desportos, Centro de Educação Física e Desportos, Universidade Federal do Espírito Santo (UFES), Vitória, ES - Brazil
| |
Collapse
|
19
|
Fujita N, Fujino H, Sakamoto H, Takegaki J, Deie M. Time course of ubiquitin-proteasome and macroautophagy-lysosome pathways in skeletal muscle in rats with heart failure. Biomed Res 2016; 36:383-92. [PMID: 26700592 DOI: 10.2220/biomedres.36.383] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Patients with heart failure have limited exercise capacity due to not only the myocardial dysfunction but also skeletal muscle atrophy. However, the mechanisms and time course of protein degradation in skeletal muscle during heart failure remain unclear, and there is no established standard treatment. The purpose of the present study was to investigate the time course of major protein degradation pathways in skeletal muscle during heart failure. Four-week-old male Wistar rats were randomly assigned to heart failure induced by monocrotaline or control groups. At 14 and 21 days after monocrotaline injection, the lungs, heart, and gastrocnemius and soleus muscles were removed and analyzed. There was no significant difference in body weight between the groups at 14 days after monocrotaline injection. Although there were no morphological changes in the skeletal muscle of the monocrotaline group at this time point, ubiquitin-proteasome and macroautophagylysosome pathways were activated in the monocrotaline group. Additionally, the pathways were less strongly activated in the soleus muscle than in the gastrocnemius muscle. These results suggest that physical exercise that shifts to slow muscle characteristics should begin when there is no indication of skeletal muscle atrophy to prevent exercise intolerance with heart failure.
Collapse
Affiliation(s)
- Naoto Fujita
- Department of Musculoskeletal Functional Research and Regeneration, Graduate School of Biomedicine and Health Sciences, Hiroshima University
| | | | | | | | | |
Collapse
|
20
|
Pacagnelli FL, Aguiar AF, Campos DHS, Castan EP, de Souza RWA, de Almeida FLA, Carani F, Carvalho RF, Cicogna AC, Silva MDP. Training improves the oxidative phenotype of muscle during the transition from cardiac hypertrophy to heart failure without altering MyoD and myogenin. Exp Physiol 2016; 101:1075-85. [PMID: 27219629 DOI: 10.1113/ep085552] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 05/19/2016] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? We investigated the effects of physical training on phenotypic (fibre-type content) and myogenic features (MyoD and myogenin expression) in skeletal muscle during the transition from cardiac hypertrophy to heart failure. What is the main finding and its importance? We provide new insight into skeletal muscle adaptations by showing that physical training increases the type I fibre content during the transition from cardiac hypertrophy to heart failure, without altering MyoD and myogenin expression. These results have important clinical implications for patients with heart failure, because this population has reduced muscle oxidative capacity. The purpose of this study was to investigate the effects of physical training (PT) on phenotypic features (fibre-type content) and myogenic regulatory factors (MyoD and myogenin) in rat skeletal muscle during the transition from cardiac hypertrophy to heart failure. We used the model of ascending aortic stenosis (AS) to induce heart failure in male Wistar rats. Sham-operated animals were used as age-matched controls. At 18 weeks after surgery, rats with ventricular dysfunction were randomized into the following four groups: sham-operated, untrained (Sham-U; n = 8); sham-operated, trained (Sham-T; n = 6); aortic stenosis, untrained (AS-U; n = 6); and aortic stenosis, trained (AS-T; n = 8). The AS-T and Sham-T groups were submitted to a 10 week aerobic PT programme, while the AS-U and Sham-U groups remained untrained for the same period of time. After the PT programme, the animals were killed and the soleus muscles collected for phenotypic and molecular analyses. Physical training promoted type IIa-to-I fibre conversion in the trained groups (Sham-T and AS-T) compared with the untrained groups (Sham-U and AS-U). No significant (P > 0.05) differences were found in type I or IIa fibre content in the AS-U group compared with the Sham-U group. Additionally, there were no significant (P > 0.05) differences in the myogenic regulatory factors MyoD and myogenin (gene and protein) expression between the groups. Therefore, our results indicate that PT may be a suitable strategy to improve the oxidative phenotype in skeletal muscle during the transition from cardiac hypertrophy to heart failure, without altering MyoD and myogenin.
Collapse
|
21
|
Pacagnelli FL, de Almeida Sabela AKD, Okoshi K, Mariano TB, Campos DHS, Carvalho RF, Cicogna AC, Vanderlei LCM. Preventive aerobic training exerts a cardioprotective effect on rats treated with monocrotaline. Int J Exp Pathol 2016; 97:238-47. [PMID: 27365256 DOI: 10.1111/iep.12166] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/06/2015] [Indexed: 01/18/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a chronic disease which causes overload to the right ventricle. The effect of preventive training on cardiac remodelling in this condition is still unknown. This study aimed to evaluate the influence of preventive training on hypertrophy, heart function and gene expression of calcium transport proteins in rats with monocrotaline-induced PAH. Thirty-two male Wistar rats were randomly divided into four groups: S, sedentary control; T, trained control; SM, sedentary monocrotaline; and TM, trained monocrotaline. The preventive training protocol was performed on a treadmill for 13 weeks, five times/week. The first two weeks were adopted for adaptation to training with gradual increases in speed/time. The speed of the physical training from the third to tenth weeks was gradually increased from 0.9 to 1.1 km/h for 60 min. Next, monocrotaline was applied (60 mg/kg) to induce PAH and lactate threshold analysis performed to determine the training speeds. The training speed of the TM group in the following two weeks was 0.8 km/h for 60 min and the T = 0.9 km/h for 60 min; in the final two weeks, both groups trained at the same speed and duration 0.9 km/h, 60 min. Cardiac function was assessed through echocardiography, ventricular hypertrophy through histomorphometric analysis and gene expression through RT-qPCR. Right cardiac function assessed through the peak flow velocity was SM = 75.5 cm/s vs. TM = 92.0 cm/s (P = 0.001), and ventricular hypertrophy was SM = 106.4 μm² vs. TM = 77.7 μm² (P = 0.004). There was a decrease in the gene expression of ryanodine S = 1.12 au vs. SM = 0.60 au (P = 0.02) without alterations due to training. Thus, we conclude that prior physical training exerts a cardioprotective effect on the right ventricle in the monocrotaline rat model.
Collapse
Affiliation(s)
- Francis Lopes Pacagnelli
- Department of Physiotherapy, University of Western São Paulo (UNOESTE), Presidente Prudente, São Paulo, Brazil
| | | | - Katashi Okoshi
- Department of Internal Medicine, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Thaoan Bruno Mariano
- Postgraduate Program in Animal Science, UNOESTE, Presidente Prudente, São Paulo, Brazil
| | | | | | - Antônio Carlos Cicogna
- Department of Internal Medicine, São Paulo State University, Botucatu, São Paulo, Brazil
| | | |
Collapse
|
22
|
Pacagnelli FL, Sabela AKDDA, Mariano TB, Ozaki GAT, Castoldi RC, Carmo EMD, Carvalho RF, Tomasi LC, Okoshi K, Vanderlei LCM. Fractal Dimension in Quantifying Experimental-Pulmonary-Hypertension-Induced Cardiac Dysfunction in Rats. Arq Bras Cardiol 2016; 107:33-9. [PMID: 27223643 PMCID: PMC4976954 DOI: 10.5935/abc.20160083] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/23/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Right-sided heart failure has high morbidity and mortality, and may be caused by pulmonary arterial hypertension. Fractal dimension is a differentiated and innovative method used in histological evaluations that allows the characterization of irregular and complex structures and the quantification of structural tissue changes. OBJECTIVE To assess the use of fractal dimension in cardiomyocytes of rats with monocrotaline-induced pulmonary arterial hypertension, in addition to providing histological and functional analysis. METHODS Male Wistar rats were divided into 2 groups: control (C; n = 8) and monocrotaline-induced pulmonary arterial hypertension (M; n = 8). Five weeks after pulmonary arterial hypertension induction with monocrotaline, echocardiography was performed and the animals were euthanized. The heart was dissected, the ventricles weighed to assess anatomical parameters, and histological slides were prepared and stained with hematoxylin/eosin for fractal dimension analysis, performed using box-counting method. Data normality was tested (Shapiro-Wilk test), and the groups were compared with non-paired Student t test or Mann Whitney test (p < 0.05). RESULTS Higher fractal dimension values were observed in group M as compared to group C (1.39 ± 0.05 vs. 1.37 ± 0.04; p < 0.05). Echocardiography showed lower pulmonary artery flow velocity, pulmonary acceleration time and ejection time values in group M, suggesting function worsening in those animals. CONCLUSION The changes observed confirm pulmonary-arterial-hypertension-induced cardiac dysfunction, and point to fractal dimension as an effective method to evaluate cardiac morphological changes induced by ventricular dysfunction.
Collapse
Affiliation(s)
| | | | | | | | | | - Edna Maria do Carmo
- Departamento de Fisioterapia, FCT, UNESP, Presidente Prudente, São Paulo, Brazil
| | | | | | - Katashi Okoshi
- Faculdade de Medicina, UNESP, Botucatu, São Paulo, Brazil
| | | |
Collapse
|
23
|
Sepulveda PV, Bush ED, Baar K. Pharmacology of manipulating lean body mass. Clin Exp Pharmacol Physiol 2015; 42:1-13. [PMID: 25311629 PMCID: PMC4383600 DOI: 10.1111/1440-1681.12320] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 09/29/2014] [Accepted: 09/29/2014] [Indexed: 01/04/2023]
Abstract
Dysfunction and wasting of skeletal muscle as a consequence of illness decreases the length and quality of life. Currently, there are few, if any, effective treatments available to address these conditions. Hence, the existence of this unmet medical need has fuelled large scientific efforts. Fortunately, these efforts have shown many of the underlying mechanisms adversely affecting skeletal muscle health. With increased understanding have come breakthrough disease-specific and broad spectrum interventions, some progressing through clinical development. The present review focuses its attention on the role of the antagonistic process regulating skeletal muscle mass before branching into prospective promising therapeutic targets and interventions. Special attention is given to therapies in development against cancer cachexia and Duchenne muscular dystrophy before closing remarks on design and conceptualization of future therapies are presented to the reader.
Collapse
Affiliation(s)
- Patricio V Sepulveda
- Department of Physiology, Monash University, Monash College Wellington Rd, Melbourne Victoria, Australia
| | - Ernest D Bush
- Akashi Therapeutics, Cambridge, MA, University of California Davis, Davis, CA, USA
| | - Keith Baar
- Departments of Neurobiology, Physiology and Behaviour and Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| |
Collapse
|
24
|
Effects of Fasting and Refeeding on Expression of Atrogin-1/MAFbx in Cardiac Muscle of Broiler Chickens. J Poult Sci 2015. [DOI: 10.2141/jpsa.0150036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
25
|
Aerobic exercise training prevents heart failure-induced skeletal muscle atrophy by anti-catabolic, but not anabolic actions. PLoS One 2014; 9:e110020. [PMID: 25330387 PMCID: PMC4201522 DOI: 10.1371/journal.pone.0110020] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 09/14/2014] [Indexed: 01/05/2023] Open
Abstract
Background Heart failure (HF) is associated with cachexia and consequent exercise intolerance. Given the beneficial effects of aerobic exercise training (ET) in HF, the aim of this study was to determine if the ET performed during the transition from cardiac dysfunction to HF would alter the expression of anabolic and catabolic factors, thus preventing skeletal muscle wasting. Methods and Results We employed ascending aortic stenosis (AS) inducing HF in Wistar male rats. Controls were sham-operated animals. At 18 weeks after surgery, rats with cardiac dysfunction were randomized to 10 weeks of aerobic ET (AS-ET) or to an untrained group (AS-UN). At 28 weeks, the AS-UN group presented HF signs in conjunction with high TNF-α serum levels; soleus and plantaris muscle atrophy; and an increase in the expression of TNF-α, NFκB (p65), MAFbx, MuRF1, FoxO1, and myostatin catabolic factors. However, in the AS-ET group, the deterioration of cardiac function was prevented, as well as muscle wasting, and the atrophy promoters were decreased. Interestingly, changes in anabolic factor expression (IGF-I, AKT, and mTOR) were not observed. Nevertheless, in the plantaris muscle, ET maintained high PGC1α levels. Conclusions Thus, the ET capability to attenuate cardiac function during the transition from cardiac dysfunction to HF was accompanied by a prevention of skeletal muscle atrophy that did not occur via an increase in anabolic factors, but through anti-catabolic activity, presumably caused by PGC1α action. These findings indicate the therapeutic potential of aerobic ET to block HF-induced muscle atrophy by counteracting the increased catabolic state.
Collapse
|
26
|
Abstract
Muscle RING finger 1 (MuRF1) and muscle atrophy F-box (MAFbx)/atrogin-1 were identified more than 10 years ago as two muscle-specific E3 ubiquitin ligases that are increased transcriptionally in skeletal muscle under atrophy-inducing conditions, making them excellent markers of muscle atrophy. In the past 10 years much has been published about MuRF1 and MAFbx with respect to their mRNA expression patterns under atrophy-inducing conditions, their transcriptional regulation, and their putative substrates. However, much remains to be learned about the physiological role of both genes in the regulation of mass and other cellular functions in striated muscle. Although both MuRF1 and MAFbx are enriched in skeletal, cardiac, and smooth muscle, this review will focus on the current understanding of MuRF1 and MAFbx in skeletal muscle, highlighting the critical questions that remain to be answered.
Collapse
Affiliation(s)
- Sue C Bodine
- Departments of Neurobiology, Physiology, and Behavior and Physiology and Membrane Biology, University of California Davis, Davis, California; and Northern California Veterans Affairs Health Systems, Mather, California
| | - Leslie M Baehr
- Membrane Biology, University of California Davis, Davis, California; and
| |
Collapse
|
27
|
Batt J, Ahmed SS, Correa J, Bain A, Granton J. Skeletal muscle dysfunction in idiopathic pulmonary arterial hypertension. Am J Respir Cell Mol Biol 2014; 50:74-86. [PMID: 23972212 DOI: 10.1165/rcmb.2012-0506oc] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Despite improvements in survival with disease-targeted therapies, the majority of patients with pulmonary arterial hypertension (PAH) have persistent exercise intolerance that results from impaired cardiac function and skeletal muscle dysfunction. Our intent was to understand the molecular mechanisms mediating skeletal muscle dysfunction in PAH. A total of 12 patients with PAH and 10 matched control subjects were assessed. Patients with PAH demonstrated diminished exercise capacity (lower oxygen uptake max, lower anaerobic threshold and higher minute ventilation/CO2) compared with control subjects. Quadriceps muscle cross-sectional area was significantly smaller in patients with PAH. The vastus lateralis muscle was biopsied to enable muscle fiber morphometric assessment and to determine expression levels/activation of proteins regulating (1) muscle mass, (2) mitochondria biogenesis and shaping machinery, and (3) excitation-contraction coupling. Patients with PAH demonstrated a decreased type I/type II muscle fiber ratio, with a smaller cross-sectional area in the type I fibers. Diminished AKT and p70S6 kinase phosphorylation, with increased atrogin-1 and muscle RING-finger protein-1 transcript levels, were evident in the PAH muscle, suggesting engagement of cellular signaling networks stimulating ubiquitin-proteasome-mediated proteolysis of muscle, with concurrent depression of networks mediating muscle hypertrophy. Although there were no differences in expression/activation of proteins associated with mitochondrial biogenesis or fission (MTCO2 [cytochrome C oxidase subunit II]/succinate dehydrogenase flavoprotein subunit A, mitochondrial transcription factor A, nuclear respiratory factor-1/dynamin-related protein 1 phosphorylation), protein levels of a positive regulator of mitochondrial fusion, Mitofusin2, were significantly lower in patients with PAH. Patients with PAH demonstrated increased phosphorylation of ryanodine receptor 1 receptors, suggesting that altered sarcoplasmic reticulum Ca(++) sequestration may impair excitation-contraction coupling in the PAH muscle. These data suggest that muscle dysfunction in PAH results from a combination of muscle atrophy and intrinsically impaired contractility.
Collapse
Affiliation(s)
- Jane Batt
- 1 Keenan Research Center, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and
| | | | | | | | | |
Collapse
|
28
|
Voltarelli VA, Bechara LRG, Bacurau AVN, Mattos KC, Dourado PMM, Bueno CR, Casarini DE, Negrao CE, Brum PC. Lack of β2 -adrenoceptors aggravates heart failure-induced skeletal muscle myopathy in mice. J Cell Mol Med 2014; 18:1087-97. [PMID: 24629015 PMCID: PMC4508148 DOI: 10.1111/jcmm.12253] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 01/20/2014] [Indexed: 12/17/2022] Open
Abstract
Skeletal myopathy is a hallmark of heart failure (HF) and has been associated with a poor prognosis. HF and other chronic degenerative diseases share a common feature of a stressed system: sympathetic hyperactivity. Although beneficial acutely, chronic sympathetic hyperactivity is one of the main triggers of skeletal myopathy in HF. Considering that β2 -adrenoceptors mediate the activity of sympathetic nervous system in skeletal muscle, we presently evaluated the contribution of β2 -adrenoceptors for the morphofunctional alterations in skeletal muscle and also for exercise intolerance induced by HF. Male WT and β2 -adrenoceptor knockout mice on a FVB genetic background (β2 KO) were submitted to myocardial infarction (MI) or SHAM surgery. Ninety days after MI both WT and β2 KO mice presented to cardiac dysfunction and remodelling accompanied by significantly increased norepinephrine and epinephrine plasma levels, exercise intolerance, changes towards more glycolytic fibres and vascular rarefaction in plantaris muscle. However, β2 KO MI mice displayed more pronounced exercise intolerance and skeletal myopathy when compared to WT MI mice. Skeletal muscle atrophy of infarcted β2 KO mice was paralleled by reduced levels of phosphorylated Akt at Ser 473 while increased levels of proteins related with the ubiquitin--proteasome system, and increased 26S proteasome activity. Taken together, our results suggest that lack of β2 -adrenoceptors worsen and/or anticipate the skeletal myopathy observed in HF.
Collapse
Affiliation(s)
- Vanessa A Voltarelli
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Wei B, Dui W, Liu D, Xing Y, Yuan Z, Ji G. MST1, a key player, in enhancing fast skeletal muscle atrophy. BMC Biol 2013; 11:12. [PMID: 23374633 PMCID: PMC3606410 DOI: 10.1186/1741-7007-11-12] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 02/01/2013] [Indexed: 02/06/2023] Open
Abstract
Background Skeletal muscle undergoes rapid atrophy upon denervation and the underlying mechanisms are complicated. FOXO3a has been implicated as a major mediator of muscle atrophy, but how its subcellular location and activity is controlled during the pathogenesis of muscle atrophy remains largely unknown. MST1 (Mammalian Sterile 20-like kinase 1) is identified as a central component of the Hippo signaling pathway. MST1 has been shown to mediate phosphorylation of FOXO3a at Ser207. Whether this MST1-FOXO signaling cascade exerts any functional consequence on cellular homeostasis remains to be investigated. Result We identified that MST1 kinase was expressed widely in skeletal muscles and was dramatically up-regulated in fast- but not slow-dominant skeletal muscles immediately following denervation. The results of our histological and biochemical studies demonstrated that deletion of MST1 significantly attenuated denervation-induced skeletal muscle wasting and decreased expression of Atrogin-1 and LC3 genes in fast-dominant skeletal muscles from three- to five-month-old adult mice. Further studies indicated that MST1, but not MST2, remarkably increased FOXO3a phosphorylation level at Ser207 and promoted its nuclear translocation in atrophic fast-dominant muscles. Conclusions We have established that MST1 kinase plays an important role in regulating denervation-induced skeletal muscle atrophy. During the early stage of muscle atrophy, the up-regulated MST1 kinase promoted progression of neurogenic atrophy in fast-dominant skeletal muscles through activation of FOXO3a transcription factors.
Collapse
Affiliation(s)
- Bin Wei
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Datun Road 15, Beijing 100101, China
| | | | | | | | | | | |
Collapse
|
30
|
Kostrominova TY, Reiner DS, Haas RH, Ingermanson R, McDonough PM. Automated methods for the analysis of skeletal muscle fiber size and metabolic type. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 306:275-332. [PMID: 24016528 DOI: 10.1016/b978-0-12-407694-5.00007-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
It is of interest to quantify the size, shape, and metabolic subtype of skeletal muscle fibers in many areas of biomedical research. To do so, skeletal muscle samples are sectioned transversely to the length of the muscle and labeled for extracellular or membrane proteins to delineate the fiber boundaries and additionally for biomarkers related to function or metabolism. The samples are digitally photographed and the fibers "outlined" for quantification of fiber cross-sectional area (CSA) using pointing devices interfaced to a computer, which is tedious, prone to error, and can be nonobjective. Here, we review methods for characterizing skeletal muscle fibers and describe new automated techniques, which rapidly quantify CSA and biomarkers. We discuss the applications of these methods to the characterization of mitochondrial dysfunctions, which underlie a variety of human afflictions, and we present a novel approach, utilizing images from the online Human Protein Atlas to predict relationships between fiber-specific protein expression, function, and metabolism.
Collapse
|
31
|
Treatment with pharmacological PPARα agonists stimulates the ubiquitin proteasome pathway and myofibrillar protein breakdown in skeletal muscle of rodents. Biochim Biophys Acta Gen Subj 2012; 1830:2105-17. [PMID: 23041501 DOI: 10.1016/j.bbagen.2012.09.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 09/28/2012] [Accepted: 09/28/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND Treatment of hyperlipidemic patients with fibrates, agonists of peroxisome proliferator-activated receptor α (PPARα), provokes muscle atrophy as a side effect. The molecular mechanism underlying this phenomenon is still unknown. We tested the hypothesis that activation of PPARα leads to an up-regulation of the ubiquitin proteasome system (UPS) which plays a major role in protein degradation in muscle. METHODS Rats, wild-type and PPARα-deficient mice (PPARα(-/-)) were treated with synthetic PPARα agonists (clofibrate, WY-14,643) to study their effect on the UPS and myofibrillar protein breakdown in muscle. RESULTS In rats and wild-type mice but not PPARα(-/-) mice, clofibrate or WY-14,643 caused increases in mRNA and protein levels of the ubiquitin ligases atrogin-1 and MuRF1 in muscle. Wild-type mice treated with WY-14,643 had a greater 3-methylhistidine release from incubated muscle and lesser muscle weights. In addition, wild-type mice but not PPARα(-/-) mice treated with WY-14,643 had higher amounts of ubiquitin-protein conjugates, a decreased activity of PI3K/Akt1 signalling, and an increased activity of FoxO1 transcription factor in muscle. Reporter gene and gel shift experiments revealed that the atrogin-1 and MuRF1 promoter do not contain functional PPARα DNA-binding sites. CONCLUSIONS These findings indicate that fibrates stimulate ubiquitination of proteins in skeletal muscle which in turn stimulates protein degradation. Up-regulation of ubiquitin ligases is probably not mediated by PPARα-dependent gene transcription but by PPARα-dependent inhibition of the PI3K/Akt1 signalling pathway leading to activation of FoxO1. GENERAL SIGNIFICANCE PPARα plays a role in the regulation of the ubiquitin proteasome system.
Collapse
|
32
|
CHEN K, CHENG HH, ZHOU RJ. Molecular mechanisms and functions of autophagy and the ubiq-uitin-proteasome pathway. YI CHUAN = HEREDITAS 2012; 34:5-18. [DOI: 10.3724/sp.j.1005.2012.00005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
33
|
Paffett ML, Lucas SN, Campen MJ. Resveratrol reverses monocrotaline-induced pulmonary vascular and cardiac dysfunction: a potential role for atrogin-1 in smooth muscle. Vascul Pharmacol 2011; 56:64-73. [PMID: 22146233 DOI: 10.1016/j.vph.2011.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 10/31/2011] [Accepted: 11/20/2011] [Indexed: 12/19/2022]
Abstract
Arterial remodeling contributes to elevated pulmonary artery (PA) pressures and right ventricular hypertrophy seen in pulmonary hypertension (PH). Resveratrol, a sirtuin-1 (SIRT1) pathway activator, can prevent the development of PH in a commonly used animal model, but it is unclear whether it can reverse established PH pathophysiology. Furthermore, atrophic ubiquitin ligases, such as atrogin-1 and MuRF-1, are known to be induced by SIRT1 activators but have not been characterized in hypertrophic vascular disease. Therefore, we hypothesized that monocrotaline (MCT)-induced PH would attenuate atrophy pathways in the PA while, conversely, SIRT1 activation (resveratrol) would reverse indices of PH and restore atrophic gene expression. Thus, we injected Sprague-Dawley rats with MCT (50 mg/kg i.p.) or saline at Day 0, and then treated with oral resveratrol or sildenafil from days 28-42 post-MCT injection. Oral resveratrol attenuated established MCT-induced PH indices, including right ventricular systolic pressure, right ventricular hypertrophy, and medial thickening of intrapulmonary arteries. Resveratrol also normalized PA atrogin-1 mRNA expression, which was significantly reduced by MCT. In cultured human PA smooth muscle cells (hPASMC), resveratrol significantly inhibited PDGF-stimulated proliferation and cellular hypertrophy, which was also associated with improvements in atrogin-1 levels. In addition, SIRT1 inhibition augmented hPASMC proliferation, as assessed by DNA mass, and suppressed atrogin mRNA expression. These findings demonstrate an inverse relationship between indices of PH and PA atrogin expression that is SIRT1 dependent and may reflect a novel role for SIRT1 in PASMCs opposing cellular hypertrophy and proliferation.
Collapse
Affiliation(s)
- Michael L Paffett
- College of Pharmacy, Division of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA
| | | | | |
Collapse
|
34
|
Freeman LM. Cachexia and sarcopenia: emerging syndromes of importance in dogs and cats. J Vet Intern Med 2011; 26:3-17. [PMID: 22111652 DOI: 10.1111/j.1939-1676.2011.00838.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 09/29/2011] [Accepted: 10/12/2011] [Indexed: 01/07/2023] Open
Abstract
Cachexia is the loss of lean body mass (LBM) that affects a large proportion of dogs and cats with congestive heart failure (CHF), chronic kidney disease (CKD), cancer, and a variety of other chronic diseases. Sarcopenia, the loss of LBM that occurs with aging, is a related syndrome, although sarcopenia occurs in the absence of disease. As many of the diseases associated with muscle loss are more common in aging, cachexia and sarcopenia often are concurrent problems. Both cachexia and sarcopenia have important clinical implications because they are associated with increased morbidity and mortality. The pathophysiology of these 2 syndromes is complex and multifactorial, but recent studies have provided new information that has helped to clarify mechanisms and identify potential new targets for treatment. Newly identified mechanisms and pathways that mediate cachexia appear to act by increasing energy requirements, decreasing energy intake, impairing nutrient absorption, and causing metabolic alterations. Whereas cachexia and sarcopenia are important areas of research for drug development in people, they are only beginning to be recognized in veterinary medicine. Greater awareness and earlier diagnosis will help provide practical approaches to managing body weight and lean tissue in dogs and cats, as well as more directed targets for treatment.
Collapse
Affiliation(s)
- L M Freeman
- Department of Clinical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, USA.
| |
Collapse
|
35
|
Wüst RCI, Myers DS, Stones R, Benoist D, Robinson PA, Boyle JP, Peers C, White E, Rossiter HB. Regional skeletal muscle remodeling and mitochondrial dysfunction in right ventricular heart failure. Am J Physiol Heart Circ Physiol 2011; 302:H402-11. [PMID: 22037189 DOI: 10.1152/ajpheart.00653.2011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Exercise intolerance is a cardinal symptom of right ventricular heart failure (RV HF) and skeletal muscle adaptations play a role in this limitation. We determined regional remodeling of muscle structure and mitochondrial function in a rat model of RV HF induced by monocrotaline injection (MCT; 60 mg·kg(-1); n = 11). Serial sections of the plantaris were stained for fiber type, succinate dehydrogenase (SDH) activity and capillaries. Mitochondrial function was assessed in permeabilized fibers using respirometry, and isolated complex activity by blue native gel electrophoresis (BN PAGE). All measurements were compared with saline-injected control animals (CON; n = 12). Overall fiber cross-sectional area was smaller in MCT than CON: 1,843 ± 114 vs. 2,322 ± 120 μm(2) (P = 0.009). Capillary-to-fiber ratio was lower in MCT in the oxidative plantaris region (1.65 ± 0.09 vs. 1.93 ± 0.07; P = 0.03), but not in the glycolytic region. SDH activity (P = 0.048) and maximal respiratory rate (P = 0.012) were each ∼15% lower in all fibers in MCT. ADP sensitivity was reduced in both skeletal muscle regions in MCT (P = 0.032), but normalized by rotenone. A 20% lower complex I/IV activity in MCT was confirmed by BN PAGE. MCT-treatment was associated with lower mitochondrial volume density (lower SDH activity), quality (lower complex I activity), and fewer capillaries per fiber area in oxidative skeletal muscle. These features are consistent with structural and functional remodeling of the determinants of oxygen supply potential and utilization that may contribute to exercise intolerance and reduced quality of life in patients with RV HF.
Collapse
Affiliation(s)
- Rob C I Wüst
- Institute of Membrane and Systems Biology, University of Leeds, Leeds, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
de Souza PAT, Matheus SMM, Castan EP, Campos DHS, Cicogna AC, Carvalho RF, Dal-Pai-Silva M. Morphological aspects of neuromuscular junctions and gene expression of nicotinic acetylcholine receptors (nAChRs) in skeletal muscle of rats with heart failure. J Mol Histol 2011; 42:557-65. [PMID: 21928074 DOI: 10.1007/s10735-011-9354-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 08/27/2011] [Indexed: 12/19/2022]
Abstract
HF is syndrome initiated by a reduction in cardiac function and it is characterized by the activation of compensatory mechanisms. Muscular fatigue and dyspnoea are the more common symptoms in HF; these may be due in part to specific skeletal muscle myopathy characterized by reduced oxidative capacity, a shift from slow fatigue resistant type I to fast less fatigue resistant type II fibers and downregulation of myogenic regulatory factors (MRFs) gene expression that can regulate gene expression of nicotinic acetylcholine receptors (nAChRs). In chronic heart failure, skeletal muscle phenotypic changes could influence the maintenance of the neuromuscular junction morphology and nAChRs gene expression during this syndrome. Two groups of rats were studied: control (CT) and Heart Failure (HF), induced by a single intraperitoneal injection of monocrotaline (MCT). At the end of the experiment, HF was evaluated by clinical signs and animals were sacrificed. Soleus (SOL) muscles were removed and processed for morphological, morphometric and molecular NMJ analyses. Our major finding was an up-regulation in the gene expression of the alpha1 and epsilon subunits of nAChR and a spot pattern of nAChR in SOL skeletal muscle in this acute monocrotaline induced HF. Our results suggest a remodeling of nAChR alpha1 and epsilon subunit during heart failure and may provide valuable information for understanding the skeletal muscle myopathy that occurs during this syndrome.
Collapse
|
37
|
Castillero E, Nieto-Bona MP, Fernández-Galaz C, Martín AI, López-Menduiña M, Granado M, Villanúa MA, López-Calderón A. Fenofibrate, a PPAR{alpha} agonist, decreases atrogenes and myostatin expression and improves arthritis-induced skeletal muscle atrophy. Am J Physiol Endocrinol Metab 2011; 300:E790-9. [PMID: 21304067 DOI: 10.1152/ajpendo.00590.2010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Arthritis is a chronic inflammatory illness that induces cachexia, which has a direct impact on morbidity and mortality. Fenofibrate, a selective PPARα activator prescribed to treat human dyslipidemia, has been reported to decrease inflammation in rheumatoid arthritis patients. The aim of this study was to elucidate whether fenofibrate is able to ameliorate skeletal muscle wasting in adjuvant-induced arthritis, an experimental model of rheumatoid arthritis. On day 4 after adjuvant injection, control and arthritic rats were treated with 300 mg/kg fenofibrate until day 15, when all rats were euthanized. Fenofibrate decreased external signs of arthritis and liver TNFα and blocked arthritis-induced decreased in PPARα expression in the gastrocnemius muscle. Arthritis decreased gastrocnemius weight, which results from a decrease in cross-section area and myofiber size, whereas fenofibrate administration to arthritic rats attenuated the decrease in both gastrocnemius weight and fast myofiber size. Fenofibrate treatment prevented arthritis-induced increase in atrogin-1 and MuRF1 expression in the gastrocnemius. Neither arthritis nor fenofibrate administration modify Akt-FoxO3 signaling. Myostatin expression was not modified by arthritis, but fenofibrate decreased myostatin expression in the gastrocnemius of arthritic rats. Arthritis increased muscle expression of MyoD, PCNA, and myogenin in the rats treated with vehicle but not in those treated with fenofibrate. The results indicate that, in experimental arthritis, fenofibrate decreases skeletal muscle atrophy through inhibition of the ubiquitin-proteasome system and myostatin.
Collapse
Affiliation(s)
- Estíbaliz Castillero
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense, Madrid, Spain 28040.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Bertaglia RS, Reissler J, Lopes FS, Cavalcante WLG, Carani FR, Padovani CR, Rodrigues SA, Cigogna AC, Carvalho RF, Fernandes AAH, Gallacci M, Silva MDP. Differential morphofunctional characteristics and gene expression in fast and slow muscle of rats with monocrotaline-induced heart failure. J Mol Histol 2011; 42:205-15. [DOI: 10.1007/s10735-011-9325-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 03/28/2011] [Indexed: 12/01/2022]
|