1
|
Han X, Li P, Jiang M, Cao Y, Wang Y, Jiang L, Liu X, Wu W. Autophagy in skeletal muscle dysfunction of chronic obstructive pulmonary disease: implications, mechanisms, and perspectives. J Zhejiang Univ Sci B 2025; 26:227-239. [PMID: 40082202 PMCID: PMC11906388 DOI: 10.1631/jzus.b2300680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/22/2023] [Indexed: 03/16/2025]
Abstract
Skeletal muscle dysfunction is a common extrapulmonary comorbidity of chronic obstructive pulmonary disease (COPD) and is associated with decreased quality-of-life and survival in patients. The autophagy lysosome pathway is one of the proteolytic systems that significantly affect skeletal muscle structure and function. Intriguingly, both promoting and inhibiting autophagy have been observed to improve COPD skeletal muscle dysfunction, yet the mechanism is unclear. This paper first reviewed the effects of macroautophagy and mitophagy on the structure and function of skeletal muscle in COPD, and then explored the mechanism of autophagy mediating the dysfunction of skeletal muscle in COPD. The results showed that macroautophagy- and mitophagy-related proteins were significantly increased in COPD skeletal muscle. Promoting macroautophagy in COPD improves myogenesis and replication capacity of muscle satellite cells, while inhibiting macroautophagy in COPD myotubes increases their diameters. Mitophagy helps to maintain mitochondrial homeostasis by removing impaired mitochondria in COPD. Autophagy is a promising target for improving COPD skeletal muscle dysfunction, and further research should be conducted to elucidate the specific mechanisms by which autophagy mediates COPD skeletal muscle dysfunction, with the aim of enhancing our understanding in this field.
Collapse
Affiliation(s)
- Xiaoyu Han
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Peijun Li
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Meiling Jiang
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Yuanyuan Cao
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Yingqi Wang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Linhong Jiang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaodan Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
- Institute of Rehabilitation Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China.
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai 201203, China.
| | - Weibing Wu
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
2
|
Li D, Pei Y, Liang L, Wang Z, Gai X, Sun Y. ADAMTS4 Reduction Contributes to Extracellular Matrix Deposition and Impaired Myogenesis in the Skeletal Muscle of Cigarette Smoke-Exposed Mice. Biomedicines 2025; 13:474. [PMID: 40002887 PMCID: PMC11853528 DOI: 10.3390/biomedicines13020474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Background: The extracellular matrix (ECM) plays a critical role in the proper regeneration of skeletal muscle. ECM remodeling has been reported in the skeletal muscle of chronic obstructive pulmonary disease (COPD), while the mechanisms remain poorly understood. Methods: In this study, we examined the dynamic interplay between ECM components and ECM enzymes in COPD skeletal muscle and cigarette smoke (CS) extract-treated C2C12 cells. C2C12 cells were further used to evaluate the role of a disintegrin and metalloproteinase with thrombospondin motif 4 (ADAMTS4) in ECM remodeling and myogenesis. Results: Chronic CS exposure induced the development of COPD and comorbid sarcopenia in C57BL/6J mice. Muscle fibrosis was observed in the gastrocnemius muscle of CS-exposed mice, accompanied by an upregulation of protein expression but a downregulation of mRNA levels of fibronectin and versican. We found that the discrepancy of mRNA and protein expression was attributed to the aberrant secretion of some ECM enzymes belonging to matrix metalloproteinases and ADAMTS proteases, especially ADAMTS4. CS exposure reduced ADAMTS4 expression in gastrocnemius muscles and C2C12 cells, and Adamts4 knockdown induced fibronectin and versican accumulation and impeded myogenic process. Conclusions: Considering that recent studies have indicated an impaired skeletal muscle regeneration in COPD, we suggested that the restrained production of ADAMTS4 in response to CS could be involved in the damaged muscle regeneration through regulating skeletal muscle ECM in COPD. Targeting ECM enzymes may benefit the rehabilitation of COPD-related sarcopenia.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoyan Gai
- Department of Respiratory and Critical Care Medicine, Research Center for Chronic Airway Diseases, Peking University Third Hospital, Peking University Health Science Center, Beijing 100191, China; (D.L.); (Y.P.); (L.L.); (Z.W.)
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Research Center for Chronic Airway Diseases, Peking University Third Hospital, Peking University Health Science Center, Beijing 100191, China; (D.L.); (Y.P.); (L.L.); (Z.W.)
| |
Collapse
|
3
|
Betz MW, De Brandt J, Aussieker T, Monsegue AP, Houtvast DCJ, Gehlert S, Verdijk LB, van Loon LJC, Gosker HR, Langen RJC, Derave W, Burtin C, Spruit MA, Snijders T. Muscle fibre satellite cells are located at a greater distance from capillaries in patients with COPD compared with healthy controls. ERJ Open Res 2024; 10:00203-2024. [PMID: 39351378 PMCID: PMC11440426 DOI: 10.1183/23120541.00203-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/13/2024] [Indexed: 10/04/2024] Open
Abstract
Background COPD is a disease characterised by skeletal muscle dysfunction. A spatial relationship exists between satellite cells and muscle fibre capillaries, which has been suggested to be of major importance for satellite cell function. In the present study we compared the spatial relationship between satellite cells and capillaries in patients with COPD and age-matched healthy older adults. Methods Muscle biopsies were obtained from the vastus lateralis of n=18 patients with COPD (8 female, 10 male; age 66±5 years, mild-to-severe airflow obstruction) and n=18 age-, sex- and body mass index-matched healthy control adults (8 female, 10 male; age 68±5 years). Immunohistochemistry was used to assess type I/II muscle fibre size, distribution, myonuclear content, satellite cell number and fibre capillarisation. In addition, type I/II muscle fibre satellite cell distance to its nearest capillary was assessed. Results The percentage of type II muscle fibres was significantly greater in patients with COPD (62±10%) compared with controls (50±12%, p<0.05). Muscle fibre capillarisation was significantly lower in patients with COPD compared with controls (p<0.05). While satellite cell content was not different between groups, type I and type II satellite cell distance to its nearest capillary was significantly greater in patients with COPD (type I: 21.3±4.8 µm; type II: 26.7±9.3 µm) compared with controls (type I: 16.1±3.5 µm; type II: 22.7±5.8 µm; p<0.05). Conclusion Satellite cells are located at a greater distance from their nearest capillary in patients with COPD compared with age-matched controls. This increased distance could play a role in impaired satellite cell function in patients with COPD.
Collapse
Affiliation(s)
- Milan W Betz
- NUTRIM Research Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Department of Human Biology, Maastricht, The Netherlands
| | - Jana De Brandt
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, BIOMED - Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Thorben Aussieker
- NUTRIM Research Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Department of Human Biology, Maastricht, The Netherlands
| | - Alejandra P Monsegue
- NUTRIM Research Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Department of Human Biology, Maastricht, The Netherlands
| | - Dion C J Houtvast
- NUTRIM Research Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Department of Human Biology, Maastricht, The Netherlands
| | - Sebastian Gehlert
- Department for Biosciences of Sports, Institute for Sport Science, University of Hildesheim, Hildesheim, Germany
| | - Lex B Verdijk
- NUTRIM Research Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Department of Human Biology, Maastricht, The Netherlands
| | - Luc J C van Loon
- NUTRIM Research Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Department of Human Biology, Maastricht, The Netherlands
| | - Harry R Gosker
- NUTRIM, Maastricht University Medical Centre+, Department of Respiratory Medicine, Maastricht, The Netherlands
| | - Ramon J C Langen
- NUTRIM, Maastricht University Medical Centre+, Department of Respiratory Medicine, Maastricht, The Netherlands
| | - Wim Derave
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| | - Chris Burtin
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, BIOMED - Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Martijn A Spruit
- NUTRIM, Maastricht University Medical Centre+, Department of Respiratory Medicine, Maastricht, The Netherlands
- Department of Research and Education, CIRO+, Horn, The Netherlands
| | - Tim Snijders
- NUTRIM Research Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Department of Human Biology, Maastricht, The Netherlands
| |
Collapse
|
4
|
Stevens NE, Loreti M, Ramirez-Sanchez I, Dos Reis FCG, Sacco A, Breen EC, Nogueira L. Cigarette smoke exposure impairs early-stage recovery from lengthening contraction-induced muscle injury in male mice. Physiol Rep 2024; 12:e70064. [PMID: 39328164 PMCID: PMC11427903 DOI: 10.14814/phy2.70064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/05/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024] Open
Abstract
The use of tobacco cigarettes produces locomotor muscle weakness and fatigue intolerance. Also, smokers and chronic obstructive pulmonary disease patients have a greater incidence of muscle injury and a deficient myogenic response. However, the effects of smoke exposure on the recovery from eccentric exercise-induced muscle injuries are unknown. Mice were exposed daily to cigarette smoke (CS) or room air (Air) for 4 months; the anterior crural muscles from one limb were injured by a lengthening contractions protocol (LCP) and recovered for 7 days. Lung compliance was greater, and body weights were lower, in CS-exposed than in the Air group. In LCP-subjected limbs, CS exposure lowered tibialis anterior myofiber cross-sectional area, decreased the size of centrally nucleated myofibers, and decreased extensor digitorum longus (EDL) mass, but did not affect EDL force from both limbs. CS exposure upregulated the mRNA levels of several myogenic (Pax7, Myf5, nNOS) genes in the EDL. The combination of CS exposure and LCP decreased Myf5 and nNOS mRNA levels and exacerbated pro-inflammatory mRNA levels. These data suggest that smoke exposure leads to an excessive pro-inflammatory response in regenerating muscle that is associated with a lower muscle mass recovery from a type of injury that often occurs during strenuous exercise.
Collapse
Affiliation(s)
- Nicole E Stevens
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Mafalda Loreti
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Israel Ramirez-Sanchez
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, IPN, Mexico City, Mexico
| | - Felipe C G Dos Reis
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Ellen C Breen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Leonardo Nogueira
- School of Exercise and Nutritional Sciences, College of Health and Human Services, San Diego State University, San Diego, California, USA
| |
Collapse
|
5
|
Yin A, Fu W, Elengickal A, Kim J, Liu Y, Bigot A, Mamchaoui K, Call JA, Yin H. Chronic hypoxia impairs skeletal muscle repair via HIF-2α stabilization. J Cachexia Sarcopenia Muscle 2024; 15:631-645. [PMID: 38333911 PMCID: PMC10995261 DOI: 10.1002/jcsm.13436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/28/2023] [Accepted: 01/02/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Chronic hypoxia and skeletal muscle atrophy commonly coexist in patients with COPD and CHF, yet the underlying physio-pathological mechanisms remain elusive. Muscle regeneration, driven by muscle stem cells (MuSCs), holds therapeutic potential for mitigating muscle atrophy. This study endeavours to investigate the influence of chronic hypoxia on muscle regeneration, unravel key molecular mechanisms, and explore potential therapeutic interventions. METHODS Experimental mice were exposed to prolonged normobaric hypoxic air (15% pO2, 1 atm, 2 weeks) to establish a chronic hypoxia model. The impact of chronic hypoxia on body composition, muscle mass, muscle strength, and the expression levels of hypoxia-inducible factors HIF-1α and HIF-2α in MuSC was examined. The influence of chronic hypoxia on muscle regeneration, MuSC proliferation, and the recovery of muscle mass and strength following cardiotoxin-induced injury were assessed. The muscle regeneration capacities under chronic hypoxia were compared between wildtype mice, MuSC-specific HIF-2α knockout mice, and mice treated with HIF-2α inhibitor PT2385, and angiotensin converting enzyme (ACE) inhibitor lisinopril. Transcriptomic analysis was performed to identify hypoxia- and HIF-2α-dependent molecular mechanisms. Statistical significance was determined using analysis of variance (ANOVA) and Mann-Whitney U tests. RESULTS Chronic hypoxia led to limb muscle atrophy (EDL: 17.7%, P < 0.001; Soleus: 11.5% reduction in weight, P < 0.001) and weakness (10.0% reduction in peak-isometric torque, P < 0.001), along with impaired muscle regeneration characterized by diminished myofibre cross-sectional areas, increased fibrosis (P < 0.001), and incomplete strength recovery (92.3% of pre-injury levels, P < 0.05). HIF-2α stabilization in MuSC under chronic hypoxia hindered MuSC proliferation (26.1% reduction of MuSC at 10 dpi, P < 0.01). HIF-2α ablation in MuSC mitigated the adverse effects of chronic hypoxia on muscle regeneration and MuSC proliferation (30.9% increase in MuSC numbers at 10 dpi, P < 0.01), while HIF-1α ablation did not have the same effect. HIF-2α stabilization under chronic hypoxia led to elevated local ACE, a novel direct target of HIF-2α. Notably, pharmacological interventions with PT2385 or lisinopril enhanced muscle regeneration under chronic hypoxia (PT2385: 81.3% increase, P < 0.001; lisinopril: 34.6% increase in MuSC numbers at 10 dpi, P < 0.05), suggesting their therapeutic potential for alleviating chronic hypoxia-associated muscle atrophy. CONCLUSIONS Chronic hypoxia detrimentally affects skeletal muscle regeneration by stabilizing HIF-2α in MuSC and thereby diminishing MuSC proliferation. HIF-2α increases local ACE levels in skeletal muscle, contributing to hypoxia-induced regenerative deficits. Administration of HIF-2α or ACE inhibitors may prove beneficial to ameliorate chronic hypoxia-associated muscle atrophy and weakness by improving muscle regeneration under chronic hypoxia.
Collapse
Affiliation(s)
- Amelia Yin
- Center for Molecular MedicineThe University of GeorgiaAthensGAUSA
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Wenyan Fu
- Center for Molecular MedicineThe University of GeorgiaAthensGAUSA
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Anthony Elengickal
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Joonhee Kim
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Yang Liu
- Center for Molecular MedicineThe University of GeorgiaAthensGAUSA
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Anne Bigot
- Sorbonne Université, Inserm, Institut de MyologieCentre de Recherche en MyologieParisFrance
| | - Kamal Mamchaoui
- Sorbonne Université, Inserm, Institut de MyologieCentre de Recherche en MyologieParisFrance
| | - Jarrod A. Call
- Department of Physiology and PharmacologyThe University of GeorgiaAthensGAUSA
| | - Hang Yin
- Center for Molecular MedicineThe University of GeorgiaAthensGAUSA
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| |
Collapse
|
6
|
Zheng G, Li C, Chen X, Deng Z, Xie T, Huo Z, Wei X, Huang Y, Zeng X, Luo Y, Bai J. HDAC9 inhibition reduces skeletal muscle atrophy and enhances regeneration in mice with cigarette smoke-induced COPD. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167023. [PMID: 38218381 DOI: 10.1016/j.bbadis.2024.167023] [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: 06/10/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
Cigarette smoke (CS) is the major risk factor for chronic obstructive pulmonary disease (COPD), and sarcopenia is one of the significant comorbidities of COPD. However, the pathogenesis of CS-related deficient skeletal muscle regeneration has yet to be clarified. The impact of CS on myoblast differentiation was examined, and then we determined which HDAC influenced the myogenic process and muscle atrophy in vitro and in vivo. Finally, we further investigated the potential mechanisms via RNA sequencing. Long-term CS exposure activated skeletal muscle primary satellite cells (SCs) while inhibiting differentiation, and defective myogenesis was also observed in C2C12 cells treated with CS extract (CSE). The level of HDAC9 changed in vitro and in vivo in CS exposure models as well as COPD patients, as detected by bioinformatics analysis. Our data showed that CSE impaired myogenic capacity and myotube formation in C2C12 cells via HDAC9. Moreover, inhibition of HDAC9 in mice exposed to CS prevented skeletal muscle dysfunction and promoted SC differentiation. The results of RNA-Seq analysis and verification indicated that HDAC9 knockout improved muscle differentiation in CS-exposed mice, probably by acting on the AKT/mTOR pathway and inhibiting the P53/P21 pathway. More importantly, the serum of HDAC9 KO mice exposed to CS alleviated the differentiation impairment of C2C12 cells caused by serum intervention in CS-exposed mice, and this effect was inhibited by LY294002 (an AKT/mTOR pathway inhibitor). These results suggest that HDAC9 plays an essential role in the defective regeneration induced by chronic exposure to CS.
Collapse
Affiliation(s)
- Guixian Zheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Chao Li
- Department of Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, Hunan 410219, China
| | - Xiaoli Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zhaohui Deng
- Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Zhuzhou, Hunan 412000, China
| | - Ting Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zengyu Huo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xinyan Wei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yanbing Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xia Zeng
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, China
| | - Yu Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, China
| | - Jing Bai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
7
|
Borges BM, Ramos RBC, Preite NW, Kaminski VDL, Alves de Castro P, Camacho M, Maximo MF, Fill TP, Calich VLG, Traynor AM, Sarikaya-Bayram Ö, Doyle S, Bayram Ö, de Campos CBL, Zelanis A, Goldman GH, Loures FV. Transcriptional profiling of a fungal granuloma reveals a low metabolic activity of Paracoccidioides brasiliensis yeasts and an actively regulated host immune response. Front Cell Infect Microbiol 2023; 13:1268959. [PMID: 37868350 PMCID: PMC10585178 DOI: 10.3389/fcimb.2023.1268959] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/11/2023] [Indexed: 10/24/2023] Open
Abstract
Granulomas are important immunological structures in the host defense against the fungus Paracoccidioides brasiliensis, the main etiologic agent of Paracoccidioidomycosis (PCM), a granulomatous systemic mycosis endemic in Latin America. We have performed transcriptional and proteomic studies of yeasts present in the pulmonary granulomas of PCM aiming to identify relevant genes and proteins that act under stressing conditions. C57BL/6 mice were infected with 1x106 yeasts and after 8- and 12-weeks of infection, granulomatous lesions were obtained for extraction of fungal and murine RNAs and fungal proteins. Dual transcriptional profiling was done comparing lung cells and P. brasiliensis yeasts from granulomas with uninfected lung cells and the original yeast suspension used in the infection, respectively. Mouse transcripts indicated a lung malfunction, with low expression of genes related to muscle contraction and organization. In addition, an increased expression of transcripts related to the activity of neutrophils, eosinophils, macrophages, lymphocytes as well as an elevated expression of IL-1β, TNF-α, IFN-γ, IL-17 transcripts were observed. The increased expression of transcripts for CTLA-4, PD-1 and arginase-1, provided evidence of immune regulatory mechanisms within the granulomatous lesions. Also, our results indicate iron as a key element for the granuloma to function, where a high number of transcripts related to fungal siderophores for iron uptake was observed, a mechanism of fungal virulence not previously described in granulomas. Furthermore, transcriptomics and proteomics analyzes indicated a low fungal activity within the granuloma, as demonstrated by the decreased expression of genes and proteins related to energy metabolism and cell cycle.
Collapse
Affiliation(s)
- Bruno Montanari Borges
- Institute of Science and Technology (ICT), Federal University of São Paulo (UNIFESP), São José dos Campos, SP, Brazil
| | - Rafael Berton Correia Ramos
- Institute of Science and Technology (ICT), Federal University of São Paulo (UNIFESP), São José dos Campos, SP, Brazil
| | - Nycolas Willian Preite
- Institute of Science and Technology (ICT), Federal University of São Paulo (UNIFESP), São José dos Campos, SP, Brazil
| | - Valéria de Lima Kaminski
- Institute of Science and Technology (ICT), Federal University of São Paulo (UNIFESP), São José dos Campos, SP, Brazil
| | - Patrícia Alves de Castro
- Faculty of Pharmaceutical Science of Ribeirão Preto (FCFRP), University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Maurício Camacho
- Institute of Science and Technology (ICT), Federal University of São Paulo (UNIFESP), São José dos Campos, SP, Brazil
| | | | - Taicia Pacheco Fill
- Institute of Chemistry, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Vera Lúcia Garcia Calich
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Aimee M. Traynor
- Department of Biology, Maynooth University, Maynooth, County Kildare, Ireland
| | | | - Sean Doyle
- Department of Biology, Maynooth University, Maynooth, County Kildare, Ireland
| | - Özgür Bayram
- Department of Biology, Maynooth University, Maynooth, County Kildare, Ireland
| | | | - André Zelanis
- Institute of Science and Technology (ICT), Federal University of São Paulo (UNIFESP), São José dos Campos, SP, Brazil
| | - Gustavo H. Goldman
- Faculty of Pharmaceutical Science of Ribeirão Preto (FCFRP), University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Flávio Vieira Loures
- Institute of Science and Technology (ICT), Federal University of São Paulo (UNIFESP), São José dos Campos, SP, Brazil
| |
Collapse
|
8
|
Gallagher H, Hendrickse PW, Pereira MG, Bowen TS. Skeletal muscle atrophy, regeneration, and dysfunction in heart failure: Impact of exercise training. JOURNAL OF SPORT AND HEALTH SCIENCE 2023; 12:557-567. [PMID: 37040849 PMCID: PMC10466197 DOI: 10.1016/j.jshs.2023.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/30/2022] [Accepted: 02/20/2023] [Indexed: 05/31/2023]
Abstract
This review highlights some established and some more contemporary mechanisms responsible for heart failure (HF)-induced skeletal muscle wasting and weakness. We first describe the effects of HF on the relationship between protein synthesis and degradation rates, which determine muscle mass, the involvement of the satellite cells for continual muscle regeneration, and changes in myofiber calcium homeostasis linked to contractile dysfunction. We then highlight key mechanistic effects of both aerobic and resistance exercise training on skeletal muscle in HF and outline its application as a beneficial treatment. Overall, HF causes multiple impairments related to autophagy, anabolic-catabolic signaling, satellite cell proliferation, and calcium homeostasis, which together promote fiber atrophy, contractile dysfunction, and impaired regeneration. Although both wasting and weakness are partly rescued by aerobic and resistance exercise training in HF, the effects of satellite cell dynamics remain poorly explored.
Collapse
Affiliation(s)
- Harrison Gallagher
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Paul W Hendrickse
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Marcelo G Pereira
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - T Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
9
|
Henrot P, Dupin I, Schilfarth P, Esteves P, Blervaque L, Zysman M, Gouzi F, Hayot M, Pomiès P, Berger P. Main Pathogenic Mechanisms and Recent Advances in COPD Peripheral Skeletal Muscle Wasting. Int J Mol Sci 2023; 24:ijms24076454. [PMID: 37047427 PMCID: PMC10095391 DOI: 10.3390/ijms24076454] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a worldwide prevalent respiratory disease mainly caused by tobacco smoke exposure. COPD is now considered as a systemic disease with several comorbidities. Among them, skeletal muscle dysfunction affects around 20% of COPD patients and is associated with higher morbidity and mortality. Although the histological alterations are well characterized, including myofiber atrophy, a decreased proportion of slow-twitch myofibers, and a decreased capillarization and oxidative phosphorylation capacity, the molecular basis for muscle atrophy is complex and remains partly unknown. Major difficulties lie in patient heterogeneity, accessing patients' samples, and complex multifactorial process including extrinsic mechanisms, such as tobacco smoke or disuse, and intrinsic mechanisms, such as oxidative stress, hypoxia, or systemic inflammation. Muscle wasting is also a highly dynamic process whose investigation is hampered by the differential protein regulation according to the stage of atrophy. In this review, we report and discuss recent data regarding the molecular alterations in COPD leading to impaired muscle mass, including inflammation, hypoxia and hypercapnia, mitochondrial dysfunction, diverse metabolic changes such as oxidative and nitrosative stress and genetic and epigenetic modifications, all leading to an impaired anabolic/catabolic balance in the myocyte. We recapitulate data concerning skeletal muscle dysfunction obtained in the different rodent models of COPD. Finally, we propose several pathways that should be investigated in COPD skeletal muscle dysfunction in the future.
Collapse
Affiliation(s)
- Pauline Henrot
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
- CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC 1401, Service de Pneumologie, F-33604 Pessac, France
| | - Isabelle Dupin
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
| | - Pierre Schilfarth
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
- CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC 1401, Service de Pneumologie, F-33604 Pessac, France
| | - Pauline Esteves
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
| | - Léo Blervaque
- PhyMedExp, INSERM-CNRS-Montpellier University, F-34090 Montpellier, France
| | - Maéva Zysman
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
- CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC 1401, Service de Pneumologie, F-33604 Pessac, France
| | - Fares Gouzi
- PhyMedExp, INSERM-CNRS-Montpellier University, CHRU Montpellier, F-34090 Montpellier, France
| | - Maurice Hayot
- PhyMedExp, INSERM-CNRS-Montpellier University, CHRU Montpellier, F-34090 Montpellier, France
| | - Pascal Pomiès
- PhyMedExp, INSERM-CNRS-Montpellier University, F-34090 Montpellier, France
| | - Patrick Berger
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
- CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC 1401, Service de Pneumologie, F-33604 Pessac, France
| |
Collapse
|
10
|
Henrot P, Blervaque L, Dupin I, Zysman M, Esteves P, Gouzi F, Hayot M, Pomiès P, Berger P. Cellular interplay in skeletal muscle regeneration and wasting: insights from animal models. J Cachexia Sarcopenia Muscle 2023; 14:745-757. [PMID: 36811134 PMCID: PMC10067506 DOI: 10.1002/jcsm.13103] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/24/2022] [Accepted: 09/02/2022] [Indexed: 02/24/2023] Open
Abstract
Skeletal muscle wasting, whether related to physiological ageing, muscle disuse or to an underlying chronic disease, is a key determinant to quality of life and mortality. However, cellular basis responsible for increased catabolism in myocytes often remains unclear. Although myocytes represent the vast majority of skeletal muscle cellular population, they are surrounded by numerous cells with various functions. Animal models, mostly rodents, can help to decipher the mechanisms behind this highly dynamic process, by allowing access to every muscle as well as time-course studies. Satellite cells (SCs) play a crucial role in muscle regeneration, within a niche also composed of fibroblasts and vascular and immune cells. Their proliferation and differentiation is altered in several models of muscle wasting such as cancer, chronic kidney disease or chronic obstructive pulmonary disease (COPD). Fibro-adipogenic progenitor cells are also responsible for functional muscle growth and repair and are associated in disease to muscle fibrosis such as in chronic kidney disease. Other cells have recently proven to have direct myogenic potential, such as pericytes. Outside their role in angiogenesis, endothelial cells and pericytes also participate to healthy muscle homoeostasis by promoting SC pool maintenance (so-called myogenesis-angiogenesis coupling). Their role in chronic diseases muscle wasting has been less studied. Immune cells are pivotal for muscle repair after injury: Macrophages undergo a transition from the M1 to the M2 state along with the transition between the inflammatory and resolutive phase of muscle repair. T regulatory lymphocytes promote and regulate this transition and are also able to activate SC proliferation and differentiation. Neural cells such as terminal Schwann cells, motor neurons and kranocytes are notably implicated in age-related sarcopenia. Last, newly identified cells in skeletal muscle, such as telocytes or interstitial tenocytes could play a role in tissular homoeostasis. We also put a special focus on cellular alterations occurring in COPD, a chronic and highly prevalent respiratory disease mainly linked to tobacco smoke exposure, where muscle wasting is strongly associated with increased mortality, and discuss the pros and cons of animal models versus human studies in this context. Finally, we discuss resident cells metabolism and present future promising leads for research, including the use of muscle organoids.
Collapse
Affiliation(s)
- Pauline Henrot
- Centre de Recherche Cardio-thoracique de Bordeaux, Univ-Bordeaux, Pessac, France.,Centre de Recherche Cardio-thoracique de Bordeaux, INSERM, Pessac, France.,CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Pessac, France
| | - Léo Blervaque
- PhyMedExp, INSERM-CNRS-Montpellier University, Montpellier, France
| | - Isabelle Dupin
- Centre de Recherche Cardio-thoracique de Bordeaux, Univ-Bordeaux, Pessac, France.,Centre de Recherche Cardio-thoracique de Bordeaux, INSERM, Pessac, France
| | - Maéva Zysman
- Centre de Recherche Cardio-thoracique de Bordeaux, Univ-Bordeaux, Pessac, France.,Centre de Recherche Cardio-thoracique de Bordeaux, INSERM, Pessac, France.,CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Pessac, France
| | - Pauline Esteves
- Centre de Recherche Cardio-thoracique de Bordeaux, Univ-Bordeaux, Pessac, France.,Centre de Recherche Cardio-thoracique de Bordeaux, INSERM, Pessac, France
| | - Fares Gouzi
- PhyMedExp, INSERM-CNRS-Montpellier University, CHRU Montpellier, Montpellier, France
| | - Maurice Hayot
- PhyMedExp, INSERM-CNRS-Montpellier University, CHRU Montpellier, Montpellier, France
| | - Pascal Pomiès
- PhyMedExp, INSERM-CNRS-Montpellier University, Montpellier, France
| | - Patrick Berger
- Centre de Recherche Cardio-thoracique de Bordeaux, Univ-Bordeaux, Pessac, France.,Centre de Recherche Cardio-thoracique de Bordeaux, INSERM, Pessac, France.,CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Pessac, France
| |
Collapse
|
11
|
Impaired muscle stem cell function and abnormal myogenesis in acquired myopathies. Biosci Rep 2023; 43:232343. [PMID: 36538023 PMCID: PMC9829652 DOI: 10.1042/bsr20220284] [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: 10/12/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle possesses a high plasticity and a remarkable regenerative capacity that relies mainly on muscle stem cells (MuSCs). Molecular and cellular components of the MuSC niche, such as immune cells, play key roles to coordinate MuSC function and to orchestrate muscle regeneration. An abnormal infiltration of immune cells and/or imbalance of pro- and anti-inflammatory cytokines could lead to MuSC dysfunctions that could have long lasting effects on muscle function. Different genetic variants were shown to cause muscular dystrophies that intrinsically compromise MuSC function and/or disturb their microenvironment leading to impaired muscle regeneration that contributes to disease progression. Alternatively, many acquired myopathies caused by comorbidities (e.g., cardiopulmonary or kidney diseases), chronic inflammation/infection, or side effects of different drugs can also perturb MuSC function and their microenvironment. The goal of this review is to comprehensively summarize the current knowledge on acquired myopathies and their impact on MuSC function. We further describe potential therapeutic strategies to restore MuSC regenerative capacity.
Collapse
|
12
|
Jaitovich A. Impaired regenerative capacity contributes to skeletal muscle dysfunction in chronic obstructive pulmonary disease. Am J Physiol Cell Physiol 2022; 323:C974-C989. [PMID: 35993519 PMCID: PMC9484993 DOI: 10.1152/ajpcell.00292.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 01/18/2023]
Abstract
Locomotor skeletal muscle dysfunction is a relevant comorbidity of chronic obstructive pulmonary disease (COPD) and is strongly associated with worse clinical outcomes including higher mortality. Over the last decades, a large body of literature helped characterize the process, defining the disruptive muscle phenotype caused by COPD that involves reduction in muscle mass, force-generation capacity, fatigue-tolerance, and regenerative potential following injury. A major limitation in the field has been the scarcity of well-calibrated animal models to conduct mechanistic research based on loss- and gain-of-function studies. This article provides an overall description of the process, the tools available to mechanistically investigate it, and the potential role of mitochondrially driven metabolic signals on the regulation muscle regeneration after injury in COPD. Finally, a description of future avenues to further expand on the area is proposed based on very recent evidence involving mitochondrial metabolic cues affecting myogenesis.
Collapse
Affiliation(s)
- Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine, Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| |
Collapse
|
13
|
Abati E, Manini A, Comi GP, Corti S. Inhibition of myostatin and related signaling pathways for the treatment of muscle atrophy in motor neuron diseases. Cell Mol Life Sci 2022; 79:374. [PMID: 35727341 PMCID: PMC9213329 DOI: 10.1007/s00018-022-04408-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 11/26/2022]
Abstract
Myostatin is a negative regulator of skeletal muscle growth secreted by skeletal myocytes. In the past years, myostatin inhibition sparked interest among the scientific community for its potential to enhance muscle growth and to reduce, or even prevent, muscle atrophy. These characteristics make it a promising target for the treatment of muscle atrophy in motor neuron diseases, namely, amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), which are rare neurological diseases, whereby the degeneration of motor neurons leads to progressive muscle loss and paralysis. These diseases carry a huge burden of morbidity and mortality but, despite this unfavorable scenario, several therapeutic advancements have been made in the past years. Indeed, a number of different curative therapies for SMA have been approved, leading to a revolution in the life expectancy and outcomes of SMA patients. Similarly, tofersen, an antisense oligonucleotide, is now undergoing clinical trial phase for use in ALS patients carrying the SOD1 mutation. However, these therapies are not able to completely halt or reverse progression of muscle damage. Recently, a trial evaluating apitegromab, a myostatin inhibitor, in SMA patients was started, following positive results from preclinical studies. In this context, myostatin inhibition could represent a useful strategy to tackle motor symptoms in these patients. The aim of this review is to describe the myostatin pathway and its role in motor neuron diseases, and to summarize and critically discuss preclinical and clinical studies of myostatin inhibitors in SMA and ALS. Then, we will highlight promises and pitfalls related to the use of myostatin inhibitors in the human setting, to aid the scientific community in the development of future clinical trials.
Collapse
Affiliation(s)
- Elena Abati
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, Neurology Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Neurology Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Arianna Manini
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, Neurology Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Giacomo Pietro Comi
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, Neurology Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Neurology Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, Neurology Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, University of Milan, Milan, Italy.
- Neurology Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
14
|
Balnis J, Drake LA, Singer DV, Vincent CE, Korponay TC, D’Armiento J, Lee CG, Elias JA, Singer HA, Jaitovich A. Deaccelerated Myogenesis and Autophagy in Genetically Induced Pulmonary Emphysema. Am J Respir Cell Mol Biol 2022; 66:623-637. [PMID: 35286819 PMCID: PMC9163640 DOI: 10.1165/rcmb.2021-0351oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/31/2022] [Indexed: 01/18/2023] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD)-pulmonary emphysema often develop locomotor muscle dysfunction, which entails reduced muscle mass and force-generation capacity and is associated with worse outcomes, including higher mortality. Myogenesis contributes to adult muscle integrity during injury-repair cycles. Injurious events crucially occur in the skeletal muscles of patients with COPD in the setting of exacerbations and infections, which lead to acute decompensations for limited periods of time, after which patients typically fail to recover the baseline status they had before the acute event. Autophagy, which is dysregulated in muscles from patients with COPD, is a key regulator of muscle stem-satellite- cells activation and myogenesis, yet very little research has so far mechanistically investigated the role of autophagy dysregulation in COPD muscles. Using a genetically inducible interleukin-13-driven pulmonary emphysema model leading to muscle dysfunction, and confirmed with a second genetic animal model, we found a significant myogenic dysfunction associated with the reduced proliferative capacity of satellite cells. Transplantation experiments followed by lineage tracing suggest that an intrinsic defect in satellite cells, and not in the COPD environment, plays a dominant role in the observed myogenic dysfunction. RNA sequencing analysis and direct observation of COPD mice satellite cells suggest dysregulated autophagy. Moreover, while autophagy flux experiments with bafilomycin demonstrated deacceleration of autophagosome turnover in COPD mice satellite cells, spermidine-induced autophagy stimulation leads to a higher replication rate and myogenesis in these animals. Our data suggest that pulmonary emphysema causes disrupted myogenesis, which could be improved with stimulation of autophagy and satellite cells activation, leading to an attenuated muscle dysfunction.
Collapse
Affiliation(s)
- Joseph Balnis
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Lisa A. Drake
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Diane V. Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Catherine E. Vincent
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Tanner C. Korponay
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Jeanine D’Armiento
- Departments of Anesthesiology and Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, New York, New York; and
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Jack A. Elias
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Harold A. Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| |
Collapse
|
15
|
Baker LA, O'Sullivan TF, Robinson KA, Graham-Brown MPM, Major RW, Ashford RU, Smith AC, Philp A, Watson EL. Primary skeletal muscle cells from chronic kidney disease patients retain hallmarks of cachexia in vitro. J Cachexia Sarcopenia Muscle 2022; 13:1238-1249. [PMID: 35029054 PMCID: PMC8978027 DOI: 10.1002/jcsm.12802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/23/2021] [Accepted: 08/23/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Skeletal muscle wasting and dysfunction are common characteristics noted in people who suffer from chronic kidney disease (CKD). The mechanisms by which this occurs are complex, and although progress has been made, the key underpinning mechanisms are not yet fully elucidated. With work to date primarily conducted in nephrectomy-based animal models, translational capacity to our patient population has been challenging. This could be overcome if rationale developing work could be conducted in human based models with greater translational capacity. This could be achieved using cells derived from patient biopsies, if they retain phenotypic traits noted in vivo. METHODS Here, we performed a systematic characterization of CKD derived muscle cells (CKD; n = 10; age: 54.40 ± 15.53 years; eGFR: 22.25 ± 13.22 ml/min/1.73 m2 ) in comparison with matched controls (CON; n = 10; age: 58.66 ± 14.74 years; eGFR: 85.81 ± 8.09 ml/min/1.73 m2 ). Harvested human derived muscle cells (HDMCs) were taken through proliferative and differentiation phases and investigated in the context of myogenic progression, inflammation, protein synthesis, and protein breakdown. Follow up investigations exposed HDMC myotubes from each donor type to 0, 0.4, and 100 nM of IGF-1 in order to investigate any differences in anabolic resistance. RESULTS Harvested human derived muscle cells isolated from CKD patients displayed higher rates of protein degradation (P = 0.044) alongside elevated expression of both TRIM63 (2.28-fold higher, P = 0.054) and fbox32 (6.4-fold higher, P < 0.001) in comparison with CONs. No differences were noted in rates of protein synthesis under basal conditions (P > 0.05); however, CKD derived cells displayed a significant degree of anabolic resistance in response to IGF-1 stimulation (both doses) in comparison with matched CONs (0.4 nm: P < 0.001; 100 nM: P < 0.001). CONCLUSIONS In summary, we report for the first time that HDMCs isolated from people suffering from CKD display key hallmarks of the well documented in vivo phenotype. Not only do these findings provide further mechanistic insight into CKD specific cachexia, but they also demonstrate this is a reliable and suitable model in which to perform targeted experiments to begin to develop novel therapeutic strategies targeting the CKD associated decline in skeletal muscle mass and function.
Collapse
Affiliation(s)
- Luke A Baker
- Department of Health Sciences, University of Leicester, Leicester, UK
| | | | | | - Matthew P M Graham-Brown
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK.,Department of Cardiovascular Science, NIHR Leicester Cardiovascular Biomedical Research Unit, Leicester, UK
| | - Rupert W Major
- Department of Health Sciences, University of Leicester, Leicester, UK.,John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Robert U Ashford
- Leicester Orthopaedics, University Hospitals of Leicester, Leicester, UK.,Department of Cancer Studies, University of Leicester, Leicester, UK
| | - Alice C Smith
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Andrew Philp
- Mitochondrial Metabolism and Ageing Laboratory, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, UNSW Medicine, UNSW, Sydney, NSW, Australia
| | - Emma L Watson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| |
Collapse
|
16
|
Nucera F, Mumby S, Paudel KR, Dharwal V, DI Stefano A, Casolaro V, Hansbro PM, Adcock IM, Caramori G. Role of oxidative stress in the pathogenesis of COPD. Minerva Med 2022; 113:370-404. [PMID: 35142479 DOI: 10.23736/s0026-4806.22.07972-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chronic inhalation of cigarette smoke is a prominent cause of chronic obstructive pulmonary disease (COPD) and provides an important source of exogenous oxidants. In addition, several inflammatory and structural cells are a source of endogenous oxidants in the lower airways of COPD patients, even in former smokers. This suggests that oxidants play a key role in the pathogenesis of COPD. This oxidative stress is counterbalanced by the protective effects of the various endogenous antioxidant defenses of the lower airways. A large amount of data from animal models and patients with COPD have shown that both the stable phase of the disease, and during exacerbations, have increased oxidative stress in the lower airways compared with age-matched smokers with normal lung function. Thus, counteracting the increased oxidative stress may produce clinical benefits in COPD patients. Smoking cessation is currently the most effective treatment of COPD patients and reduces oxidative stress in the lower airways. In addition, many drugs used to treat COPD have some antioxidant effects, however, it is still unclear if their clinical efficacy is related to pharmacological modulation of the oxidant/antioxidant balance. Several new antioxidant compounds are in development for the treatment of COPD.
Collapse
Affiliation(s)
- Francesco Nucera
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy -
| | - Sharon Mumby
- Airways Diseases Section, Faculty of Medicine, Imperial College London, National Heart and Lung Institute, London, UK
| | - Keshav R Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Vivek Dharwal
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Antonino DI Stefano
- Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell'Apparato Cardio Respiratorio, Istituti Clinici Scientifici Maugeri, IRCCS, Veruno, Novara, Italy
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry Scuola Medica Salernitana, University of Salerno, Salerno, Italy
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Ian M Adcock
- Airways Diseases Section, Faculty of Medicine, Imperial College London, National Heart and Lung Institute, London, UK
| | - Gaetano Caramori
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| |
Collapse
|
17
|
Catteau M, Passerieux E, Blervaque L, Gouzi F, Ayoub B, Hayot M, Pomiès P. Response to Electrostimulation Is Impaired in Muscle Cells from Patients with Chronic Obstructive Pulmonary Disease. Cells 2021; 10:3002. [PMID: 34831227 PMCID: PMC8616440 DOI: 10.3390/cells10113002] [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: 10/06/2021] [Revised: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 11/16/2022] Open
Abstract
Among the comorbidities associated with chronic obstructive pulmonary disease (COPD), skeletal muscle weakness and atrophy are known to affect patient survival rate. In addition to muscle deconditioning, various systemic and intrinsic factors have been implicated in COPD muscle dysfunction but an impaired COPD muscle adaptation to contraction has never been extensively studied. We submitted cultured myotubes from nine healthy subjects and nine patients with COPD to an endurance-type protocol of electrical pulse stimulation (EPS). EPS induced a decrease in the diameter, covered surface and expression of MHC1 in COPD myotubes. Although the expression of protein degradation markers was not affected, expression of the protein synthesis marker mTOR was not induced in COPD compared to healthy myotubes after EPS. The expression of the differentiation markers p16INK4a and p21 was impaired, while expression of Myf5 and MyoD tended to be affected in COPD muscle cells in response to EPS. The expression of mitochondrial biogenesis markers PGC1α and MFN2 was affected and expression of TFAM and COX1 tended to be reduced in COPD compared to healthy myotubes upon EPS. Lipid peroxidation was increased and the expression of the antioxidant enzymes SOD2 and GPx4 was affected in COPD compared to healthy myotubes in response to EPS. Thus, we provide evidence of an impaired response of COPD muscle cells to contraction, which might be involved in the muscle weakness observed in patients with COPD.
Collapse
Affiliation(s)
- Matthias Catteau
- PhyMedExp, University of Montpellier—INSERM—CNRS, 34295 Montpellier, France; (M.C.); (E.P.); (L.B.)
| | - Emilie Passerieux
- PhyMedExp, University of Montpellier—INSERM—CNRS, 34295 Montpellier, France; (M.C.); (E.P.); (L.B.)
| | - Léo Blervaque
- PhyMedExp, University of Montpellier—INSERM—CNRS, 34295 Montpellier, France; (M.C.); (E.P.); (L.B.)
| | - Farés Gouzi
- PhyMedExp, University of Montpellier—INSERM—CNRS—CHRU Montpellier, 34295 Montpellier, France; (F.G.); (B.A.); (M.H.)
| | - Bronia Ayoub
- PhyMedExp, University of Montpellier—INSERM—CNRS—CHRU Montpellier, 34295 Montpellier, France; (F.G.); (B.A.); (M.H.)
| | - Maurice Hayot
- PhyMedExp, University of Montpellier—INSERM—CNRS—CHRU Montpellier, 34295 Montpellier, France; (F.G.); (B.A.); (M.H.)
| | - Pascal Pomiès
- PhyMedExp, University of Montpellier—INSERM—CNRS, 34295 Montpellier, France; (M.C.); (E.P.); (L.B.)
| |
Collapse
|
18
|
Lee SJ. Targeting the myostatin signaling pathway to treat muscle loss and metabolic dysfunction. J Clin Invest 2021; 131:148372. [PMID: 33938454 DOI: 10.1172/jci148372] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Since the discovery of myostatin (MSTN; also known as GDF-8) as a critical regulator of skeletal muscle mass in 1997, there has been an extensive effort directed at understanding the cellular and physiological mechanisms underlying MSTN activity, with the long-term goal of developing strategies and agents capable of blocking MSTN signaling to treat patients with muscle loss. Considerable progress has been made in elucidating key components of this regulatory system, and in parallel with this effort has been the development of numerous biologics that have been tested in clinical trials for a wide range of indications, including muscular dystrophy, sporadic inclusion body myositis, spinal muscular atrophy, cachexia, muscle loss due to aging or following falls, obesity, and type 2 diabetes. Here, I review what is known about the MSTN regulatory system and the current state of efforts to target this pathway for clinical applications.
Collapse
Affiliation(s)
- Se-Jin Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA.,University of Connecticut School of Medicine, Department of Genetics and Genome Sciences, Farmington, Connecticut, USA
| |
Collapse
|
19
|
Fernández-Garibay X, Ortega MA, Cerro-Herreros E, Comelles J, Martínez E, Artero R, Fernández-Costa JM, Ramón-Azcón J. Bioengineered in vitro3D model of myotonic dystrophy type 1 human skeletal muscle. Biofabrication 2021; 13. [PMID: 33836519 DOI: 10.1088/1758-5090/abf6ae] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/09/2021] [Indexed: 02/06/2023]
Abstract
Myotonic dystrophy type 1 (DM1) is the most common hereditary myopathy in the adult population. The disease is characterized by progressive skeletal muscle degeneration that produces severe disability. At present, there is still no effective treatment for DM1 patients, but the breakthroughs in understanding the molecular pathogenic mechanisms in DM1 have allowed the testing of new therapeutic strategies. Animal models andin vitrotwo-dimensional cell cultures have been essential for these advances. However, serious concerns exist regarding how faithfully these models reproduce the biological complexity of the disease. Biofabrication tools can be applied to engineer human three-dimensional (3D) culture systems that complement current preclinical research models. Here, we describe the development of the firstin vitro3D model of DM1 human skeletal muscle. Transdifferentiated myoblasts from patient-derived fibroblasts were encapsulated in micromolded gelatin methacryloyl-carboxymethyl cellulose methacrylate hydrogels through photomold patterning on functionalized glass coverslips. These hydrogels present a microstructured topography that promotes myoblasts alignment and differentiation resulting in highly aligned myotubes from both healthy and DM1 cells in a long-lasting cell culture. The DM1 3D microtissues recapitulate the molecular alterations detected in patient biopsies. Importantly, fusion index analyses demonstrate that 3D micropatterning significantly improved DM1 cell differentiation into multinucleated myotubes compared to standard cell cultures. Moreover, the characterization of the 3D cultures of DM1 myotubes detects phenotypes as the reduced thickness of myotubes that can be used for drug testing. Finally, we evaluated the therapeutic effect of antagomiR-23b administration on bioengineered DM1 skeletal muscle microtissues. AntagomiR-23b treatment rescues both molecular DM1 hallmarks and structural phenotype, restoring myotube diameter to healthy control sizes. Overall, these new microtissues represent an improvement over conventional cell culture models and can be used as biomimetic platforms to establish preclinical studies for myotonic dystrophy.
Collapse
Affiliation(s)
- Xiomara Fernández-Garibay
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| | - María A Ortega
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| | - Estefanía Cerro-Herreros
- University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Translational Genomics Group, Incliva Health Research Institute, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Joint Unit Incliva- CIPF, Dr Moliner 50, E46100 Burjassot, Valencia, Spain
| | - Jordi Comelles
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain.,Department of Electronics and Biomedical Engineering, University of Barcelona (UB), c/Martí i Franquès 1-11, E08028 Barcelona, Spain
| | - Elena Martínez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain.,Department of Electronics and Biomedical Engineering, University of Barcelona (UB), c/Martí i Franquès 1-11, E08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red (CIBER), Av. Monforte de Lemos 3-5, Pabellón 11, Planta 0, E28029 Madrid, Spain
| | - Rubén Artero
- University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Translational Genomics Group, Incliva Health Research Institute, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Joint Unit Incliva- CIPF, Dr Moliner 50, E46100 Burjassot, Valencia, Spain
| | - Juan M Fernández-Costa
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| | - Javier Ramón-Azcón
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain.,Institució Catalana de Reserca I Estudis Avançats (ICREA), Passeig de Lluís Companys, 23, E08010 Barcelona, Spain
| |
Collapse
|
20
|
Joseph J, Doles JD. Disease-associated metabolic alterations that impact satellite cells and muscle regeneration: perspectives and therapeutic outlook. Nutr Metab (Lond) 2021; 18:33. [PMID: 33766031 PMCID: PMC7992337 DOI: 10.1186/s12986-021-00565-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 03/15/2021] [Indexed: 11/10/2022] Open
Abstract
Many chronic disease patients experience a concurrent loss of lean muscle mass. Skeletal muscle is a dynamic tissue maintained by continuous protein turnover and progenitor cell activity. Muscle stem cells, or satellite cells, differentiate (by a process called myogenesis) and fuse to repair and regenerate muscle. During myogenesis, satellite cells undergo extensive metabolic alterations; therefore, pathologies characterized by metabolic derangements have the potential to impair myogenesis, and consequently exacerbate skeletal muscle wasting. How disease-associated metabolic disruptions in satellite cells might be contributing to wasting is an important question that is largely neglected. With this review we highlight the impact of various metabolic disruptions in disease on myogenesis and skeletal muscle regeneration. We also discuss metabolic therapies with the potential to improve myogenesis, skeletal muscle regeneration, and ultimately muscle mass.
Collapse
Affiliation(s)
- Josiane Joseph
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, MN, USA
| | - Jason D Doles
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
21
|
Barlow J, Sfyri PP, Mitchell R, Verpoorten S, Scully D, Andreou C, Papadopoulos P, Patel K, Matsakas A. Platelet releasate normalises the compromised muscle regeneration in a mouse model of hyperlipidaemia. Exp Physiol 2021; 106:700-713. [PMID: 33450106 DOI: 10.1113/ep088937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/12/2021] [Indexed: 12/19/2022]
Abstract
NEW FINDINGS What is the central question of this study? What is the impact of obesity-independent hyperlipidaemia on skeletal muscle stem cell function of ApoE-deficient (ApoE-/- ) mice? What is the main finding and its importance? Compromised muscle stem cell function accounts for the impaired muscle regeneration in hyperlipidaemic ApoE-/- mice. Importantly, impaired muscle regeneration is normalised by administration of platelet releasate. ABSTRACT Muscle satellite cells are important stem cells for skeletal muscle regeneration and repair after injury. ApoE-deficient mice, an established mouse model of hyperlipidaemia and atherosclerosis, show evidence of oxidative stress-induced lesions and fat infiltration in skeletal muscle followed by impaired repair after injury. However, the mechanisms underpinning attenuated muscle regeneration remain to be fully defined. Key to addressing the latter is to understand the properties of muscle stem cells from ApoE-deficient mice and their myogenic potential. Muscle stem cells from ApoE-deficient mice were cultured both ex vivo (on single fibres) and in vitro (primary myoblasts) and their myogenic capacity was determined. Skeletal muscle regeneration was studied on days 5 and 10 after cardiotoxin injury. ApoE-deficient muscle stem cells showed delayed activation and differentiation on single muscle fibres ex vivo. Impaired proliferation and differentiation profiles were also evident on isolated primary muscle stem cells in culture. ApoE-deficient mice displayed impaired skeletal muscle regeneration after acute injury in vivo. Administration of platelet releasate in ApoE-deficient mice reversed the deficits of muscle regeneration after acute injury to wild-type levels. These findings indicate that muscle stem cell myogenic potential is perturbed in skeletal muscle of a mouse model of hyperlipidaemia. We propose that platelet releasate could be a therapeutic intervention for conditions with associated myopathy such as peripheral arterial disease.
Collapse
Affiliation(s)
- Joseph Barlow
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Pagona Panagiota Sfyri
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Rob Mitchell
- School of Biological Sciences, University of Reading, Reading, UK
| | - Sandrine Verpoorten
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - David Scully
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Charalampos Andreou
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Petros Papadopoulos
- Department of Hematology, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| |
Collapse
|
22
|
Jaitovich A. Hypercapnic Respiratory Failure-Driven Skeletal Muscle Dysfunction: It Is Time for Animal Model-Based Mechanistic Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:129-138. [PMID: 33788191 DOI: 10.1007/978-3-030-63046-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Dysfunction of locomotor muscles is frequent in chronic pulmonary diseases and strongly associated with worse outcomes including higher mortality. Although these associations have been corroborated over the last decades, there is poor mechanistic understanding of the process, in part due to the lack of adequate animal models to investigate this process. Most of the mechanistic research has so far been accomplished using relevant individual stimuli such as low oxygen or high CO2 delivered to otherwise healthy animals as surrogates of the phenomena occurring in the clinical setting. This review advocates for the development of a syndromic model in which skeletal muscle dysfunction is investigated as a comorbidity of a well-validated pulmonary disease model, which could potentially allow discovering meaningful mechanisms and pathways and lead to more substantial progress to treat this devastating condition.
Collapse
Affiliation(s)
- Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY, USA. .,Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
23
|
Meys R, Stoffels AAF, de Brandt J, van Hees HWH, Franssen FME, Sillen MJH, Wouters EFM, Burtin C, Klijn P, Bij de Vaate E, van den Borst B, Otker JM, Donkers J, Schleich FN, Hayot M, Pomiès P, Everaert I, Derave W, Spruit MA. Beta-alanine supplementation in patients with COPD receiving non-linear periodised exercise training or neuromuscular electrical stimulation: protocol of two randomised, double-blind, placebo-controlled trials. BMJ Open 2020; 10:e038836. [PMID: 32928863 PMCID: PMC7488791 DOI: 10.1136/bmjopen-2020-038836] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Exercise intolerance is common in patients with chronic obstructive pulmonary disease (COPD) and, although multifactorial, it is largely caused by lower-limb muscle dysfunction. Research has shown that patients with severe to very severe COPD have significantly lower levels of muscle carnosine, which acts as a pH buffer and antioxidant. Beta-alanine (BA) supplementation has been shown to consistently elevate muscle carnosine in a variety of populations and may therefore improve exercise tolerance and lower-limb muscle function. The primary objective of the current studies is to assess the beneficial effects of BA supplementation in enhancing exercise tolerance on top of two types of exercise training (non-linear periodised exercise (NLPE) training or neuromuscular electrical stimulation (NMES)) in patients with COPD. METHODS AND ANALYSIS Two randomised, double-blind, placebo-controlled trials have been designed. Patients will routinely receive either NLPE (BASE-TRAIN trial) or NMES (BASE-ELECTRIC trial) as part of standard exercise-based care during their 8-to-10 week pulmonary rehabilitation (PR) programme. A total of 222 patients with COPD (2×77 = 154 patients in the BASE-TRAIN trial and 2×34 = 68 patients in the BASE-ELECTRIC trial) will be recruited from two specialised PR centres in The Netherlands. For study purposes, patients will receive 3.2 g of oral BA supplementation or placebo per day. Exercise tolerance is the primary outcome, which will be assessed using the endurance shuttle walk test (BASE-TRAIN) or the constant work rate cycle test (BASE-ELECTRIC). Furthermore, quadriceps muscle strength and endurance, cognitive function, carnosine levels (in muscle), BA levels (in blood and muscle), markers of oxidative stress and inflammation (in blood, muscles and lungs), physical activity and quality of life will be measured. ETHICS AND DISSEMINATION Both trials were approved by CMO Regio Arnhem-Nijmegen, The Netherlands (NL70781.091.19. and NL68757.091.19). TRIAL REGISTRATION NUMBER NTR8427 (BASE-TRAIN) and NTR8419 (BASE-ELECTRIC).
Collapse
Affiliation(s)
- Roy Meys
- Department of Research and Development, CIRO, Horn, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Anouk A F Stoffels
- Department of Research and Development, CIRO, Horn, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
- Department of Pulmonary Diseases, Radboud UMC Dekkerswald, Nijmegen, The Netherlands
| | - Jana de Brandt
- Reval Rehabilitation Research, Biomedical Research Institute, Faculty of Rehabilitation Sciences, Hasselt University, DIepenbeek, Belgium
| | | | - Frits M E Franssen
- Department of Research and Development, CIRO, Horn, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | | | - Emiel F M Wouters
- Department of Research and Development, CIRO, Horn, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Chris Burtin
- Reval Rehabilitation Research, Biomedical Research Institute, Faculty of Rehabilitation Sciences, Hasselt University, DIepenbeek, Belgium
| | - Peter Klijn
- Department of Pulmonology, Merem Pulmonary Rehabilitation Centre, Hilversum, The Netherlands
- Department of Pulmonary Medicine, Amsterdam UMC, Amsterdam, The Netherlands
| | - Eline Bij de Vaate
- Department of Pulmonology, Merem Pulmonary Rehabilitation Centre, Hilversum, The Netherlands
| | - Bram van den Borst
- Department of Pulmonary Diseases, Radboud UMC Dekkerswald, Nijmegen, The Netherlands
| | - Jacqueline M Otker
- Patient Advisory Council, Lung Foundation Netherlands, Amersfoort, The Netherlands
- Client Council, CIRO, Horn, The Netherlands
| | | | - Florence N Schleich
- Department of Respiratory Medicine, CHU Sart-Tilman Liege, GIGA I3, Liege, Belgium
| | - Maurice Hayot
- PhyMedExp, INSERM - CNRS, University of Montpellier - Montpellier CHU, Montpellier, France
| | - Pascal Pomiès
- PhyMedExp, INSERM - CNRS, University of Montpellier - Montpellier CHU, Montpellier, France
| | - Inge Everaert
- Department of Movement and Sport Sciences, University Ghent, Ghent, Belgium
| | - Wim Derave
- Department of Movement and Sport Sciences, University Ghent, Ghent, Belgium
| | - Martijn A Spruit
- Department of Research and Development, CIRO, Horn, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
- Reval Rehabilitation Research, Biomedical Research Institute, Faculty of Rehabilitation Sciences, Hasselt University, DIepenbeek, Belgium
| |
Collapse
|
24
|
Corvelyn M, De Beukelaer N, Duelen R, Deschrevel J, Van Campenhout A, Prinsen S, Gayan-Ramirez G, Maes K, Weide G, Desloovere K, Sampaolesi M, Costamagna D. Muscle Microbiopsy to Delineate Stem Cell Involvement in Young Patients: A Novel Approach for Children With Cerebral Palsy. Front Physiol 2020; 11:945. [PMID: 32848872 PMCID: PMC7424076 DOI: 10.3389/fphys.2020.00945] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/14/2020] [Indexed: 12/18/2022] Open
Abstract
Cerebral palsy (CP), the single largest cause of childhood physical disability, is characterized firstly by a lesion in the immature brain, and secondly by musculoskeletal problems that progress with age. Previous research reported altered muscle properties, such as reduced volume and satellite cell (SC) numbers and hypertrophic extracellular matrix compared to typically developing (TD) children (>10 years). Unfortunately, data on younger CP patients are scarce and studies on SCs and other muscle stem cells in CP are insufficient or lacking. Therefore, it remains difficult to understand the early onset and trajectory of altered muscle properties in growing CP children. Because muscle stem cells are responsible for postnatal growth, repair and remodeling, multiple adult stem cell populations from young CP children could play a role in altered muscle development. To this end, new methods for studying muscle samples of young children, valid to delineate the features and to elucidate the regenerative potential of muscle tissue, are necessary. Using minimal invasive muscle microbiopsy, which was applied in young subjects under general anaesthesia for the first time, we aimed to isolate and characterize muscle stem cell-derived progenitors of TD children and patients with CP. Data of 15 CP patients, 3–9 years old, and 5 aged-matched TD children were reported. The muscle microbiopsy technique was tolerated well in all participants. Through the explant technique, we provided muscle stem cell-derived progenitors from the Medial Gastrocnemius. Via fluorescent activated cell sorting, using surface markers CD56, ALP, and PDGFRa, we obtained SC-derived progenitors, mesoangioblasts and fibro-adipogenic progenitors, respectively. Adipogenic, skeletal, and smooth muscle differentiation assays confirmed the cell identity and ability to give rise to different cell types after appropriate stimuli. Myogenic differentiation in CP SC-derived progenitors showed enhanced fusion index and altered myotube formation based on MYOSIN HEAVY CHAIN expression, as well as disorganization of nuclear spreading, which were not observed in TD myotubes. In conclusion, the microbiopsy technique allows more focused muscle research in young CP patients. Current results show altered differentiation abilities of muscle stem cell-derived progenitors and support the hypothesis of their involvement in CP-altered muscle growth.
Collapse
Affiliation(s)
- Marlies Corvelyn
- Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Nathalie De Beukelaer
- Neurorehabilitation Group, Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
| | - Robin Duelen
- Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Jorieke Deschrevel
- Laboratory of Respiratory Disease and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Anja Van Campenhout
- Pediatric Orthopedics, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Sandra Prinsen
- Pediatric Orthopedics, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Ghislaine Gayan-Ramirez
- Laboratory of Respiratory Disease and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Karen Maes
- Laboratory of Respiratory Disease and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Guido Weide
- Neurorehabilitation Group, Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium.,Laboratory of Respiratory Disease and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Kaat Desloovere
- Neurorehabilitation Group, Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
| | - Maurilio Sampaolesi
- Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Domiziana Costamagna
- Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Neurorehabilitation Group, Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Catteau M, Gouzi F, Blervaque L, Passerieux E, Blaquière M, Ayoub B, Bughin F, Mercier J, Hayot M, Pomiès P. Effects of a human microenvironment on the differentiation of human myoblasts. Biochem Biophys Res Commun 2020; 525:968-973. [DOI: 10.1016/j.bbrc.2020.03.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 03/02/2020] [Indexed: 11/29/2022]
|
26
|
Su J, Li J, Lu Y, Li N, Li P, Wang Z, Wu W, Liu X. The rat model of COPD skeletal muscle dysfunction induced by progressive cigarette smoke exposure: a pilot study. BMC Pulm Med 2020; 20:74. [PMID: 32293377 PMCID: PMC7092612 DOI: 10.1186/s12890-020-1109-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/09/2020] [Indexed: 12/13/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) skeletal muscle dysfunction is a prevalent malady that significantly affects patients’ prognosis and quality of life. Although the study of this disease has attracted considerable attention, a definite animal model is yet to be established. This study investigates whether smoke exposure could lead to the development of a COPD skeletal muscle dysfunction model in rats. Methods Sprague Dawley rats were randomly divided into model (MG, n = 8) and control groups (CG, n = 6). The MG was exposed to cigarette smoke for 16 weeks while the CG was not. The body weight and forelimb grip strength of rats were monitored monthly. The pulmonary function and the strength of tibialis anterior muscle were assessed in vitro and compared after establishing the model. The histological changes in lung and gastrocnemius muscles were observed. The expressions of interleukin (IL)-6, IL-8, and tumor necrosis factor (TNF)-α were detected by ELISA, while the expressions of Atrogin-1 and MuRF1 in the gastrocnemius muscle were determined by Western blotting. Results Smoke exposure slowly increases the body weight and forelimb grip strength of MG rats, compared to CG rats. However, it significantly decreases the pulmonary ventilation function and the skeletal muscle contractility of the MG in vitro. Histologically, the lung tissues of MG show typical pathological manifestations of emphysema, while the skeletal muscles present muscular atrophy. The expressions of IL-6, IL-8, and TNF-α in MG rats are significantly higher than those measured in CG rats. Increased levels of Atrogin-1 and MuRF1 were also detected in the gastrocnemius muscle tissue of MG. Conclusion Progressive smoking exposure decreases the contractile function of skeletal muscles, leading to muscular atrophy. It also increases the expressions of inflammatory and muscle protein degradation factors in COPD rats. This indicates that smoke exposure could be used to establish a COPD skeletal muscle dysfunction model in rats.
Collapse
Affiliation(s)
- Jianqing Su
- Department of Sports Medicine, Shanghai University of Sport, Shanghai, 200438, China
| | - Jian Li
- Department of Sports Medicine, Shanghai University of Sport, Shanghai, 200438, China
| | - Yufan Lu
- Department of Sports Medicine, Shanghai University of Sport, Shanghai, 200438, China
| | - Ning Li
- Department of Sports Medicine, Shanghai University of Sport, Shanghai, 200438, China
| | - Peijun Li
- Department of Sports Medicine, Shanghai University of Sport, Shanghai, 200438, China
| | - Zhengrong Wang
- Department of Sports Medicine, Shanghai University of Sport, Shanghai, 200438, China
| | - Weibing Wu
- Department of Sports Medicine, Shanghai University of Sport, Shanghai, 200438, China.
| | - Xiaodan Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Institute of Rehabilitation Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
27
|
Kargl CK, Nie Y, Evans S, Stout J, Shannahan JH, Kuang S, Gavin TP. Factors secreted from high glucose treated endothelial cells impair expansion and differentiation of human skeletal muscle satellite cells. J Physiol 2019; 597:5109-5124. [DOI: 10.1113/jp278165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/26/2019] [Indexed: 12/12/2022] Open
Affiliation(s)
| | - Yaohui Nie
- Department of Health and KinesiologyPurdue University
| | | | | | | | - Shihuan Kuang
- Department of Animal SciencesPurdue University West Lafayette IN USA
| | | |
Collapse
|
28
|
Jaitovich A, Barreiro E. Skeletal Muscle Dysfunction in Chronic Obstructive Pulmonary Disease. What We Know and Can Do for Our Patients. Am J Respir Crit Care Med 2019; 198:175-186. [PMID: 29554438 DOI: 10.1164/rccm.201710-2140ci] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle dysfunction occurs in patients with chronic obstructive pulmonary disease (COPD) and affects both ventilatory and nonventilatory muscle groups. It represents a very important comorbidity that is associated with poor quality of life and reduced survival. It results from a complex combination of functional, metabolic, and anatomical alterations leading to suboptimal muscle work. Muscle atrophy, altered fiber type and metabolism, and chest wall remodeling, in the case of the respiratory muscles, are relevant etiological contributors to this process. Muscle dysfunction worsens during COPD exacerbations, rendering patients progressively less able to perform activities of daily living, and it is also associated with poor outcomes. Muscle recovery measures consisting of a combination of pulmonary rehabilitation, optimized nutrition, and other strategies are associated with better prognosis when administered in stable patients as well as after exacerbations. A deeper understanding of this process' pathophysiology and clinical relevance will facilitate the use of measures to alleviate its effects and potentially improve patients' outcomes. In this review, a general overview of skeletal muscle dysfunction in COPD is offered to highlight its relevance and magnitude to expert practitioners and scientists as well as to the average clinician dealing with patients with chronic respiratory diseases.
Collapse
Affiliation(s)
- Ariel Jaitovich
- 1 Division of Pulmonary and Critical Care Medicine and.,2 Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Esther Barreiro
- 3 Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, Institut Hospital del Mar d'Investigacions Mèdiques-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, Barcelona, Spain; and.,4 Centro de Investigación en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
29
|
Bensaid S, Fabre C, Fourneau J, Cieniewski-Bernard C. Impact of different methods of induction of cellular hypoxia: focus on protein homeostasis signaling pathways and morphology of C2C12 skeletal muscle cells differentiated into myotubes. J Physiol Biochem 2019; 75:367-377. [PMID: 31267382 DOI: 10.1007/s13105-019-00687-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/15/2019] [Indexed: 12/25/2022]
Abstract
Hypoxia, occurring in several pathologies, has deleterious effects on skeletal muscle, in particular on protein homeostasis. Different induction methods of hypoxia are commonly used in cellular models to investigate the alterations of muscular function consecutive to hypoxic stress. However, a consensus is not clearly established concerning hypoxia induction methodology. Our aim was to compare oxygen deprivation with chemically induced hypoxia using cobalt chloride (CoCl2) or desferrioxamine (DFO) on C2C12 myotubes which were either cultured in hypoxia chamber at an oxygen level of 4% or treated with CoCl2 or DFO. For each method of hypoxia induction, we determined their impact on muscle cell morphology and on expression or activation status of key signaling proteins of synthesis and degradation pathways. The expression of HIF-1α increased whatever the method of hypoxia induction. Myotube diameter and protein content decreased exclusively for C2C12 myotubes submitted to physiological hypoxia (4% O2) or treated with CoCl2. Results were correlated with a hypophosphorylation of key proteins regulated synthesis pathway (Akt, GSK3-β and P70S6K). Similarly, the phosphorylation of FoxO1 decreased and the autophagy-related LC3-II was overexpressed with 4% O2 and CoCl2 conditions. Our results demonstrated that in vitro oxygen deprivation and the use of mimetic agent such as CoCl2, unlike DFO, induced similar responses on myotube morphology and atrophy/hypertrophy markers. Thus, physiological hypoxia or its artificial induction using CoCl2 can be used to understand finely the molecular changes in skeletal muscle cells and to evaluate new therapeutics for hypoxia-related muscle disorders.
Collapse
Affiliation(s)
- Samir Bensaid
- Team Physical Activity, Muscle, Health, University Lille - EA 7369 - URePSSS, 59000, Lille, France.,Research Pole, CHU Lille, 59000, Lille, France
| | - Claudine Fabre
- Team Physical Activity, Muscle, Health, University Lille - EA 7369 - URePSSS, 59000, Lille, France
| | - Julie Fourneau
- Team Physical Activity, Muscle, Health, University Lille - EA 7369 - URePSSS, 59000, Lille, France
| | | |
Collapse
|
30
|
CD34 regulates the skeletal muscle response to hypoxia. J Muscle Res Cell Motil 2019; 40:309-318. [PMID: 31222587 DOI: 10.1007/s10974-019-09525-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 06/13/2019] [Indexed: 01/25/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) can sometimes be associated with skeletal muscle atrophy. Hypoxemic episodes, which occur during disease exacerbation and daily physical activity, are frequent in COPD patients. However, the link between hypoxemia and muscle atrophy remains unclear, along with mechanisms of muscle hypoxic stress response. Myogenic progenitors (MPs) and fibro/adipogenic progenitors (FAPs) express CD34 and participate to muscle mass maintenance. Although there is evidence linking CD34 expression and muscle repair, the link between CD34 expression, muscle wasting and the hypoxic stress observed in COPD has never been studied. Using a 2-day model of exposure to hypoxic conditions, we investigated the impact of hypoxia on skeletal muscle wasting and function, and elucidated the importance of CD34 expression in that response. A 2-day exposure to hypoxic conditions induces muscle atrophy, which was significantly worse in Cd34-/- mice compared to wild type (WT). Moreover, the lack of CD34 expression negatively impacts the maximal strength of the extensor digitorum longus muscle in response to hypoxia. Following exposure to hypoxic conditions, FAPs (which support MPs differentiation and myogenesis) are significantly lower in Cd34-/- mice compared to WT animals while the expression of myogenic regulatory factors and degradation factors (Atrogin) are similar. CD34 expression is important in the maintenance of muscle mass and function in response to hypoxic stress. These results highlight a new potential role for CD34 in muscle mass maintenance in hypoxic stress such as observed in COPD.
Collapse
|
31
|
Gea J, Martínez-Llorens J. Muscle Dysfunction in Chronic Obstructive Pulmonary Disease: Latest Developments. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.arbr.2018.07.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
32
|
Marchetti N. Anabolic Medications for Muscle Wasting in Chronic Obstructive Pulmonary Disease. Is the Evidence Getting Stronger? Am J Respir Crit Care Med 2019; 199:256-258. [DOI: 10.1164/rccm.201807-1327ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Nathaniel Marchetti
- Department of Thoracic Medicine and SurgeryLewis Katz School of Medicine at Temple UniversityPhiladelphia, Pennsylvania
| |
Collapse
|
33
|
Polkey MI, Praestgaard J, Berwick A, Franssen FME, Singh D, Steiner MC, Casaburi R, Tillmann HC, Lach-Trifilieff E, Roubenoff R, Rooks DS. Activin Type II Receptor Blockade for Treatment of Muscle Depletion in Chronic Obstructive Pulmonary Disease. A Randomized Trial. Am J Respir Crit Care Med 2019; 199:313-320. [PMID: 30095981 PMCID: PMC6363975 DOI: 10.1164/rccm.201802-0286oc] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 08/10/2018] [Indexed: 02/07/2023] Open
Abstract
RATIONALE Bimagrumab is a fully human monoclonal antibody that blocks the activin type II receptors, preventing the activity of myostatin and other negative skeletal muscle regulators. OBJECTIVES To assess the effects of bimagrumab on skeletal muscle mass and function in patients with chronic obstructive pulmonary disease (COPD) and reduced skeletal muscle mass. METHODS Sixty-seven patients with COPD (mean FEV1, 1.05 L [41.6% predicted]; aged 40-80 yr; body mass index < 20 kg/m2 or appendicular skeletal muscle mass index ≤ 7.25 [men] and ≤ 5.67 [women] kg/m2), received two doses of either bimagrumab 30 mg/kg intravenously (n = 33) or placebo (n = 34) (Weeks 0 and 8) over 24 weeks. MEASUREMENTS AND MAIN RESULTS We assessed changes in thigh muscle volume (cubic centimeters) as the primary endpoint along with 6-minute-walk distance (meters), safety, and tolerability. Fifty-five (82.1%) patients completed the study. Thigh muscle volume increased by Week 4 and remained increased at Week 24 in bimagrumab-treated patients, whereas no changes were observed with placebo (Week 4: +5.9% [SD, 3.4%] vs. 0.0% [3.3%], P < 0.001; Week 8: +7.0% [3.7%] vs. -0.7% [2.8%], P < 0.001; Week 16: +7.8% [5.1%] vs. -0.9% [4.5%], P < 0.001; Week 24: +5.0% [4.9%] vs. -1.3% [4.3%], P < 0.001). Over 24 weeks, 6-minute-walk distance did not increase significantly in either group. Adverse events in the bimagrumab group included muscle-related symptoms, diarrhea, and acne, most of which were mild in severity. CONCLUSIONS Blocking the action of negative muscle regulators through the activin type II receptors with bimagrumab treatment safely increased skeletal muscle mass but did not improve functional capacity in patients with COPD and low muscle mass. Clinical trial registered with www.clinicaltrials.gov (NCT01669174).
Collapse
Affiliation(s)
- Michael I. Polkey
- National Institute for Health Research Respiratory Biomedical Research Unit, Royal Brompton and Harefield National Health Service Foundation Trust and Imperial College London, London, United Kingdom
| | | | - Amy Berwick
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Frits M. E. Franssen
- Department of Research and Education, CIRO, Center of Expertise for Chronic Organ Failure, Horn, the Netherlands
| | - Dave Singh
- Centre for Respiratory Medicine and Allergy, University of Manchester and the Medicines Evaluation Unit, University Hospital of South Manchester National Health Service Foundation Trust, Manchester, United Kingdom
| | - Michael C. Steiner
- Centre for Exercise and Rehabilitation Science, National Institute for Health Research Leicester Biomedical Research Centre, Respiratory, Glenfield Hospital, Leicester, United Kingdom
| | - Richard Casaburi
- Rehabilitation Clinical Trials Center, Los Angeles Biomedical Research Institute, Harbor-University of California Los Angeles Medical Center, Torrance, California; and
| | | | | | | | - Daniel S. Rooks
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| |
Collapse
|
34
|
Coppolino I, Ruggeri P, Nucera F, Cannavò MF, Adcock I, Girbino G, Caramori G. Role of Stem Cells in the Pathogenesis of Chronic Obstructive Pulmonary Disease and Pulmonary Emphysema. COPD 2018; 15:536-556. [DOI: 10.1080/15412555.2018.1536116] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Irene Coppolino
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Paolo Ruggeri
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Francesco Nucera
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Mario Francesco Cannavò
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Ian Adcock
- Airways Disease Section, National Heart and Lung Institute, Royal Brompton Hospital Biomedical Research Unit, Imperial College, London, UK
| | - Giuseppe Girbino
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Gaetano Caramori
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| |
Collapse
|
35
|
Gea J, Martínez-Llorens J. Muscle Dysfunction in Chronic Obstructive Pulmonary Disease: Latest Developments. Arch Bronconeumol 2018; 55:237-238. [PMID: 30297244 DOI: 10.1016/j.arbres.2018.07.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/17/2018] [Accepted: 07/17/2018] [Indexed: 11/24/2022]
Affiliation(s)
- Joaquim Gea
- Servicio de Neumología, Hospital del Mar-IMIM, DCEXS, Universitat Pompeu Fabra. CIBERES, ISCiii, Barcelona, España.
| | - Juana Martínez-Llorens
- Servicio de Neumología, Hospital del Mar-IMIM, DCEXS, Universitat Pompeu Fabra. CIBERES, ISCiii, Barcelona, España
| |
Collapse
|
36
|
Gouzi F, Blaquière M, Catteau M, Bughin F, Maury J, Passerieux E, Ayoub B, Mercier J, Hayot M, Pomiès P. Oxidative stress regulates autophagy in cultured muscle cells of patients with chronic obstructive pulmonary disease. J Cell Physiol 2018; 233:9629-9639. [PMID: 29943813 DOI: 10.1002/jcp.26868] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 05/23/2018] [Indexed: 12/21/2022]
Abstract
The proteolytic autophagy pathway is enhanced in the lower limb muscles of patients with chronic obstructive pulmonary disease (COPD). Reactive oxygen species (ROS) have been shown to regulate autophagy in the skeletal muscles, but the role of oxidative stress in the muscle autophagy of patients with COPD is unknown. We used cultured myoblasts and myotubes from the quadriceps of eight healthy subjects and twelve patients with COPD (FEV1% predicted: 102.0% and 32.0%, respectively; p < 0.0001). We compared the autophagosome formation, the expression of autophagy markers, and the autophagic flux in healthy subjects and the patients with COPD, and we evaluated the effects of the 3-methyladenine (3-MA) autophagy inhibitor on the atrophy of COPD myotubes. Autophagy was also assessed in COPD myotubes treated with an antioxidant molecule, ascorbic acid. Autophagosome formation was increased in COPD myoblasts and myotubes (p = 0.011; p < 0.001), and the LC3 2/LC3 1 ratio (p = 0.002), SQSTM1 mRNA and protein expression (p = 0.023; p = 0.007), BNIP3 expression (p = 0.031), and autophagic flux (p = 0.002) were higher in COPD myoblasts. Inhibition of autophagy with 3-MA increased the COPD myotube diameter (p < 0.001) to a level similar to the diameter of healthy subject myotubes. Treatment of COPD myotubes with ascorbic acid decreased ROS concentration (p < 0.001), ROS-induced protein carbonylation (p = 0.019), the LC3 2/LC3 1 ratio (p = 0.037), the expression of SQSTM1 (p < 0.001) and BNIP3 (p < 0.001), and increased the COPD myotube diameter (p < 0.001). Thus, autophagy signaling is enhanced in cultured COPD muscle cells. Furthermore, the oxidative stress level contributes to the regulation of autophagy, which is involved in the atrophy of COPD myotubes in vitro.
Collapse
Affiliation(s)
- Fares Gouzi
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Department of Clinical Physiology, CHRU of Montpellier, Montpellier, France
| | - Marine Blaquière
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Department of Clinical Physiology, CHRU of Montpellier, Montpellier, France
| | - Matthias Catteau
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - François Bughin
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Department of Clinical Physiology, CHRU of Montpellier, Montpellier, France
| | - Jonathan Maury
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Clinique du Souffle "La Solane," Fontalvie/5-Santé Group, Osséja, France
| | - Emilie Passerieux
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Bronia Ayoub
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Department of Clinical Physiology, CHRU of Montpellier, Montpellier, France
| | - Jacques Mercier
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Department of Clinical Physiology, CHRU of Montpellier, Montpellier, France
| | - Maurice Hayot
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Department of Clinical Physiology, CHRU of Montpellier, Montpellier, France
| | - Pascal Pomiès
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| |
Collapse
|
37
|
Lu JJ, Wang Q, Xie LH, Zhang Q, Sun SH. Tumor necrosis factor-like weak inducer of apoptosis regulates quadriceps muscle atrophy and fiber-type alteration in a rat model of chronic obstructive pulmonary disease. Tob Induc Dis 2017; 15:43. [PMID: 29151827 PMCID: PMC5679159 DOI: 10.1186/s12971-017-0148-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 10/27/2017] [Indexed: 12/25/2022] Open
Abstract
Background In chronic obstructive pulmonary disease (COPD), weakness and muscle mass loss of the quadriceps muscle has been demonstrated to predict survival and mortality rates of patients. Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK), as a member of the TNF superfamily, has recently been identified as a key regulator of skeletal muscle wasting and metabolic dysfunction. So our aim was to study the role of TWEAK during quadriceps muscle atrophy and fiber-type transformation in COPD model rats and its possible pathway. Methods Forty-four healthy male adult Wistar rats were randomly divided into two groups: A normal control group (n = 16) and a COPD model group (n = 28). The COPD group was exposed to cigarette smoke for 90 d and injected with porcine pancreatic elastase on day 15, whereas the control group was injected with saline alone. Following treatment, weights of the quadriceps muscles were measured and hematoxylin and eosin staining was performed to identify structural changes in lung and quadriceps muscle tissue. Immunohistochemical staining was also conducted to determine the localization of TWEAK, nuclear factor (NF)-κB, muscle ring finger (MuRF)-1 and proliferator-activated coactivator (PGC)-1a proteins in the quadriceps muscle, and western blotting was used to assess the level of protein expression. Results Compared with controls, COPD model rats exhibited significantly lower quadriceps muscle weight (P < 0.05) accompanied by fiber atrophy and disordered fiber arrangement, a wide gap between adjacent muscle fibers, a significant reduction in nuclear number (P < 0.05) and an uneven size distribution. The proportion of fiber types was also significantly altered (P < 0.05). In addition, TWEAK expression in the quadriceps muscle of COPD model rats was significantly higher than that in control rats (P < 0.05), and was significantly associated with quadriceps atrophy and fiber-type alteration (P < 0.05). Levels of NF-κB, MuRF1 and PGC-1α expression also significantly differed between the two groups (P < 0.05). Conclusions Collectively these data suggest that increased levels of TWEAK may lead to skeletal muscle atrophy and fiber-type alteration, which in turn may be associated with activation of the ubiquitin-proteasome pathway, involving NF-κB, MuRF1 and PGC-1α as potential regulatory factors. These preliminary results in rats suggest that TWEAK may be a therapeutic target for the treatment of muscle atrophy in COPD.
Collapse
Affiliation(s)
- Jun-Juan Lu
- Department of Respiratory Medicine, The Third XiangYa Hospital of Central South University, 138 Tongzipo Road, Changsha, Hunan 410013 People's Republic of China
| | - Qing Wang
- Department of Respiratory Medicine, Changsha Central Hospital, Changsha, Hunan 410004 People's Republic of China
| | - Li Hua Xie
- Department of Respiratory Medicine, The Third XiangYa Hospital of Central South University, 138 Tongzipo Road, Changsha, Hunan 410013 People's Republic of China
| | - Qiang Zhang
- Department of Respiratory Medicine, The Third XiangYa Hospital of Central South University, 138 Tongzipo Road, Changsha, Hunan 410013 People's Republic of China
| | - Sheng Hua Sun
- Department of Respiratory Medicine, The Third XiangYa Hospital of Central South University, 138 Tongzipo Road, Changsha, Hunan 410013 People's Republic of China
| |
Collapse
|
38
|
Zhu M, Liu Z, Gao M, Zhang Y, Li Y, Ling S, Zhang P, Zhao C, Jiang L, Liu Y, Li Q, Li D, Hu S, Li Y. The effect of Bu Zhong Yi Qi decoction on simulated weightlessness‑induced muscle atrophy and its mechanisms. Mol Med Rep 2017; 16:5165-5174. [PMID: 28849026 PMCID: PMC5647051 DOI: 10.3892/mmr.2017.7287] [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: 08/10/2016] [Accepted: 05/11/2017] [Indexed: 12/17/2022] Open
Abstract
Microgravity has been previously demonstrated to induce skeletal muscle atrophy, loss of muscle force and disorders in myogenesis and metabolism. Current pharmacological strategies exhibit poor efficacy. Bu Zhong Yi Qi decoction (BZ) is a well-known traditional Chinese medicine decoction used for myasthenia gravis. In the present study, its effect on unloading induced muscle atrophy was investigated. The mousetail suspension model was used to simulate weightlessness induced muscle atrophy. The results indicated that BZ could significantly protect muscles from simulated weightlessness-induced atrophy. To elucidate the underlying mechanisms, drugCIPHER-CS methods were introduced to predict its potential targets, significantly enriched pathways and biological processes. The results demonstrated that the calcium signaling pathway, citrate cycle, biosynthetic and lipid metabolic process are affected by BZ. Among the targets, nuclear receptor corepressor 1 (NCoR1) is one of the most important proteins involved in myogenesis and metabolism. The results indicated that BZ significantly downregulated NCoR 1 expression, and further induced muscle differentiation and metabolism by regulating NCoR1-associated gene expression in vivo and in vitro. In summary, the present study indicated that may be effective in combating weightlessness-induced muscle atrophy. Combined with bioinformatics, the underlying mechanism for this decoction was investigated, which provided an improved understanding of this decoction.
Collapse
Affiliation(s)
- Mu Zhu
- Department of Chinese Materia Medica, School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing 100019, P.R. China
| | - Zhongyang Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Institute of Radiation Medicine, Beijing 102206, P.R. China
| | - Mingze Gao
- Department of Chinese Materia Medica, School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing 100019, P.R. China
| | - Yan Zhang
- Department of Chinese Materia Medica, School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing 100019, P.R. China
| | - Yuheng Li
- State Key Laboratory of Space Medicine Fundamentals and Applications, China Astronaut Research and Training Center, Beijing 100094, P.R. China
| | - Shukuan Ling
- State Key Laboratory of Space Medicine Fundamentals and Applications, China Astronaut Research and Training Center, Beijing 100094, P.R. China
| | - Pengfei Zhang
- State Key Laboratory of Space Medicine Fundamentals and Applications, China Astronaut Research and Training Center, Beijing 100094, P.R. China
| | - Chenyang Zhao
- State Key Laboratory of Space Medicine Fundamentals and Applications, China Astronaut Research and Training Center, Beijing 100094, P.R. China
| | - Lijun Jiang
- State Key Laboratory of Space Medicine Fundamentals and Applications, China Astronaut Research and Training Center, Beijing 100094, P.R. China
| | - Yu Liu
- State Key Laboratory of Space Medicine Fundamentals and Applications, China Astronaut Research and Training Center, Beijing 100094, P.R. China
| | - Qi Li
- State Key Laboratory of Space Medicine Fundamentals and Applications, China Astronaut Research and Training Center, Beijing 100094, P.R. China
| | - Dong Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Institute of Radiation Medicine, Beijing 102206, P.R. China
| | - Sumin Hu
- Department of Chinese Materia Medica, School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing 100019, P.R. China
| | - Yingxian Li
- State Key Laboratory of Space Medicine Fundamentals and Applications, China Astronaut Research and Training Center, Beijing 100094, P.R. China
| |
Collapse
|
39
|
Martin NRW, Aguilar-Agon K, Robinson GP, Player DJ, Turner MC, Myers SD, Lewis MP. Hypoxia Impairs Muscle Function and Reduces Myotube Size in Tissue Engineered Skeletal Muscle. J Cell Biochem 2017; 118:2599-2605. [PMID: 28294416 PMCID: PMC5518201 DOI: 10.1002/jcb.25982] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 02/21/2017] [Indexed: 12/21/2022]
Abstract
Contemporary tissue engineered skeletal muscle models display a high degree of physiological accuracy compared with native tissue, and therefore may be excellent platforms to understand how various pathologies affect skeletal muscle. Chronic obstructive pulmonary disease (COPD) is a lung disease which causes tissue hypoxia and is characterized by muscle fiber atrophy and impaired muscle function. In the present study we exposed engineered skeletal muscle to varying levels of oxygen (O2; 21–1%) for 24 h in order to see if a COPD like muscle phenotype could be recreated in vitro, and if so, at what degree of hypoxia this occurred. Maximal contractile force was attenuated in hypoxia compared to 21% O2; with culture at 5% and 1% O2 causing the most pronounced effects with 62% and 56% decrements in force, respectively. Furthermore at these levels of O2, myotubes within the engineered muscles displayed significant atrophy which was not seen at higher O2 levels. At the molecular level we observed increases in mRNA expression of MuRF‐1 only at 1% O2 whereas MAFbx expression was elevated at 10%, 5%, and 1% O2. In addition, p70S6 kinase phosphorylation (a downstream effector of mTORC1) was reduced when engineered muscle was cultured at 1% O2, with no significant changes seen above this O2 level. Overall, these data suggest that engineered muscle exposed to O2 levels of ≤5% adapts in a manner similar to that seen in COPD patients, and thus may provide a novel model for further understanding muscle wasting associated with tissue hypoxia. J. Cell. Biochem. 118: 2599–2605, 2017. © 2017 The Authors. Journal of Cellular Biochemistry Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Neil R W Martin
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Kathyrn Aguilar-Agon
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - George P Robinson
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Darren J Player
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Mark C Turner
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Stephen D Myers
- Department of Sport and Exercise Sciences, University of Chichester, Chichester, UK
| | - Mark P Lewis
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| |
Collapse
|
40
|
Chronic obstructive pulmonary disease and malnutrition in developing countries. Curr Opin Pulm Med 2017; 23:139-148. [DOI: 10.1097/mcp.0000000000000356] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
41
|
Abstract
With aging and other muscle wasting diseases, men and women undergo similar pathological changes in skeletal muscle: increased inflammation, enhanced oxidative stress, mitochondrial dysfunction, satellite cell senescence, elevated apoptosis and proteasome activity, and suppressed protein synthesis and myocyte regeneration. Decreased food intake and physical activity also indirectly contribute to muscle wasting. Sex hormones also play important roles in maintaining skeletal muscle homeostasis. Testosterone is a potent anabolic factor promoting muscle protein synthesis and muscular regeneration. Estrogens have a protective effect on skeletal muscle by attenuating inflammation; however, the mechanisms of estrogen action in skeletal muscle are less well characterized than those of testosterone. Age- and/or disease-induced alterations in sex hormones are major contributors to muscle wasting. Hence, men and women may respond differently to catabolic conditions because of their hormonal profiles. Here we review the similarities and differences between men and women with common wasting conditions including sarcopenia and cachexia due to cancer, end-stage renal disease/chronic kidney disease, liver disease, chronic heart failure, and chronic obstructive pulmonary disease based on the literature in clinical studies. In addition, the responses in men and women to the commonly used therapeutic agents and their efficacy to improve muscle mass and function are also reviewed.
Collapse
|
42
|
Emerging therapies for the treatment of skeletal muscle wasting in chronic obstructive pulmonary disease. Pharmacol Ther 2016; 166:56-70. [DOI: 10.1016/j.pharmthera.2016.06.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2016] [Indexed: 12/18/2022]
|
43
|
Chaillou T, Lanner JT. Regulation of myogenesis and skeletal muscle regeneration: effects of oxygen levels on satellite cell activity. FASEB J 2016; 30:3929-3941. [PMID: 27601440 DOI: 10.1096/fj.201600757r] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/15/2016] [Indexed: 12/11/2022]
Abstract
Reduced oxygen (O2) levels (hypoxia) are present during embryogenesis and exposure to altitude and in pathologic conditions. During embryogenesis, myogenic progenitor cells reside in a hypoxic microenvironment, which may regulate their activity. Satellite cells are myogenic progenitor cells localized in a local environment, suggesting that the O2 level could affect their activity during muscle regeneration. In this review, we present the idea that O2 levels regulate myogenesis and muscle regeneration, we elucidate the molecular mechanisms underlying myogenesis and muscle regeneration in hypoxia and depict therapeutic strategies using changes in O2 levels to promote muscle regeneration. Severe hypoxia (≤1% O2) appears detrimental for myogenic differentiation in vitro, whereas a 3-6% O2 level could promote myogenesis. Hypoxia impairs the regenerative capacity of injured muscles. Although it remains to be explored, hypoxia may contribute to the muscle damage observed in patients with pathologies associated with hypoxia (chronic obstructive pulmonary disease, and peripheral arterial disease). Hypoxia affects satellite cell activity and myogenesis through mechanisms dependent and independent of hypoxia-inducible factor-1α. Finally, hyperbaric oxygen therapy and transplantation of hypoxia-conditioned myoblasts are beneficial procedures to enhance muscle regeneration in animals. These therapies may be clinically relevant to treatment of patients with severe muscle damage.-Chaillou, T. Lanner, J. T. Regulation of myogenesis and skeletal muscle regeneration: effects of oxygen levels on satellite cell activity.
Collapse
Affiliation(s)
- Thomas Chaillou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
44
|
Involvement of the FoxO1/MuRF1/Atrogin-1 Signaling Pathway in the Oxidative Stress-Induced Atrophy of Cultured Chronic Obstructive Pulmonary Disease Myotubes. PLoS One 2016; 11:e0160092. [PMID: 27526027 PMCID: PMC4987766 DOI: 10.1371/journal.pone.0160092] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 07/13/2016] [Indexed: 12/16/2022] Open
Abstract
Oxidative stress is thought to be one of the most important mechanisms implicated in the muscle wasting of chronic obstructive pulmonary disease (COPD) patients, but its role has never been demonstrated. We therefore assessed the effects of both pro-oxidant and antioxidant treatments on the oxidative stress levels and atrophic signaling pathway of cultured COPD myotubes. Treatment of cultured COPD myotubes with the pro-oxidant molecule H2O2 resulted in increased ROS production (P = 0.002) and protein carbonylation (P = 0.050), in association with a more pronounced atrophy of the myotubes, as reflected by a reduced diameter (P = 0.003), and the activated expression of atrophic markers MuRF1 and FoxO1 (P = 0.022 and P = 0.030, respectively). Conversely, the antioxidant molecule ascorbic acid induced a reduction in ROS production (P<0.001) and protein carbonylation (P = 0.019), and an increase in the myotube diameter (P<0.001) to a level similar to the diameter of healthy subject myotubes, in association with decreased expression levels of MuRF1, atrogin-1 and FoxO1 (P<0.001, P = 0.002 and P = 0.042, respectively). A significant negative correlation was observed between the variations in myotube diameter and the variations in the expression of MuRF1 after antioxidant treatment (P = 0.047). Moreover, ascorbic acid was able to prevent the H2O2-induced atrophy of COPD myotubes. Last, the proteasome inhibitor MG132 restored the basal atrophy level of the COPD myotubes and also suppressed the H2O2-induced myotube atrophy. These findings demonstrate for the first time the involvement of oxidative stress in the atrophy of COPD peripheral muscle cells in vitro, via the FoxO1/MuRF1/atrogin-1 signaling pathway of the ubiquitin/proteasome system.
Collapse
|
45
|
Sente T, Van Berendoncks AM, Jonckheere AI, Rodenburg RJ, Lauwers P, Van Hoof V, Wouters A, Lardon F, Hoymans VY, Vrints CJ. Primary skeletal muscle myoblasts from chronic heart failure patients exhibit loss of anti-inflammatory and proliferative activity. BMC Cardiovasc Disord 2016; 16:107. [PMID: 27228977 PMCID: PMC4880810 DOI: 10.1186/s12872-016-0278-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 05/13/2016] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Peripheral skeletal muscle wasting is a common finding with adverse effects in chronic heart failure (HF). Whereas its clinical relevance is beyond doubt, the underlying pathophysiological mechanisms are not yet fully elucidated. We aimed to introduce and characterize the primary culture of skeletal muscle cells from individual HF patients as a supportive model to study this muscle loss. METHODS AND RESULTS Primary myoblast and myotubes cultures were successfully propagated from the m. vastus lateralis of 6 HF patients with reduced ejection fraction (HFrEF; LVEF <45 %) and 6 age and gender-matched healthy donors. HFrEF cultures were not different from healthy donors in terms of morphology, such as myoblast size, shape and actin microfilament. Differentiation and fusion indexes were identical between groups. Myoblast proliferation in logarithmic growth phase, however, was attenuated in the HFrEF group (p = 0.032). In addition, HFrEF myoblasts are characterized by a reduced TNFR2 expression and IL-6 secretion (p = 0.017 and p = 0.016; respectively). CONCLUSION Biopsy derived primary skeletal muscle myoblasts of HFrEF patients produce similar morphological and myogenic differentiation responses as myoblasts of healthy donors, though demonstrate loss of anti-inflammatory and proliferative activity.
Collapse
Affiliation(s)
- Tahnee Sente
- Laboratory of Cellular and Molecular Cardiology, Antwerp University Hospital, Edegem, Belgium. .,Department of Translational Pathophysiological Research, Cardiovascular Diseases, University of Antwerp, Wilrijk, Belgium.
| | | | - An I Jonckheere
- Department of Pediatric Neurology, UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Richard J Rodenburg
- Department of Pediatrics, Nijmegen Center for Mitochondrial Disorders, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Patrick Lauwers
- Department of Thoracic and Vascular Surgery, Antwerp University Hospital, Edegem, Belgium
| | - Viviane Van Hoof
- Department of Biochemistry, Antwerp University Hospital, Edegem, Belgium
| | - An Wouters
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Wilrijk, Belgium
| | - Filip Lardon
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Wilrijk, Belgium
| | - Vicky Y Hoymans
- Laboratory of Cellular and Molecular Cardiology, Antwerp University Hospital, Edegem, Belgium.,Department of Cardiology, Antwerp University Hospital, Edegem, Belgium
| | - Christiaan J Vrints
- Laboratory of Cellular and Molecular Cardiology, Antwerp University Hospital, Edegem, Belgium.,Department of Translational Pathophysiological Research, Cardiovascular Diseases, University of Antwerp, Wilrijk, Belgium.,Department of Cardiology, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
46
|
Gea J, Pascual S, Casadevall C, Orozco-Levi M, Barreiro E. Muscle dysfunction in chronic obstructive pulmonary disease: update on causes and biological findings. J Thorac Dis 2015; 7:E418-38. [PMID: 26623119 DOI: 10.3978/j.issn.2072-1439.2015.08.04] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Respiratory and/or limb muscle dysfunction, which are frequently observed in chronic obstructive pulmonary disease (COPD) patients, contribute to their disease prognosis irrespective of the lung function. Muscle dysfunction is caused by the interaction of local and systemic factors. The key deleterious etiologic factors are pulmonary hyperinflation for the respiratory muscles and deconditioning secondary to reduced physical activity for limb muscles. Nonetheless, cigarette smoke, systemic inflammation, nutritional abnormalities, exercise, exacerbations, anabolic insufficiency, drugs and comorbidities also seem to play a relevant role. All these factors modify the phenotype of the muscles, through the induction of several biological phenomena in patients with COPD. While respiratory muscles improve their aerobic phenotype (percentage of oxidative fibers, capillarization, mitochondrial density, enzyme activity in the aerobic pathways, etc.), limb muscles exhibit the opposite phenotype. In addition, both muscle groups show oxidative stress, signs of damage and epigenetic changes. However, fiber atrophy, increased number of inflammatory cells, altered regenerative capacity; signs of apoptosis and autophagy, and an imbalance between protein synthesis and breakdown are rather characteristic features of the limb muscles, mostly in patients with reduced body weight. Despite that significant progress has been achieved in the last decades, full elucidation of the specific roles of the target biological mechanisms involved in COPD muscle dysfunction is still required. Such an achievement will be crucial to adequately tackle with this relevant clinical problem of COPD patients in the near-future.
Collapse
Affiliation(s)
- Joaquim Gea
- Servei de Pneumologia, Muscle & Respiratory System Research Unit (URMAR), Hospital del Mar-I.M.I.M., Experimental Sciences and Health Department (CEXS), Universitat Pompeu Fabra, CIBERES, ISCIII, Barcelona, Catalonia, Spain
| | - Sergi Pascual
- Servei de Pneumologia, Muscle & Respiratory System Research Unit (URMAR), Hospital del Mar-I.M.I.M., Experimental Sciences and Health Department (CEXS), Universitat Pompeu Fabra, CIBERES, ISCIII, Barcelona, Catalonia, Spain
| | - Carme Casadevall
- Servei de Pneumologia, Muscle & Respiratory System Research Unit (URMAR), Hospital del Mar-I.M.I.M., Experimental Sciences and Health Department (CEXS), Universitat Pompeu Fabra, CIBERES, ISCIII, Barcelona, Catalonia, Spain
| | - Mauricio Orozco-Levi
- Servei de Pneumologia, Muscle & Respiratory System Research Unit (URMAR), Hospital del Mar-I.M.I.M., Experimental Sciences and Health Department (CEXS), Universitat Pompeu Fabra, CIBERES, ISCIII, Barcelona, Catalonia, Spain
| | - Esther Barreiro
- Servei de Pneumologia, Muscle & Respiratory System Research Unit (URMAR), Hospital del Mar-I.M.I.M., Experimental Sciences and Health Department (CEXS), Universitat Pompeu Fabra, CIBERES, ISCIII, Barcelona, Catalonia, Spain
| |
Collapse
|
47
|
Mattick CS, Landis AE, Allenby BR, Genovese NJ. Anticipatory Life Cycle Analysis of In Vitro Biomass Cultivation for Cultured Meat Production in the United States. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2015; 49:11941-11949. [PMID: 26383898 DOI: 10.1021/acs.est.5b01614] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Cultured, or in vitro, meat consists of edible biomass grown from animal stem cells in a factory, or carnery. In the coming decades, in vitro biomass cultivation could enable the production of meat without the need to raise livestock. Using an anticipatory life cycle analysis framework, the study described herein examines the environmental implications of this emerging technology and compares the results with published impacts of beef, pork, poultry, and another speculative analysis of cultured biomass. While uncertainty ranges are large, the findings suggest that in vitro biomass cultivation could require smaller quantities of agricultural inputs and land than livestock; however, those benefits could come at the expense of more intensive energy use as biological functions such as digestion and nutrient circulation are replaced by industrial equivalents. From this perspective, large-scale cultivation of in vitro meat and other bioengineered products could represent a new phase of industrialization with inherently complex and challenging trade-offs.
Collapse
Affiliation(s)
- Carolyn S Mattick
- School of Public Health, University of Texas , 1200 Hermann Pressler Drive, Houston, Texas 77030, United States
| | - Amy E Landis
- Glenn Department of Civil Engineering, Clemson University , Clemson, South Carolina 29634, United States
| | - Braden R Allenby
- School of Sustainable Engineering and the Built Environment and ASU Lincoln Center for Applied Ethics, Arizona State University , Tempe, Arizona 85287, United States
| | - Nicholas J Genovese
- Department of Medicine, University of Minnesota , Minneapolis, Minnesota 55455, United States
| |
Collapse
|