1
|
Liu Y, Chen H, Zhang Y, Shang Q, Zhao W, Zhang Y, Qiu W, Qin W, Lin F, He J, Liu H, Chen X, Gong Y, Liu L, Jiang Y, Ren H, Jiang X, Shen G. Plumbagin alleviates muscle atrophy in female mice through inhibiting the DANCR/NF-κB axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156282. [PMID: 39616734 DOI: 10.1016/j.phymed.2024.156282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/12/2024] [Accepted: 11/19/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Muscle atrophy is a condition of the skeletal muscular system closely related to inflammation and significantly affects a person's quality of life and physical activity. It is characterized primarily by the progressive loss of muscle mass, strength, and function. Plumbagin (PB), the main bioactive component of the traditional Chinese medicine Plumbago zeylanica L., has bFeen shown to treat various inflammatory diseases, such as osteoporosis, osteoarthritis, and sepsis. Furthermore, many biological processes, including inflammation, involve differentiation antagonistic nonprotein-coding RNA (DANCR). However, their role and clinical importance in myogenesis and amyotrophy are not well understood. PURPOSE This study aimed to explore the role of DANCR and the inflammatory response in the anti-muscle atrophy effects of PB. METHODS The expression of DANCR in muscle atrophic mice and during myogenic differentiation was examined using quantitative reverse transcription PCR (RT‒qPCR). The mechanism of DANCR in muscle atrophy was confirmed through gene knockdown, RNA sequencing (RNA-seq), RNA pull-down, RNA immunoprecipitation (RIP), immunofluorescence (IF), and luciferase reporter gene assays. Bioinformatics was utilized to investigate the mechanism by which PB treatment affects muscle atrophy. The relationship between PB and DANCR was verified by surface plasmon resonance (SPR) and RT‒qPCR. Additionally, the role of PB in muscle atrophy was explored through its control of DANCR-mediated regulation of the NF-κB pathway. Finally, the effect of PB on the myogenic differentiation of human skeletal muscle cells (HsKMCs) was investigated. RESULTS DANCR expression was upregulated in the muscle tissues of mice with muscle atrophy and downregulated during myogenic differentiation. Knockout of DANCR promoted myogenic differentiation and significantly alleviated the loss of muscle mass, strength, and function in mice with muscle atrophy. The primary mechanism involved DANCR directly binding to the p65 protein to regulate NF-κB pathway activity. Experiments revealed that PB could target the degradation of DANCR, reduce the nuclear entry of p65, and inhibit the activation of the NF-κB pathway. Consequently, PB significantly inhibited myotube atrophy and the inflammatory response in HsKMCs and promoted their myogenic differentiation by regulating the NF-κB pathway. CONCLUSIONS Our results suggest that PB regulates myogenesis and prevents amyotrophy by targeting the degradation of DANCR and inhibiting the activation of the NF-κB pathway. This study reveals the crucial role of DANCR in maintaining muscle physiology during muscle atrophy and identifies PB as an effective drug that can target DANCR degradation to alleviate muscle atrophy.
Collapse
Affiliation(s)
- Yu Liu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Honglin Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - You Zhang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qi Shang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Wenhua Zhao
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Yuzhuo Zhang
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Weiyu Qiu
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Weicheng Qin
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Feng Lin
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiahui He
- The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou 510130, China
| | - Huiwen Liu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xingda Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yan Gong
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Lingjuan Liu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yixuan Jiang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Hui Ren
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China; Guangzhou Medical University, Guangzhou 511436, China.
| | - Xiaobing Jiang
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China; Guangzhou Medical University, Guangzhou 511436, China.
| | - Gengyang Shen
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China; Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
2
|
Damanti S, Senini E, De Lorenzo R, Merolla A, Santoro S, Festorazzi C, Messina M, Vitali G, Sciorati C, Rovere-Querini P. Acute Sarcopenia: Mechanisms and Management. Nutrients 2024; 16:3428. [PMID: 39458423 PMCID: PMC11510680 DOI: 10.3390/nu16203428] [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: 08/26/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Acute sarcopenia refers to the swift decline in muscle function and mass following acute events such as illness, surgery, trauma, or burns that presents significant challenges in hospitalized older adults. METHODS narrative review to describe the mechanisms and management of acute sarcopenia. RESULTS The prevalence of acute sarcopenia ranges from 28% to 69%, likely underdiagnosed due to the absence of muscle mass and function assessments in most clinical settings. Systemic inflammation, immune-endocrine dysregulation, and anabolic resistance are identified as key pathophysiological factors. Interventions include early mobilization, resistance exercise, neuromuscular electrical stimulation, and nutritional strategies such as protein supplementation, leucine, β-hydroxy-β-methyl-butyrate, omega-3 fatty acids, and creatine monohydrate. Pharmaceuticals show variable efficacy. CONCLUSIONS Future research should prioritize serial monitoring of muscle parameters, identification of predictive biomarkers, and the involvement of multidisciplinary teams from hospital admission to address sarcopenia. Early and targeted interventions are crucial to improve outcomes and prevent long-term disability associated with acute sarcopenia.
Collapse
Affiliation(s)
- Sarah Damanti
- Internal Medicine Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (S.D.); (G.V.); (P.R.-Q.)
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Eleonora Senini
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Rebecca De Lorenzo
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Aurora Merolla
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Simona Santoro
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Costanza Festorazzi
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Marco Messina
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Giordano Vitali
- Internal Medicine Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (S.D.); (G.V.); (P.R.-Q.)
| | - Clara Sciorati
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Patrizia Rovere-Querini
- Internal Medicine Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (S.D.); (G.V.); (P.R.-Q.)
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| |
Collapse
|
3
|
Zhang Y, Dos Santos M, Huang H, Chen K, Iyengar P, Infante R, Polanco PM, Brekken RA, Cai C, Caijgas A, Cano Hernandez K, Xu L, Bassel-Duby R, Liu N, Olson EN. A molecular pathway for cancer cachexia-induced muscle atrophy revealed at single-nucleus resolution. Cell Rep 2024; 43:114587. [PMID: 39116208 PMCID: PMC11472345 DOI: 10.1016/j.celrep.2024.114587] [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: 02/14/2024] [Revised: 05/14/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer cachexia is a prevalent and often fatal wasting condition that cannot be fully reversed with nutritional interventions. Muscle atrophy is a central component of the syndrome, but the mechanisms whereby cancer leads to skeletal muscle atrophy are not well understood. We performed single-nucleus multi-omics on skeletal muscles from a mouse model of cancer cachexia and profiled the molecular changes in cachexic muscle. Our results revealed the activation of a denervation-dependent gene program that upregulates the transcription factor myogenin. Further studies showed that a myogenin-myostatin pathway promotes muscle atrophy in response to cancer cachexia. Short hairpin RNA inhibition of myogenin or inhibition of myostatin through overexpression of its endogenous inhibitor follistatin prevented cancer cachexia-induced muscle atrophy in mice. Our findings uncover a molecular basis of muscle atrophy associated with cancer cachexia and highlight potential therapeutic targets for this disorder.
Collapse
Affiliation(s)
- Yichi Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Matthieu Dos Santos
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Huocong Huang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kenian Chen
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Puneeth Iyengar
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Rodney Infante
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Patricio M Polanco
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rolf A Brekken
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chunyu Cai
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ambar Caijgas
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Karla Cano Hernandez
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ning Liu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
4
|
Compton SLE, Heymsfield SB, Brown JC. Nutritional Mechanisms of Cancer Cachexia. Annu Rev Nutr 2024; 44:77-98. [PMID: 39207878 DOI: 10.1146/annurev-nutr-062122-015646] [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] [Indexed: 09/04/2024]
Abstract
Cancer cachexia is a complex systemic wasting syndrome. Nutritional mechanisms that span energy intake, nutrient metabolism, body composition, and energy balance may be impacted by, and may contribute to, the development of cachexia. To date, clinical management of cachexia remains elusive. Leaning on discoveries and novel methodologies from other fields of research may bolster new breakthroughs that improve nutritional management and clinical outcomes. Characteristics that compare and contrast cachexia and obesity may reveal opportunities for cachexia research to adopt methodology from the well-established field of obesity research. This review outlines the known nutritional mechanisms and gaps in the knowledge surrounding cancer cachexia. In parallel, we present how obesity may be a different side of the same coin and how obesity research has tackled similar research questions. We present insights into how cachexia research may utilize nutritional methodology to expand our understanding of cachexia to improve definitions and clinical care in future directions for the field.
Collapse
Affiliation(s)
- Stephanie L E Compton
- Cancer Energetics Unit, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA;
| | - Steven B Heymsfield
- Metabolism and Body Composition Unit, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Justin C Brown
- Cancer Energetics Unit, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA;
| |
Collapse
|
5
|
Lord SO, Dawson PW, Chunthorng-Orn J, Ng J, Baehr LM, Hughes DC, Sridhar P, Knowles T, Bodine SC, Lai YC. Uncovering the mechanisms of MuRF1-induced ubiquitylation and revealing similarities with MuRF2 and MuRF3. Biochem Biophys Rep 2024; 37:101636. [PMID: 38283190 PMCID: PMC10818185 DOI: 10.1016/j.bbrep.2023.101636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/12/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
MuRF1 (Muscle-specific RING finger protein 1; gene name TRIM63) is a ubiquitin E3 ligase, associated with the progression of muscle atrophy. As a RING (Really Interesting New Gene) type E3 ligase, its unique activity of ubiquitylation is driven by a specific interaction with a UBE2 (ubiquitin conjugating enzyme). Our understanding of MuRF1 function remains unclear as candidate UBE2s have not been fully elucidated. In the present study, we screened human ubiquitin dependent UBE2s in vitro and found that MuRF1 engages in ubiquitylation with UBE2D, UBE2E, UBE2N/V families and UBE2W. MuRF1 can cause mono-ubiquitylation, K48- and K63-linked polyubiquitin chains in a UBE2 dependent manner. Moreover, we identified a two-step UBE2 dependent mechanism whereby MuRF1 is monoubiquitylated by UBE2W which acts as an anchor for UBE2N/V to generate polyubiquitin chains. With the in vitro ubiquitylation assay, we also found that MuRF2 and MuRF3 not only share the same UBE2 partners as MuRF1 but can also directly ubiquitylate the same substrates: Titin (A168-A170), Desmin, and MYLPF (Myosin Light Chain, Phosphorylatable, Fast Skeletal Muscle; also called Myosin Light Regulatory Chain 2). In summary, our work presents new insights into the mechanisms that underpin MuRF1 activity and reveals overlap in MuRF-induced ubiquitylation which could explain their partial redundancy in vivo.
Collapse
Affiliation(s)
- Samuel O. Lord
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Peter W.J. Dawson
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| | | | - Jimi Ng
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Leslie M. Baehr
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - David C. Hughes
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Pooja Sridhar
- School of Biosciences, University of Birmingham, Birmingham, UK
| | - Timothy Knowles
- School of Biosciences, University of Birmingham, Birmingham, UK
| | - Sue C. Bodine
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Yu-Chiang Lai
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
6
|
Qin H, Rui J, Lao J. Heme oxygenase-1 therapy attenuates muscle atrophy following global brachial plexus avulsion in juvenile rats. Muscle Nerve 2023; 68:789-797. [PMID: 37698285 DOI: 10.1002/mus.27972] [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: 01/21/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 09/13/2023]
Abstract
INTRODUCTION/AIMS Brachial plexus injury can seriously affect distal target muscle function, and long-term denervation leads to irreversible structural damage. In the present study, we examined the effect of hemin, a heme oxygenase-1 (HO-1) inducer, on intrinsic forepaw muscle atrophy induced by pan-plexus injury in juvenile rats, as well as its underlying mechanism. METHODS A global brachial plexus avulsion (GBPA) model of rat was established, and thirty 6-wk-old male rats were randomly divided into five groups: control, GBPA plus scramble small intering RNA (siRNA), GBPA plus scramble siRNA plus hemin, GBPA plus HO-1 siRNA, and GBPA plus HO-1 siRNA plus hemin. Hemin (50 mg/kg) was administered intraperitoneally once daily and the siRNA (5 μg) was injected intramuscularly twice a week. Intrinsic forepaw muscles were used for analysis. Myofiber cross-sectional area (CSA), capillary-to-fiber ratio (C/F), and fiber-type composition were assessed. The levels of inflammatory factors, ubiquitin-protein ligases, and autophagy-related proteins were also measured. RESULTS We found that hemin treatment could effectively ameliorate denervated intrinsic forepaw muscle atrophy and suppress type I to II myofiber-type conversion. Hemin treatment failed to prevent muscle capillary loss after denervation. The levels of inflammatory factors (tumor necrosis factor alpha [TNFα] and interleukin 6 [IL-6]), ubiquitin-protein ligases (MuRF-1 and MAFbx), and autophagy-related proteins (BNIP3 and LC3B-II/I ratio) were increased by denervation and HO-1 therapy attenuated the increment. DISCUSSION Upregulation of HO-1 might potentially be an effective strategy to alleviate denervation-related muscle atrophy and might be a promising adjunctive treatment to improve hand function in children with pan-plexus injury.
Collapse
Affiliation(s)
- Hongjiu Qin
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hand Reconstruction (Fudan University), Shanghai, China
- Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, China
- Institute of Hand Surgery, Fudan University, Shanghai, China
| | - Jing Rui
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hand Reconstruction (Fudan University), Shanghai, China
- Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, China
- Institute of Hand Surgery, Fudan University, Shanghai, China
| | - Jie Lao
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hand Reconstruction (Fudan University), Shanghai, China
- Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, China
- Institute of Hand Surgery, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Calvani R, Picca A, Coelho-Júnior HJ, Tosato M, Marzetti E, Landi F. "Diet for the prevention and management of sarcopenia". Metabolism 2023:155637. [PMID: 37352971 DOI: 10.1016/j.metabol.2023.155637] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 06/25/2023]
Abstract
Sarcopenia is a geriatric condition characterized by a progressive loss of skeletal muscle mass and strength, with an increased risk of adverse health outcomes (e.g., falls, disability, institutionalization, reduced quality of life, mortality). Pharmacological remedies are currently unavailable for preventing the development of sarcopenia, halting its progression, or impeding its negative health outcomes. The most effective strategies to contrast sarcopenia rely on the adoption of healthier lifestyle behaviors, including adherence to high-quality diets and regular physical activity. In this review, the role of nutrition in the prevention and management of sarcopenia is summarized. Special attention is given to current "blockbuster" dietary regimes and agents used to counteract age-related muscle wasting, together with their putative mechanisms of action. Issues related to the design and implementation of effective nutritional strategies are discussed, with a focus on unanswered questions on the most appropriate timing of nutritional interventions to preserve muscle health and function into old age. A brief description is also provided on new technologies that can facilitate the development and implementation of personalized nutrition plans to contrast sarcopenia.
Collapse
Affiliation(s)
- Riccardo Calvani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy.
| | - Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy; Department of Medicine and Surgery, LUM University, 70100 Casamassima, Italy.
| | - Hélio José Coelho-Júnior
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Matteo Tosato
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy.
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy.
| | - Francesco Landi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy.
| |
Collapse
|
8
|
Ferrer M, Anthony TG, Ayres JS, Biffi G, Brown JC, Caan BJ, Cespedes Feliciano EM, Coll AP, Dunne RF, Goncalves MD, Grethlein J, Heymsfield SB, Hui S, Jamal-Hanjani M, Lam JM, Lewis DY, McCandlish D, Mustian KM, O'Rahilly S, Perrimon N, White EP, Janowitz T. Cachexia: A systemic consequence of progressive, unresolved disease. Cell 2023; 186:1824-1845. [PMID: 37116469 PMCID: PMC11059056 DOI: 10.1016/j.cell.2023.03.028] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/15/2023] [Accepted: 03/23/2023] [Indexed: 04/30/2023]
Abstract
Cachexia, a systemic wasting condition, is considered a late consequence of diseases, including cancer, organ failure, or infections, and contributes to significant morbidity and mortality. The induction process and mechanistic progression of cachexia are incompletely understood. Refocusing academic efforts away from advanced cachexia to the etiology of cachexia may enable discoveries of new therapeutic approaches. Here, we review drivers, mechanisms, organismal predispositions, evidence for multi-organ interaction, model systems, clinical research, trials, and care provision from early onset to late cachexia. Evidence is emerging that distinct inflammatory, metabolic, and neuro-modulatory drivers can initiate processes that ultimately converge on advanced cachexia.
Collapse
Affiliation(s)
- Miriam Ferrer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; MRC Cancer Unit, University of Cambridge, Hutchison Research Centre, Cambridge Biomedical Campus, Cambridge CB2 0XZ, UK
| | - Tracy G Anthony
- Department of Nutritional Sciences, Rutgers School of Environmental and Biological Sciences, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Janelle S Ayres
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Giulia Biffi
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - Justin C Brown
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Bette J Caan
- Kaiser Permanente Northern California Division of Research, Oakland, CA 94612, USA
| | | | - Anthony P Coll
- Wellcome Trust-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Richard F Dunne
- University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA
| | - Marcus D Goncalves
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jonas Grethlein
- Ruprecht Karl University of Heidelberg, Heidelberg 69117, Germany
| | - Steven B Heymsfield
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Sheng Hui
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Mariam Jamal-Hanjani
- Department of Medical Oncology, University College London Hospitals, London WC1E 6DD, UK; Cancer Research UK Lung Cancer Centre of Excellence and Cancer Metastasis Laboratory, University College London Cancer Institute, London WC1E 6DD, UK
| | - Jie Min Lam
- Cancer Research UK Lung Cancer Centre of Excellence and Cancer Metastasis Laboratory, University College London Cancer Institute, London WC1E 6DD, UK
| | - David Y Lewis
- The Beatson Institute for Cancer Research, Cancer Research UK, Glasgow G61 1BD, UK
| | - David McCandlish
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Karen M Mustian
- University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA
| | - Stephen O'Rahilly
- Wellcome Trust-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Eileen P White
- Rutgers Cancer Institute of New Jersey, Department of Molecular Biology and Biochemistry, Rutgers University, The State University of New Jersey, New Brunswick, NJ 08901, USA; Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| | - Tobias Janowitz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Northwell Health Cancer Institute, Northwell Health, New Hyde Park, NY 11042, USA.
| |
Collapse
|
9
|
Webster JM, Waaijenberg K, van de Worp WRPH, Kelders MCJM, Lambrichts S, Martin C, Verhaegen F, Van der Heyden B, Smith C, Lavery GG, Schols AMWJ, Hardy RS, Langen RCJ. 11β-HSD1 determines the extent of muscle atrophy in a model of acute exacerbation of COPD. Am J Physiol Lung Cell Mol Physiol 2023; 324:L400-L412. [PMID: 36807882 PMCID: PMC10027082 DOI: 10.1152/ajplung.00009.2022] [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] [Indexed: 02/23/2023] Open
Abstract
Muscle atrophy is an extrapulmonary complication of acute exacerbations (AE) in chronic obstructive pulmonary disease (COPD). The endogenous production and therapeutic application of glucocorticoids (GCs) have been implicated as drivers of muscle loss in AE-COPD. The enzyme 11 β-hydroxysteroid dehydrogenase 1 (11β-HSD1) activates GCs and contributes toward GC-induced muscle wasting. To explore the potential of 11βHSD1 inhibition to prevent muscle wasting here, the objective of this study was to ascertain the contribution of endogenous GC activation and amplification by 11βHSD1 in skeletal muscle wasting during AE-COPD. Emphysema was induced by intratracheal (IT) instillation of elastase to model COPD in WT and 11βHSD1/KO mice, followed by vehicle or IT-LPS administration to mimic AE. µCT scans were obtained prior and at study endpoint 48 h following IT-LPS, to assess emphysema development and muscle mass changes, respectively. Plasma cytokine and GC profiles were determined by ELISA. In vitro, myonuclear accretion and cellular response to plasma and GCs were determined in C2C12 and human primary myotubes. Muscle wasting was exacerbated in LPS-11βHSD1/KO animals compared with WT controls. RT-qPCR and western blot analysis showed elevated catabolic and suppressed anabolic pathways in muscle of LPS-11βHSD1/KO animals relative to WTs. Plasma corticosterone levels were higher in LPS-11βHSD1/KO animals, whereas C2C12 myotubes treated with LPS-11βHSD1/KO plasma or exogenous GCs displayed reduced myonuclear accretion relative to WT counterparts. This study reveals that 11β-HSD1 inhibition aggravates muscle wasting in a model of AE-COPD, suggesting that therapeutic inhibition of 11β-HSD1 may not be appropriate to prevent muscle wasting in this setting.
Collapse
Affiliation(s)
- Justine M Webster
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
- Faculty of Health, Medicine and Life Sciences, Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Kelsy Waaijenberg
- Faculty of Health, Medicine and Life Sciences, Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Wouter R P H van de Worp
- Faculty of Health, Medicine and Life Sciences, Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Marco C J M Kelders
- Faculty of Health, Medicine and Life Sciences, Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Sara Lambrichts
- Faculty of Health, Medicine and Life Sciences, Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Claire Martin
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Faculty of Health, Medicine and Life Sciences, Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Frank Verhaegen
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Brent Van der Heyden
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Charlotte Smith
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Gareth G Lavery
- Department of Biosciences, Nottingham Trent University, Nottingham, United Kingdom
| | - Annemie M W J Schols
- Faculty of Health, Medicine and Life Sciences, Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Rowan S Hardy
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
- Institute of Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Ramon C J Langen
- Faculty of Health, Medicine and Life Sciences, Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
10
|
Faye AS, Khan T, Cautha S, Kochar B. Sarcopenia in Inflammatory Bowel Diseases: Reviewing Past Work to Pave the Path for the Future. CURRENT TREATMENT OPTIONS IN GASTROENTEROLOGY 2022; 20:250-260. [PMID: 36388172 PMCID: PMC9648863 DOI: 10.1007/s11938-022-00389-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 06/16/2023]
Abstract
Purpose of the Review Sarcopenia is the loss of muscle quantity and strength. It is highly prevalent in patients with inflammatory bowel disease (IBD) and is associated with periods of ongoing inflammation. This review will summarize the prior work in the field and highlight areas for future research. Recent Findings The presence of sarcopenia has been associated with adverse outcomes in different populations. Most recently, sarcopenia has been associated with adverse postoperative outcomes and an increased likelihood of surgery in IBD. Despite this, significant heterogeneity among these studies limits the ability to draw definitive conclusions. Summary The importance of sarcopenia in inflammatory bowel disease (IBD) is only beginning to be recognized. Future studies assessing it utility both as a risk stratification tool and a modifiable factor in IBD are needed.
Collapse
Affiliation(s)
- Adam S. Faye
- Division of Gastroenterology, NYU Grossman School of Medicine, New York, NY
- Department of Population Health, NYU Grossman School of Medicine, New York, NY
| | - Tasnin Khan
- New York Institute of Technology, Old Westbury, NY
| | - Sandhya Cautha
- Department of Medicine, BronxCare Health System, Bronx, NY
| | - Bharati Kochar
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA
- The Mongan Institute, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
11
|
Pattnaik B, Patnaik N, Mittal S, Mohan A, Agrawal A, Guleria R, Madan K. Micro RNAs as potential biomarkers in tuberculosis: A systematic review. Noncoding RNA Res 2022; 7:16-26. [PMID: 35128217 PMCID: PMC8792429 DOI: 10.1016/j.ncrna.2021.12.005] [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] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 12/15/2022] Open
Abstract
Tuberculosis (TB) remains a major infectious disease across the globe. With increasing TB infections and a rise in multi-drug resistance, rapid diagnostic modalities are required to achieve TB control. Radiological investigations and microbiological tests (microscopic examination, cartridge-based nucleic acid amplification tests, and cultures) are most commonly used to diagnose TB. Histopathological/cytopathological examinations are also required for an accurate diagnosis in many patients. The causative agent, Mycobacterium tuberculosis (Mtb), is known to circumvent the host's immune system. Circulating microRNAs (miRNAs) play a crucial role in biological pathways and can be used as a potential biomarker to detect tuberculosis. miRNAs are small non-coding RNAs and negatively regulate gene expression during post-transcriptional regulation. The differential expression of miRNAs in multiple clinical samples in tuberculosis patients may be helpful as potential disease biomarkers. This review summarizes the literature on miRNAs in various clinical samples as biomarkers for TB diagnosis.
Collapse
Affiliation(s)
- Bijay Pattnaik
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Niharika Patnaik
- Centre of Excellence in Asthma & Lung Disease, Molecular Immunogenetics Lab, CSIR-Institute of Genomics & Integrative Biology, New Delhi, India
| | - Saurabh Mittal
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Anant Mohan
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Anurag Agrawal
- Centre of Excellence in Asthma & Lung Disease, Molecular Immunogenetics Lab, CSIR-Institute of Genomics & Integrative Biology, New Delhi, India
| | - Randeep Guleria
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Karan Madan
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
12
|
Kimura N, Kawahara T, Uemura Y, Atsumi T, Sumida T, Mimura T, Kawaguchi Y, Amano H, Iwasaki Y, Kaneko Y, Matsui T, Muro Y, Imura Y, Kanda T, Tanaka Y, Kawakami A, Jinnin M, Ishii T, Hiromura K, Miwa Y, Nakajima H, Kuwana M, Nishioka Y, Morinobu A, Kameda H, Kohsaka H. Branched chain amino acids in the treatment of polymyositis and dermatomyositis: a phase II/III, multi-center, randomized controlled trial. Rheumatology (Oxford) 2022; 61:4445-4454. [PMID: 35179548 DOI: 10.1093/rheumatology/keac101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/23/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To assess the efficacy and safety of branched chain amino acids (BCAAs) in the treatment of PM/DM prior to official approval of their use in Japan. METHODS Treatment naïve adults with PM/DM were enrolled in a randomized, double-blind trial to receive either TK-98 (drug name of BCAAs) or placebo in addition to conventional treatment. After 12 weeks, patients with an average manual muscle test (MMT) score <9.5 were enrolled in an open label extension study for a further 12 weeks. The primary end point was the change of the MMT score at 12 weeks. The secondary end points were the clinical response and the change of functional index (FI). RESULTS Forty-seven patients were randomized either to the TK-98 (n = 24) or placebo (n = 23) groups. The changes of MMT scores at 12 weeks were 0.70±0.19 (mean±SEM) and 0.69±0.18, respectively (P = 0.98). Thirteen patients from the TK-98 group and 12 from the placebo group were enrolled in the extension study. The MMT scores in both groups improved similarly. The increase of the FI scores of the shoulder flexion at 12 weeks was significantly greater in the TK-98 group (27.9±5.67 vs. 12.8±5.67 for the right shoulder flexion, and 27.0±5.44 vs. 13.4±5.95 for the left shoulder [P < 0.05]). Frequencies of adverse events upto 12 weeks were similar. CONCLUSION BCAAs showed no effect on the improvement of the muscle strength evaluated by MMT and the clinical response. However, they were partly effective for improving dynamic repetitive muscle functions. TRIAL REGISTRATION UMIN-CTR Clinical Trial, https://center6.umin.ac.jp/, UMIN000016233.
Collapse
Affiliation(s)
- Naoki Kimura
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Lifetime Clinical Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takuya Kawahara
- Clinical Research Promotion Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Yukari Uemura
- Clinical Research Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine, Hokkaido University, Hokkaido, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Toshihide Mimura
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Yasushi Kawaguchi
- Department of Rheumatology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Hirofumi Amano
- Department of Rheumatology and Internal Medicine, Juntendo University School of Medicine,Tokyo, Japan
| | - Yukiko Iwasaki
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan.,Department of Palliative Medicine, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Toshihiro Matsui
- Department of Rheumatology, Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Yoshinao Muro
- Department of Dermatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshitaka Imura
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Clinical Immunology and Rheumatology, Tazuke-Kofukai Medical Research Institute, Kitano Hospital, Osaka, Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences,Nagasaki, Japan
| | - Masatoshi Jinnin
- Department of Dermatology, Wakayama Medical University Graduate School of Medicine, Wakayama, Japan
| | - Tomonori Ishii
- Clinical Research, Innovation and Education Center, Tohoku University Hospital, Sendai, Japan
| | - Keiju Hiromura
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine,Maebashi, Japan
| | - Yusuke Miwa
- Department of Medicine, Division of Rheumatology, Showa University School of Medicine, Tokyo, Japan
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Chiba University Hospital, Chiba, Japan
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Yasuhiko Nishioka
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Section of Rheumatology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hideto Kameda
- Department of Internal Medicine, Division of Rheumatology, Faculty of Medicine, Toho University, Tokyo, Japan
| | - Hitoshi Kohsaka
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Rheumatology Center, Chiba-Nishi General Hospital, Matsudo, Japan
| |
Collapse
|
13
|
Ducharme JB, McKenna ZJ, Deyhle MR. Exercise mitigates the Toll of muscle atrophy: A narrative review of the effects of exercise on Toll-like receptor-4 in leukocytes and skeletal muscle. Am J Physiol Cell Physiol 2022; 322:C581-C589. [PMID: 35171696 DOI: 10.1152/ajpcell.00005.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Conditions characterized by muscle wasting such as cachexia and sarcopenia are devastating at the individual level, and they place a profound burden on public health. Evidence suggests that inflammation is likely a mechanistic contributor to the pathogenesis of these conditions. One specific molecule, lipopolysaccharide, has gained attention due to its role in initiating inflammation. Toll-like receptor-4 is the primary receptor for lipopolysaccharide and has been shown to be implicit in the downstream proinflammatory response associated with lipopolysaccharide. Importantly, Toll-like receptor-4 is expressed on various cell types throughout the human body such as leukocytes and skeletal muscle fibers and may have site-specific effects that contribute to muscle wasting conditions based on the location in which activation occurs. Accordingly, reducing proinflammatory signaling at these locations may be an effective strategy at mitigating muscle wasting. Regular exercise training is believed to elicit anti-inflammatory adaptations, but the mechanisms by which this occurs are yet to be fully understood. Understanding the mechanisms by which Toll-like receptor-4 activation contributes to muscle wasting and how exercise affects this, may allow for the development of a non-pharmacological therapeutic intervention. Therefore, in this review, we summarize the current understanding of the lipopolysaccharide/Toll-like receptor-4 axis in leukocytes and skeletal muscle fibers on the pathogenesis of muscle wasting conditions and we critically examine the current evidence regarding the effects of exercise on this axis.
Collapse
Affiliation(s)
- Jeremy B Ducharme
- Department of Health, Exercise, and Sport Sciences, University of New Mexico, Albuquerque, NM, United States
| | - Zachary J McKenna
- Department of Health, Exercise, and Sport Sciences, University of New Mexico, Albuquerque, NM, United States
| | - Michael R Deyhle
- Department of Health, Exercise, and Sport Sciences, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
14
|
Gaddas M. Le dosage de l’Insulin-Like Growth Factor-1 : les difficultés de la détermination sérique et de l’interprétation des résultats. NUTR CLIN METAB 2022. [DOI: 10.1016/j.nupar.2021.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
15
|
Daily JW, Park S. Sarcopenia Is a Cause and Consequence of Metabolic Dysregulation in Aging Humans: Effects of Gut Dysbiosis, Glucose Dysregulation, Diet and Lifestyle. Cells 2022; 11:cells11030338. [PMID: 35159148 PMCID: PMC8834403 DOI: 10.3390/cells11030338] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
Skeletal muscle mass plays a critical role in a healthy lifespan by helping to regulate glucose homeostasis. As seen in sarcopenia, decreased skeletal muscle mass impairs glucose homeostasis, but it may also be caused by glucose dysregulation. Gut microbiota modulates lipopolysaccharide (LPS) production, short-chain fatty acids (SCFA), and various metabolites that affect the host metabolism, including skeletal muscle tissues, and may have a role in the sarcopenia etiology. Here, we aimed to review the relationship between skeletal muscle mass, glucose homeostasis, and gut microbiota, and the effect of consuming probiotics and prebiotics on the development and pathological consequences of sarcopenia in the aging human population. This review includes discussions about the effects of glucose metabolism and gut microbiota on skeletal muscle mass and sarcopenia and the interaction of dietary intake, physical activity, and gut microbiome to influence sarcopenia through modulating the gut–muscle axis. Emerging evidence suggests that the microbiome can regulate both skeletal muscle mass and function, in part through modulating the metabolisms of short-chain fatty acids and branch-chain amino acids that might act directly on muscle in humans or indirectly through the brain and liver. Dietary factors such as fats, proteins, and indigestible carbohydrates and lifestyle interventions such as exercise, smoking, and alcohol intake can both help and hinder the putative gut–muscle axis. The evidence presented in this review suggests that loss of muscle mass and function are not an inevitable consequence of the aging process, and that dietary and lifestyle interventions may prevent or delay sarcopenia.
Collapse
Affiliation(s)
- James W. Daily
- Department of R & D, Daily Manufacturing Inc., Rockwell, 28138 NC, USA;
| | - Sunmin Park
- Department of Food & Nutrition, Obesity/Diabetes Center, Hoseo University, Asan 31499, Korea
- Correspondence: ; Tel.: +82-41-540-5345; Fax: +82-41-548-0670
| |
Collapse
|
16
|
Yi X, Tao J, Qian Y, Feng F, Hu X, Xu T, Jin H, Ruan H, Zheng HF, Tong P. Morroniside ameliorates inflammatory skeletal muscle atrophy via inhibiting canonical and non-canonical NF-κB and regulating protein synthesis/degradation. Front Pharmacol 2022; 13:1056460. [PMID: 36618945 PMCID: PMC9816435 DOI: 10.3389/fphar.2022.1056460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
No drug options exist for skeletal muscle atrophy in clinical, which poses a huge socio-economic burden, making development on drug interventions a general wellbeing need. Patients with a variety of pathologic conditions associated with skeletal muscle atrophy have systemically elevated inflammatory factors. Morroniside, derived from medicinal herb Cornus officinalis, possesses anti-inflammatory effect. However, whether and how morroniside combat muscle atrophy remain unknown. Here, we identified crucial genetic associations between TNFα/NF-κB pathway and grip strength based on population using 377,807 European participants from the United Kingdom Biobank dataset. Denervation increased TNFα in atrophying skeletal muscles, which inhibited myotube formation in vitro. Notably, morroniside treatment rescued TNFα-induced myotube atrophy in vitro and impeded skeletal muscle atrophy in vivo, resulting in increased body/muscles weights, No. of satellite cells, size of type IIA, IIX and IIB myofibers, and percentage of type IIA myofibers in denervated mice. Mechanistically, in vitro and/or in vivo studies demonstrated that morroniside could not only inhibit canonical and non-canonical NF-κB, inflammatory mediators (IL6, IL-1b, CRP, NIRP3, PTGS2, TNFα), but also down-regulate protein degradation signals (Follistatin, Myostatin, ALK4/5/7, Smad7/3), ubiquitin-proteasome molecules (FoxO3, Atrogin-1, MuRF1), autophagy-lysosomal molecules (Bnip3, LC3A, and LC3B), while promoting protein synthesis signals (IGF-1/IGF-1R/IRS-1/PI3K/Akt, and BMP14/BMPR2/ALK2/3/Smad5/9). Moreover, morroniside had no obvious liver and kidney toxicity. This human genetic, cells and mice pathological evidence indicates that morroniside is an efficacious and safe inflammatory muscle atrophy treatment and suggests its translational potential on muscle wasting.
Collapse
Affiliation(s)
- Xiangjiao Yi
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Jianguo Tao
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yu Qian
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Feng Feng
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xueqin Hu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Taotao Xu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Hongting Jin
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Hongfeng Ruan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
- *Correspondence: Peijian Tong, ; Hou-Feng Zheng, ; Hongfeng Ruan,
| | - Hou-Feng Zheng
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- College of Life Sciences, Zhejiang University, Hangzhou, China
- *Correspondence: Peijian Tong, ; Hou-Feng Zheng, ; Hongfeng Ruan,
| | - Peijian Tong
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
- *Correspondence: Peijian Tong, ; Hou-Feng Zheng, ; Hongfeng Ruan,
| |
Collapse
|
17
|
Minari ALA, Thomatieli-Santos RV. From skeletal muscle damage and regeneration to the hypertrophy induced by exercise: What is the role of different macrophages subsets? Am J Physiol Regul Integr Comp Physiol 2021; 322:R41-R54. [PMID: 34786967 DOI: 10.1152/ajpregu.00038.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Macrophages are one of the top players when considering immune cells involved with tissue homeostasis. Recently, increasing evidence has demonstrated that these macrophages could also present two major subsets during tissue healing; proliferative macrophages (M1-like), which are responsible for increasing myogenic cell proliferation, and restorative macrophages (M2-like), which are accountable for the end of the mature muscle myogenesis. The participation and characterization of these macrophage subsets is critical during myogenesis, not only to understand the inflammatory role of macrophages during muscle recovery but also to create supportive strategies that can improve mass muscle maintenance. Indeed, most of our knowledge about macrophage subsets comes from skeletal muscle damage protocols, and we still do not know how these subsets can contribute to skeletal muscle adaptation. This narrative review aims to collect and discuss studies demonstrating the involvement of different macrophage subsets during the skeletal muscle damage/regeneration process, showcasing an essential role of these macrophage subsets during muscle adaptation induced by acute and chronic exercise programs.
Collapse
Affiliation(s)
- André Luis Araujo Minari
- Universidade estadual Paulista, Campus Presidente Prudente, Brazil.,Universidade Federal de São Paulo, Psicobiologia, Brazil
| | - Ronaldo V Thomatieli-Santos
- Universidade Federal de São Paulo, Campus Baixada Santista, Brazil.,Universidade Federal de São Paulo, Psicobiologia, Brazil
| |
Collapse
|
18
|
Franco-Romero A, Sandri M. Role of autophagy in muscle disease. Mol Aspects Med 2021; 82:101041. [PMID: 34625292 DOI: 10.1016/j.mam.2021.101041] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 02/08/2023]
Abstract
Beside inherited muscle diseases many catabolic conditions such as insulin resistance, malnutrition, cancer growth, aging, infections, chronic inflammatory status, inactivity, obesity are characterized by loss of muscle mass, strength and function. The decrease of muscle quality and quantity increases morbidity, mortality and has a major impact on the quality of life. One of the pathogenetic mechanisms of muscle wasting is the dysregulation of the main protein and organelles quality control system of the cell: the autophagy-lysosome. This review will focus on the role of the autophagy-lysosome system in the different conditions of muscle loss. We will also dissect the signalling pathways that are involved in excessive or defective autophagy regulation. Finally, the state of the art of autophagy modulators that have been used in preclinical or clinical studies to ameliorate muscle mass will be also described.
Collapse
Affiliation(s)
- Anais Franco-Romero
- Venetian Institute of Molecular Medicine, via Orus 2, 35129, Padova, Italy; Department of Biomedical Science, University of Padova, via G. Colombo 3, 35100, Padova, Italy
| | - Marco Sandri
- Venetian Institute of Molecular Medicine, via Orus 2, 35129, Padova, Italy; Department of Biomedical Science, University of Padova, via G. Colombo 3, 35100, Padova, Italy; Myology Center, University of Padova, via G. Colombo 3, 35100, Padova, Italy; Department of Medicine, McGill University, Montreal, Canada.
| |
Collapse
|
19
|
Jayawardena TU, Kim SY, Jeon YJ. Sarcopenia; functional concerns, molecular mechanisms involved, and seafood as a nutritional intervention - review article. Crit Rev Food Sci Nutr 2021; 63:1983-2003. [PMID: 34459311 DOI: 10.1080/10408398.2021.1969889] [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] [Indexed: 12/13/2022]
Abstract
The fundamental basis for the human function is provided by skeletal muscle. Advancing age causes selective fiber atrophy, motor unit loss, and hybrid fiber formation resulting in hampered mass and strength, thus referred to as sarcopenia. Influence on the loss of independence of aged adults, contribute toward inclined healthcare costs conveys the injurious impact. The current understating of age-related skeletal muscle changes are addressed in this review, and further discusses mechanisms regulating protein turnover, although they do not completely define the process yet. Moreover, the reduced capacity of muscle regeneration due to impairment of satellite cell activation and proliferation with neuronal, immunological, hormonal factors were brought into the light of attention. Nevertheless, complete understating of sarcopenia requires disentangling it from disuse and disease. Nutritional intervention is considered a potentially preventable factor contributing to sarcopenia. Seafood is a crucial player in the fight against hunger and malnutrition, where it consists of macro and micronutrients. Hence, the review shed light on seafood as a nutritional intrusion in the treatment and prevention of sarcopenia. Understanding multiple factors will provide therapeutic targets in the prevention, treatment, and overcoming adverse effects of sarcopenia.
Collapse
Affiliation(s)
- Thilina U Jayawardena
- Department of Marine Life Sciences, Jeju National University, Jeju, Republic of Korea
| | - Seo-Young Kim
- Division of Practical Application, Honam National Institute of Biological Resources, Mokpo-si, Korea
| | - You-Jin Jeon
- Department of Marine Life Sciences, Jeju National University, Jeju, Republic of Korea.,Marine Science Institute, Jeju National University, Jeju, Jeju Self-Governing Province, Republic of Korea
| |
Collapse
|
20
|
Fang WY, Tseng YT, Lee TY, Fu YC, Chang WH, Lo WW, Lin CL, Lo YC. Triptolide prevents LPS-induced skeletal muscle atrophy via inhibiting NF-κB/TNF-α and regulating protein synthesis/degradation pathway. Br J Pharmacol 2021; 178:2998-3016. [PMID: 33788266 DOI: 10.1111/bph.15472] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/03/2021] [Accepted: 03/20/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Increasing evidence suggests systemic inflammation-caused skeletal muscle atrophy as a major clinical feature of cachexia. Triptolide obtained from Tripterygium wilfordii Hook F possesses potent anti-inflammatory and immunosuppressive effects. The present study aims to evaluate the protective effects and molecular mechanisms of triptolide on inflammation-induced skeletal muscle atrophy. EXPERIMENTAL APPROACH The effects of triptolide on skeletal muscle atrophy were investigated in LPS-treated C2C12 myotubes and C57BL/6 mice. Protein expressions and mRNA levels were analysed by western blot and qPCR, respectively. Skeletal muscle mass, volume and strength were measured by histological analysis, micro-CT and grip strength, respectively. Locomotor activity was measured using the open field test. KEY RESULTS Triptolide (10-100 fM) up-regulated protein synthesis signals (IGF-1/p-IGF-1R/IRS-1/p-Akt/p-mTOR) and down-regulated protein degradation signal atrogin-1 in C2C12 myotubes. In LPS (100 ng·ml-1 )-treated C2C12 myotubes, triptolide up-regulated MyHC, IGF-1, p-IGF-1R, IRS-1 and p-Akt. Triptolide also down-regulated ubiquitin-proteasome molecules (n-FoxO3a/atrogin-1/MuRF1), proteasome activity, autophagy-lysosomal molecules (LC3-II/LC3-I and Bnip3) and inflammatory mediators (NF-κB, Cox-2, NLRP3, IL-1β and TNF-α). However, AG1024, an IGF-1R inhibitor, suppressed triptolide-mediated effects on MyHC, myotube diameter, MuRF1 and p62 in LPS-treated C2C12 myotubes. In LPS (1 mg·kg-1 , i.p.)-challenged mice, triptolide (5 and 20 μg·kg-1 ·day-1 , i.p.) decreased plasma TNF-α levels and it increased skeletal muscle volume, cross-sectional area of myofibers, weights of the gastrocnemius and tibialis anterior muscles, forelimb grip strength and locomotion. CONCLUSIONS AND IMPLICATIONS These findings reveal that triptolide prevented LPS-induced inflammation and skeletal muscle atrophy and have implications for the discovery of novel agents for preventing muscle wasting.
Collapse
Affiliation(s)
- Wei-Yu Fang
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Ting Tseng
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - Tzu-Ying Lee
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yin-Chih Fu
- Department of Orthopedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan-Hsuan Chang
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan-Wen Lo
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Lung Lin
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Neurosurgery, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Ching Lo
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
21
|
Xu R, Wang Z. Involvement of Transcription Factor FoxO1 in the Pathogenesis of Polycystic Ovary Syndrome. Front Physiol 2021; 12:649295. [PMID: 33746783 PMCID: PMC7973228 DOI: 10.3389/fphys.2021.649295] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
FoxO1 is a member of the forkhead transcription factor family subgroup O (FoxO), which is expressed in many cell types, and participates in various pathophysiological processes, including cell proliferation, apoptosis, autophagy, metabolism, inflammatory response, cytokine expression, immune differentiation, and oxidative stress resistance. Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in the women of childbearing age, which is regulated via a variety of signaling pathways. Currently, the specific mechanism underlying the pathogenesis of PCOS is still unclear. As an important transcription factor, FoxO1 activity might be involved in the pathophysiology of PCOS. PCOS has been associated with insulin resistance and low-grade inflammatory response. Therefore, the studies regarding the role of FoxO1 in the incidence and associated complications of PCOS will help provide novel ideas for establishing the treatment strategy of PCOS.
Collapse
Affiliation(s)
- Renfeng Xu
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Provincial University Key Laboratory of Sport and Health Science, Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Zhengchao Wang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Provincial University Key Laboratory of Sport and Health Science, Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
| |
Collapse
|
22
|
Alterations in intestinal microbiota diversity, composition, and function in patients with sarcopenia. Sci Rep 2021; 11:4628. [PMID: 33633246 PMCID: PMC7907362 DOI: 10.1038/s41598-021-84031-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
16S rRNA sequencing of human fecal samples has been tremendously successful in identifying microbiome changes associated with both aging and disease. A number of studies have described microbial alterations corresponding to physical frailty and nursing home residence among aging individuals. A gut-muscle axis through which the microbiome influences skeletal muscle growth/function has been hypothesized. However, the microbiome has yet to be examined in sarcopenia. Here, we collected fecal samples of 60 healthy controls (CON) and 27 sarcopenic (Case)/possibly sarcopenic (preCase) individuals and analyzed the intestinal microbiota using 16S rRNA sequencing. We observed an overall reduction in microbial diversity in Case and preCase samples. The genera Lachnospira, Fusicantenibacter, Roseburia, Eubacterium, and Lachnoclostridium—known butyrate producers—were significantly less abundant in Case and preCase subjects while Lactobacillus was more abundant. Functional pathways underrepresented in Case subjects included numerous transporters and phenylalanine, tyrosine, and tryptophan biosynthesis suggesting that protein processing and nutrient transport may be impaired. In contrast, lipopolysaccharide biosynthesis was overrepresented in Case and PreCase subjects suggesting that sarcopenia is associated with a pro-inflammatory metagenome. These analyses demonstrate structural and functional alterations in the intestinal microbiota that may contribute to loss of skeletal muscle mass and function in sarcopenia.
Collapse
|
23
|
Dalle S, Van Roie E, Hiroux C, Vanmunster M, Coudyzer W, Suhr F, Bogaerts S, Van Thienen R, Koppo K. Omega-3 Supplementation Improves Isometric Strength But Not Muscle Anabolic and Catabolic Signaling in Response to Resistance Exercise in Healthy Older Adults. J Gerontol A Biol Sci Med Sci 2021; 76:406-414. [PMID: 33284965 PMCID: PMC7907485 DOI: 10.1093/gerona/glaa309] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Indexed: 12/11/2022] Open
Abstract
Old skeletal muscle exhibits decreased anabolic sensitivity, eventually contributing to muscle wasting. Besides anabolism, also muscle inflammation and catabolism are critical players in regulating the old skeletal muscle's sensitivity. Omega-3 fatty acids (ω-3) are an interesting candidate to reverse anabolic insensitivity via anabolic actions. Yet, it remains unknown whether ω-3 also attenuates muscle inflammation and catabolism. The present study investigates the effect of ω-3 supplementation on muscle inflammation and metabolism (anabolism/catabolism) upon resistance exercise (RE). Twenty-three older adults (65-84 years; 8♀) were randomized to receive ω-3 (~3 g/d) or corn oil (placebo [PLAC]) and engaged in a 12-week RE program (3×/wk). Before and after intervention, muscle volume, strength, and systemic inflammation were assessed, and muscle biopsies were analyzed for markers of anabolism, catabolism, and inflammation. Isometric knee-extensor strength increased in ω-3 (+12.2%), but not in PLAC (-1.4%; pinteraction = .015), whereas leg press strength improved in both conditions (+27.1%; ptime < .001). RE, but not ω-3, decreased inflammatory (p65NF-κB) and catabolic (FOXO1, LC3b) markers, and improved muscle quality. Yet, muscle volume remained unaffected by RE and ω-3. Accordingly, muscle anabolism (mTORC1) and plasma C-reactive protein remained unchanged by RE and ω-3, whereas serum IL-6 tended to decrease in ω-3 (pinteraction = .07). These results show that, despite no changes in muscle volume, RE-induced gains in isometric strength can be further enhanced by ω-3. However, ω-3 did not improve RE-induced beneficial catabolic or inflammatory adaptations. Irrespective of muscle volume, gains in strength (primary criterion for sarcopenia) might be explained by changes in muscle quality due to muscle inflammatory or catabolic signaling.
Collapse
Affiliation(s)
- Sebastiaan Dalle
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Belgium
| | - Evelien Van Roie
- Physical Activity, Sports and Health Research Group, Department of Movement Sciences, KU Leuven, Belgium
| | - Charlotte Hiroux
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Belgium
| | - Mathias Vanmunster
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Belgium
| | - Walter Coudyzer
- Department of Morphology and Medical Imaging, Faculty of Medicine, Radiology Section, KU Leuven, Belgium
| | - Frank Suhr
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Belgium
| | - Stijn Bogaerts
- Locomotor and Neurological Disorders, Department of Development and Regeneration, KU Leuven, Belgium
| | - Ruud Van Thienen
- Research Group for Neurorehabilitation, Department of Rehabilitation Sciences, KU Leuven, Belgium
| | - Katrien Koppo
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Belgium
| |
Collapse
|
24
|
Webster JM, Kempen LJAP, Hardy RS, Langen RCJ. Inflammation and Skeletal Muscle Wasting During Cachexia. Front Physiol 2020; 11:597675. [PMID: 33329046 PMCID: PMC7710765 DOI: 10.3389/fphys.2020.597675] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022] Open
Abstract
Cachexia is the involuntary loss of muscle and adipose tissue that strongly affects mortality and treatment efficacy in patients with cancer or chronic inflammatory disease. Currently, no specific treatments or interventions are available for patients developing this disorder. Given the well-documented involvement of pro-inflammatory cytokines in muscle and fat metabolism in physiological responses and in the pathophysiology of chronic inflammatory disease and cancer, considerable interest has revolved around their role in mediating cachexia. This has been supported by association studies that report increased levels of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) in some, but not all, cancers and in chronic inflammatory diseases such as chronic obstructive pulmonary disease (COPD) and rheumatoid arthritis (RA). In addition, preclinical studies including animal disease models have provided a substantial body of evidence implicating a causal contribution of systemic inflammation to cachexia. The presence of inflammatory cytokines can affect skeletal muscle through several direct mechanisms, relying on activation of the corresponding receptor expressed by muscle, and resulting in inhibition of muscle protein synthesis (MPS), elevation of catabolic activity through the ubiquitin-proteasomal system (UPS) and autophagy, and impairment of myogenesis. Additionally, systemic inflammatory mediators indirectly contribute to muscle wasting through dysregulation of tissue and organ systems, including GCs via the hypothalamus-pituitary-adrenal (HPA) axis, the digestive system leading to anorexia-cachexia, and alterations in liver and adipocyte behavior, which subsequently impact on muscle. Finally, myokines secreted by skeletal muscle itself in response to inflammation have been implicated as autocrine and endocrine mediators of cachexia, as well as potential modulators of this debilitating condition. While inflammation has been shown to play a pivotal role in cachexia development, further understanding how these cytokines contribute to disease progression is required to reveal biomarkers or diagnostic tools to help identify at risk patients, or enable the design of targeted therapies to prevent or delay the progression of cachexia.
Collapse
Affiliation(s)
- Justine M. Webster
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Laura J. A. P. Kempen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Rowan S. Hardy
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Institute for Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
- MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - Ramon C. J. Langen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
25
|
Impacts of Green Tea on Joint and Skeletal Muscle Health: Prospects of Translational Nutrition. Antioxidants (Basel) 2020; 9:antiox9111050. [PMID: 33126483 PMCID: PMC7692648 DOI: 10.3390/antiox9111050] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/14/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis and sarcopenia are two major joint and skeletal muscle diseases prevalent during aging. Osteoarthritis is a multifactorial progressive degenerative and inflammatory disorder of articular cartilage. Cartilage protection and pain management are the two most important strategies in the management of osteoarthritis. Sarcopenia, a condition of loss of muscle mass and strength, is associated with impaired neuromuscular innervation, the transition of skeletal muscle fiber type, and reduced muscle regenerative capacity. Management of sarcopenia requires addressing both skeletal muscle quantity and quality. Emerging evidence suggests that green tea catechins play an important role in maintaining healthy joints and skeletal muscle. This review covers (i) the prevalence and etiology of osteoarthritis and sarcopenia, such as excessive inflammation and oxidative stress, mitochondrial dysfunction, and reduced autophagy; (ii) the effects of green tea catechins on joint health by downregulating inflammatory signaling mediators, upregulating anabolic mediators, and modulating miRNAs expression, resulting in reduced chondrocyte death, collagen degradation, and cartilage protection; (iii) the effects of green tea catechins on skeletal muscle health via maintaining a dynamic balance between protein synthesis and degradation and boosting the synthesis of mitochondrial energy metabolism, resulting in favorable muscle homeostasis and mitigation of muscle atrophy with aging; and (iv) the current study limitations and future research directions.
Collapse
|
26
|
Peris-Moreno D, Taillandier D, Polge C. MuRF1/TRIM63, Master Regulator of Muscle Mass. Int J Mol Sci 2020; 21:ijms21186663. [PMID: 32933049 PMCID: PMC7555135 DOI: 10.3390/ijms21186663] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
The E3 ubiquitin ligase MuRF1/TRIM63 was identified 20 years ago and suspected to play important roles during skeletal muscle atrophy. Since then, numerous studies have been conducted to decipher the roles, molecular mechanisms and regulation of this enzyme. This revealed that MuRF1 is an important player in the skeletal muscle atrophy process occurring during catabolic states, making MuRF1 a prime candidate for pharmacological treatments against muscle wasting. Indeed, muscle wasting is an associated event of several diseases (e.g., cancer, sepsis, diabetes, renal failure, etc.) and negatively impacts the prognosis of patients, which has stimulated the search for MuRF1 inhibitory molecules. However, studies on MuRF1 cardiac functions revealed that MuRF1 is also cardioprotective, revealing a yin and yang role of MuRF1, being detrimental in skeletal muscle and beneficial in the heart. This review discusses data obtained on MuRF1, both in skeletal and cardiac muscles, over the past 20 years, regarding the structure, the regulation, the location and the different functions identified, and the first inhibitors reported, and aim to draw the picture of what is known about MuRF1. The review also discusses important MuRF1 characteristics to consider for the design of future drugs to maintain skeletal muscle mass in patients with different pathologies.
Collapse
|
27
|
Koutnik AP, Poff AM, Ward NP, DeBlasi JM, Soliven MA, Romero MA, Roberson PA, Fox CD, Roberts MD, D'Agostino DP. Ketone Bodies Attenuate Wasting in Models of Atrophy. J Cachexia Sarcopenia Muscle 2020; 11:973-996. [PMID: 32239651 PMCID: PMC7432582 DOI: 10.1002/jcsm.12554] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/18/2020] [Accepted: 01/30/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Cancer Anorexia Cachexia Syndrome (CACS) is a distinct atrophy disease negatively influencing multiple aspects of clinical care and patient quality of life. Although it directly causes 20% of all cancer-related deaths, there are currently no model systems that encompass the entire multifaceted syndrome, nor are there any effective therapeutic treatments. METHODS A novel model of systemic metastasis was evaluated for the comprehensive CACS (metastasis, skeletal muscle and adipose tissue wasting, inflammation, anorexia, anemia, elevated protein breakdown, hypoalbuminemia, and metabolic derangement) in both males and females. Ex vivo skeletal muscle analysis was utilized to determine ubiquitin proteasome degradation pathway activation. A novel ketone diester (R/S 1,3-Butanediol Acetoacetate Diester) was assessed in multifaceted catabolic environments to determine anti-atrophy efficacy. RESULTS Here, we show that the VM-M3 mouse model of systemic metastasis demonstrates a novel, immunocompetent, logistically feasible, repeatable phenotype with progressive tumor growth, spontaneous metastatic spread, and the full multifaceted CACS with sex dimorphisms across tissue wasting. We also demonstrate that the ubiquitin proteasome degradation pathway was significantly upregulated in association with reduced insulin-like growth factor-1/insulin and increased FOXO3a activation, but not tumor necrosis factor-α-induced nuclear factor-kappa B activation, driving skeletal muscle atrophy. Additionally, we show that R/S 1,3-Butanediol Acetoacetate Diester administration shifted systemic metabolism, attenuated tumor burden indices, reduced atrophy/catabolism and mitigated comorbid symptoms in both CACS and cancer-independent atrophy environments. CONCLUSIONS Our findings suggest the ketone diester attenuates multifactorial CACS skeletal muscle atrophy and inflammation-induced catabolism, demonstrating anti-catabolic effects of ketone bodies in multifactorial atrophy.
Collapse
Affiliation(s)
- Andrew P. Koutnik
- Department of Molecular Pharmacology and PhysiologyMorsani College of Medicine, University of South FloridaTampaFLUSA
| | - Angela M. Poff
- Department of Molecular Pharmacology and PhysiologyMorsani College of Medicine, University of South FloridaTampaFLUSA
| | - Nathan P. Ward
- Department of Cancer PhysiologyMoffitt Cancer Center, H. Lee Moffitt Cancer Center and Research InstituteTampaFLUSA
| | - Janine M. DeBlasi
- Department of Molecular Pharmacology and PhysiologyMorsani College of Medicine, University of South FloridaTampaFLUSA
| | - Maricel A. Soliven
- Department of Molecular Pharmacology and PhysiologyMorsani College of Medicine, University of South FloridaTampaFLUSA
| | | | | | - Carl D. Fox
- School of KinesiologyAuburn UniversityAuburnALUSA
| | | | - Dominic P. D'Agostino
- Department of Molecular Pharmacology and PhysiologyMorsani College of Medicine, University of South FloridaTampaFLUSA
- Institute for Human and Machine CognitionOcalaFLUSA
| |
Collapse
|
28
|
Cisplatin-Induced Skeletal Muscle Dysfunction: Mechanisms and Counteracting Therapeutic Strategies. Int J Mol Sci 2020; 21:ijms21041242. [PMID: 32069876 PMCID: PMC7072891 DOI: 10.3390/ijms21041242] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/08/2020] [Accepted: 02/09/2020] [Indexed: 12/17/2022] Open
Abstract
Among the severe side effects induced by cisplatin chemotherapy, muscle wasting is the most relevant one. This effect is a major cause for a clinical decline of cancer patients, since it is a negative predictor of treatment outcome and associated to increased mortality. However, despite its toxicity even at low doses, cisplatin remains the first-line therapy for several types of solid tumors. Thus, effective pharmacological treatments counteracting or minimizing cisplatin-induced muscle wasting are urgently needed. The dissection of the molecular pathways responsible for cisplatin-induced muscle dysfunction gives the possibility to identify novel promising therapeutic targets. In this context, the use of animal model of cisplatin-induced cachexia is very useful. Here, we report an update of the most relevant researches on the mechanisms underlying cisplatin-induced muscle wasting and on the most promising potential therapeutic options to preserve muscle mass and function.
Collapse
|
29
|
Ross CL, Zhou Y, McCall CE, Soker S, Criswell TL. The Use of Pulsed Electromagnetic Field to Modulate Inflammation and Improve Tissue Regeneration: A Review. Bioelectricity 2019; 1:247-259. [PMID: 34471827 PMCID: PMC8370292 DOI: 10.1089/bioe.2019.0026] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pulsed electromagnetic field (PEMF) is emerging as innovative treatment for regulation of inflammation, which could have significant effects on tissue regeneration. PEMF modulates inflammatory processes through the regulation of pro- and anti-inflammatory cytokine secretion during different stages of inflammatory response. Consistent outcomes in studies involving animal and human tissue have shown promise for the use of PEMF as an alternative or complementary treatment to pharmaceutical therapies. Thus, PEMF treatment could provide a novel nonpharmaceutical means of modulating inflammation in injured tissues resulting in enhanced functional recovery. This review examines the effect of PEMF on immunomodulatory cells (e.g., mesenchymal stem/stromal cells [MSCs] and macrophages [MΦ]) to better understand the potential for PEMF therapy to modulate inflammatory signaling pathways and improve tissue regeneration. This review cites published data that support the use of PEMF to improve tissue regeneration. Our studies included herein confirm anti-inflammatory effects of PEMF on MSCs and MΦ.
Collapse
Affiliation(s)
- Christina L. Ross
- Center for Integrative Medicine, Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Yu Zhou
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Charles E. McCall
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Shay Soker
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tracy L. Criswell
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
30
|
Exploring the Interface between Inflammatory and Therapeutic Glucocorticoid Induced Bone and Muscle Loss. Int J Mol Sci 2019; 20:ijms20225768. [PMID: 31744114 PMCID: PMC6888251 DOI: 10.3390/ijms20225768] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 02/02/2023] Open
Abstract
Due to their potent immunomodulatory anti-inflammatory properties, synthetic glucocorticoids (GCs) are widely utilized in the treatment of chronic inflammatory disease. In this review, we examine our current understanding of how chronic inflammation and commonly used therapeutic GCs interact to regulate bone and muscle metabolism. Whilst both inflammation and therapeutic GCs directly promote systemic osteoporosis and muscle wasting, the mechanisms whereby they achieve this are distinct. Importantly, their interactions in vivo are greatly complicated secondary to the directly opposing actions of GCs on a wide array of pro-inflammatory signalling pathways that underpin catabolic and anti-anabolic metabolism. Several clinical studies have attempted to address the net effects of therapeutic glucocorticoids on inflammatory bone loss and muscle wasting using a range of approaches. These have yielded a wide array of results further complicated by the nature of inflammatory disease, underlying the disease management and regimen of GC therapy. Here, we report the latest findings related to these pathway interactions and explore the latest insights from murine models of disease aimed at modelling these processes and delineating the contribution of pre-receptor steroid metabolism. Understanding these processes remains paramount in the effective management of patients with chronic inflammatory disease.
Collapse
|
31
|
Li Y, Zhang F, Modrak S, Little A, Zhang H. Chronic Alcohol Consumption Enhances Skeletal Muscle Wasting in Mice Bearing Cachectic Cancers: The Role of TNFα/Myostatin Axis. Alcohol Clin Exp Res 2019; 44:66-77. [PMID: 31657476 DOI: 10.1111/acer.14221] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/03/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Chronic alcohol consumption enhances cancer-associated cachexia, which is one of the major causes of decreased survival. The precise molecular mechanism of how alcohol consumption enhances cancer-associated cachexia, especially skeletal muscle loss, remains to be elucidated. METHODS We used a mouse model of chronic alcohol consumption, in which 20% (w/v) alcohol was provided as sole drinking fluid, and Lewis lung carcinoma to study the underlying mechanisms. RESULTS We found that alcohol consumption up-regulated the expression of MAFbx, MuRF-1, and LC3 in skeletal muscle, suggesting that alcohol enhanced ubiquitin-mediated proteolysis and LC3-mediated autophagy. Alcohol consumption enhanced phosphorylation of Smad2/3, p38, and ERK and decreased the phosphorylation of FOXO1. These are the signaling molecules governing protein degradation pathways. Moreover, alcohol consumption slightly up-regulated the expression of insulin receptor substrate-1, did not affect phosphatidylinositol-3 kinase, but decreased the phosphorylation of Akt and mammalian target of rapamycin (mTOR), and down-regulated the expression of Raptor and p70 ribosomal kinase S6 kinase, suggesting that alcohol impaired protein synthesis signaling pathway in skeletal muscle of tumor-bearing mice. Alcohol consumption enhanced the expression of myostatin in skeletal muscle, plasma, and tumor, but did not affect the expression of myostatin in non-tumor-bearing mice. In TNFα knockout mice, the effects of alcohol-enhanced expression of myostatin and protein degradation-related signaling molecules, and decreased protein synthesis signaling in skeletal muscle were abolished. Consequently, alcohol consumption neither affected cancer-associated cachexia nor decreased the survival of TNFα KO mice bearing cachectic cancer. CONCLUSIONS Chronic alcohol consumption enhances cancer-associated skeletal muscle loss through suppressing Akt/mTOR-mediated protein synthesis pathway and enhancing protein degradation pathways. This process is initiated by TNFα and mediated by myostatin.
Collapse
Affiliation(s)
- Yuanfei Li
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington.,Department of Oncology, (YL), The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Faya Zhang
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Samantha Modrak
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Alex Little
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Hui Zhang
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| |
Collapse
|
32
|
Abstract
Skeletal muscle atrophy is a common side effect of most human diseases. Muscle loss is not only detrimental for the quality of life but it also dramatically impairs physiological processes of the organism and decreases the efficiency of medical treatments. While hypothesized for years, the existence of an atrophying programme common to all pathologies is still incompletely solved despite the discovery of several actors and key regulators of muscle atrophy. More than a decade ago, the discovery of a set of genes, whose expression at the mRNA levels were similarly altered in different catabolic situations, opened the way of a new concept: the presence of atrogenes, i.e. atrophy-related genes. Importantly, the atrogenes are referred as such on the basis of their mRNA content in atrophying muscles, the regulation at the protein level being sometimes more complicate to elucidate. It should be noticed that the atrogenes are markers of atrophy and that their implication as active inducers of atrophy is still an open question for most of them. While the atrogene family has grown over the years, it has mostly been incremented based on data coming from rodent models. Whether the rodent atrogenes are valid for humans still remain to be established. An "atrogene" was originally defined as a gene systematically up- or down-regulated in several catabolic situations. Even if recent works often restrict this notion to the up-regulation of a limited number of proteolytic enzymes, it is important to keep in mind the big picture view. In this review, we provide an update of the validated and potential rodent atrogenes and the metabolic pathways they belong, and based on recent work, their relevance in human physio-pathological situations. We also propose a more precise definition of the atrogenes that integrates rapid recovery when catabolic stimuli are stopped or replaced by anabolic ones.
Collapse
Affiliation(s)
- Daniel Taillandier
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000, Clermont-Ferrand, France.
| | - Cécile Polge
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000, Clermont-Ferrand, France
| |
Collapse
|
33
|
Bloemberg D, Quadrilatero J. Autophagy, apoptosis, and mitochondria: molecular integration and physiological relevance in skeletal muscle. Am J Physiol Cell Physiol 2019; 317:C111-C130. [PMID: 31017800 DOI: 10.1152/ajpcell.00261.2018] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Apoptosis and autophagy are processes resulting from the integration of cellular stress and death signals. Their individual importance is highlighted by the lethality of various mouse models missing apoptosis or autophagy-related genes. In addition to their independent roles, significant overlap exists with respect to the signals that stimulate these processes as well as their effector consequences. While these cellular systems exemplify the programming redundancies that underlie many fundamental biological mechanisms, their intertwined relationship means that dysfunction can promote pathology. Although both autophagic and apoptotic signaling are active in skeletal muscle during various diseases and atrophy, their specific roles here are somewhat unique. Given our growing understanding of how specific changes at the cellular level impact whole-organism physiology, there is an equally growing interest in pharmacological manipulation of apoptosis and/or autophagy for altering human physiology and health.
Collapse
Affiliation(s)
- Darin Bloemberg
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| | - Joe Quadrilatero
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| |
Collapse
|
34
|
Fenton CG, Webster JM, Martin CS, Fareed S, Wehmeyer C, Mackie H, Jones R, Seabright AP, Lewis JW, Lai YC, Goodyear CS, Jones SW, Cooper MS, Lavery GG, Langen R, Raza K, Hardy RS. Therapeutic glucocorticoids prevent bone loss but drive muscle wasting when administered in chronic polyarthritis. Arthritis Res Ther 2019; 21:182. [PMID: 31370858 PMCID: PMC6676537 DOI: 10.1186/s13075-019-1962-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/22/2019] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Patients with rheumatoid arthritis (RA) experience extra-articular manifestations including osteoporosis and muscle wasting, which closely associate with severity of disease. Whilst therapeutic glucocorticoids (GCs) reduce inflammation in RA, their actions on muscle and bone metabolism in the context of chronic inflammation remain unclear. We utilised the TNF-tg model of chronic polyarthritis to ascertain the impact of therapeutic GCs on bone and muscle homeostasis in the context of systemic inflammation. METHODS TNF-tg and wild-type (WT) animals received either vehicle or the GC corticosterone (100 μg/ml) in drinking water at onset of arthritis. Arthritis severity and clinical parameters were measured, serum collected for ELISA and muscle and bone biopsies collected for μCT, histology and mRNA analysis. In vivo findings were examined in primary cultures of osteoblasts, osteoclasts and myotubes. RESULTS TNF-tg mice receiving GCs showed protection from inflammatory bone loss, characterised by a reduction in serum markers of bone resorption, osteoclast numbers and osteoclast activity. In contrast, muscle wasting was markedly increased in WT and TNF-tg animals receiving GCs, independently of inflammation. This was characterised by a reduction in muscle weight and fibre size, and an induction in anti-anabolic and catabolic signalling. CONCLUSIONS This study demonstrates that when given in early onset chronic polyarthritis, oral GCs partially protect against inflammatory bone loss, but induce marked muscle wasting. These results suggest that in patients with inflammatory arthritis receiving GCs, the development of interventions to manage deleterious side effects in muscle should be prioritised.
Collapse
Affiliation(s)
- C. G. Fenton
- 0000 0004 1936 7486grid.6572.6Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK ,0000 0004 1936 7486grid.6572.6Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - J. M. Webster
- 0000 0004 1936 7486grid.6572.6Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK ,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - C. S. Martin
- 0000 0004 1936 7486grid.6572.6Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - S. Fareed
- 0000 0004 1936 7486grid.6572.6Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - C. Wehmeyer
- 0000 0004 1936 7486grid.6572.6Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - H. Mackie
- 0000 0001 2193 314Xgrid.8756.cCentre of Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - R. Jones
- 0000 0004 1936 7486grid.6572.6MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| | - A. P. Seabright
- 0000 0004 1936 7486grid.6572.6School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - J. W. Lewis
- 0000 0004 1936 7486grid.6572.6Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK ,0000 0004 1936 7486grid.6572.6MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| | - Y. C. Lai
- 0000 0004 1936 7486grid.6572.6Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK ,0000 0004 1936 7486grid.6572.6MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK ,0000 0004 1936 7486grid.6572.6School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - C. S. Goodyear
- 0000 0001 2193 314Xgrid.8756.cCentre of Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - S. W Jones
- 0000 0004 1936 7486grid.6572.6Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - M. S. Cooper
- 0000 0004 1936 834Xgrid.1013.3ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - G. G. Lavery
- 0000 0004 1936 7486grid.6572.6Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK ,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - R. Langen
- 0000 0001 0481 6099grid.5012.6Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - K. Raza
- 0000 0004 1936 7486grid.6572.6Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK ,grid.412919.6Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, UK
| | - R. S. Hardy
- 0000 0004 1936 7486grid.6572.6Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK ,0000 0004 1936 7486grid.6572.6Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK ,0000 0004 1936 7486grid.6572.6MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK ,0000 0004 1936 7486grid.6572.6Institute of Clinical Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
35
|
Gueugneau M, d'Hose D, Barbé C, de Barsy M, Lause P, Maiter D, Bindels LB, Delzenne NM, Schaeffer L, Gangloff YG, Chambon C, Coudy-Gandilhon C, Béchet D, Thissen JP. Increased Serpina3n release into circulation during glucocorticoid-mediated muscle atrophy. J Cachexia Sarcopenia Muscle 2018; 9:929-946. [PMID: 29989354 PMCID: PMC6204594 DOI: 10.1002/jcsm.12315] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/13/2018] [Accepted: 04/22/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Glucocorticoids (GC) play a major role in muscle atrophy. As skeletal muscle is a secretory organ, characterization of the muscle secretome elicited by muscle atrophy should allow to better understand the cellular mechanisms and to identify circulating biomarkers of this condition. Our project aimed to identify the changes in the muscle secretome associated with GC-induced muscle atrophy and susceptible to translate into circulation. METHODS We have identified the GC-induced changes in the secretome of C2 C12 muscle cells by proteomic analysis, and then, we have determined how these changes translate into the circulation of mice or human subjects exposed to high concentrations of GC. RESULTS This approach led us to identify Serpina3n as one of the most markedly secreted protein in response to GC. Our original in vitro results were confirmed in vivo by an increased expression of Serpina3n in skeletal muscle (3.9-fold; P < 0.01) and in the serum (two-fold; P < 0.01) of mice treated with GC. We also observed increased levels of the human orthologue Serpina3 in the serum of Cushing's syndrome patients compared with healthy controls matched for age and sex (n = 9/group, 2.5-fold; P < 0.01). An increase of Serpina3n was also demonstrated in muscle atrophy models mediated by GC such as cancer cachexia (four-fold; P < 0.01), sepsis (12.5-fold; P < 0.001), or diabetes (two-fold; P < 0.01). In contrast, levels of Serpina3n both in skeletal muscle and in the circulation were reduced in several models of muscle hypertrophy induced by myostatin inhibition (P < 0.01). Furthermore, a cluster of data suggests that the regulation of muscle Serpina3n involves mTOR, an essential determinant of the muscle cell size. CONCLUSIONS Taken together, these data suggest that Serpina3n may represent a circulating biomarker of muscle atrophy associated to GC and, broadly, a reflection of dynamic changes in muscle mass.
Collapse
Affiliation(s)
- Marine Gueugneau
- Pole of Endocrinology, Diabetes and Nutrition, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium.,INRA, UMR1019, Université Clermont Auvergne, UNH, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France
| | - Donatienne d'Hose
- Pole of Endocrinology, Diabetes and Nutrition, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Caroline Barbé
- Pole of Endocrinology, Diabetes and Nutrition, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Marie de Barsy
- Pole of Endocrinology, Diabetes and Nutrition, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Pascale Lause
- Pole of Endocrinology, Diabetes and Nutrition, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Dominique Maiter
- Pole of Endocrinology, Diabetes and Nutrition, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), Université catholique de Louvain, Brussels, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), Université catholique de Louvain, Brussels, Belgium
| | - Laurent Schaeffer
- INMG, CNRS, UMR 5310, INSERM U1217, LBMC, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Yann-Gaël Gangloff
- INMG, CNRS, UMR 5310, INSERM U1217, LBMC, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Christophe Chambon
- INRA, Plateforme d'Exploration du Métabolisme Composante Protéomique, Saint Genès Champanelle, France
| | - Cécile Coudy-Gandilhon
- INRA, UMR1019, Université Clermont Auvergne, UNH, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France
| | - Daniel Béchet
- INRA, UMR1019, Université Clermont Auvergne, UNH, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France
| | - Jean-Paul Thissen
- Pole of Endocrinology, Diabetes and Nutrition, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
36
|
Pinheiro-Dardis CM, Gutierres VO, Assis RP, Peviani SM, Delfino GB, Durigan JLQ, Salvini TDF, Baviera AM, Brunetti IL. Insulin treatment reverses the increase in atrogin-1 expression in atrophied skeletal muscles of diabetic rats with acute joint inflammation. Ther Clin Risk Manag 2018; 14:275-286. [PMID: 29497304 PMCID: PMC5818839 DOI: 10.2147/tcrm.s142948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background The aim of this study was to evaluate the changes in biomarkers of skeletal muscle proteolysis (atrogin-1, muscle RING finger-1 protein [MuRF-1]) and inflammation (nuclear factor kappa-B) in skeletal muscles of rats under two catabolic conditions, diabetes mellitus (DM) and acute joint inflammation, and the effects of insulin therapy. Materials and methods Male Wistar rats were divided into groups without diabetes – normal (N), saline (NS), or ι-carrageenan (NCa) injection into the tibiotarsal joint – and groups with diabetes – diabetes (D), plus insulin (DI), saline (DS), or ι-carrageenan (DCa) injection into the tibiotarsal joint, or ι-carrageenan injection and treatment with insulin (DCaI). Three days after ι-carrageenan injection (17 days after diabetes induction), tibialis anterior (TA) and soleus (SO) skeletal muscles were used for analysis. Results DM alone caused a significant decrease in the mass of TA and SO muscles, even with low levels of atrogenes (atrogin-1, MuRF-1), which could be interpreted as an adaptive mechanism to spare muscle proteins under this catabolic condition. The loss of muscle mass was exacerbated when ι-carrageenan was administered in the joints of diabetic rats, in association with increased expression of atrogin-1, MuRF-1, and nuclear factor kappa-B. Treatment with insulin prevented the increase in atrogin-1 (TA, SO) and the loss of muscle mass (SO) in diabetic-carrageenan rats; in comparison with TA, SO muscle was more responsive to the anabolic actions of insulin. Conclusion Acute joint inflammation overcame the adaptive mechanism in diabetic rats to prevent excessive loss of muscle mass, worsening the catabolic state. The treatment of diabetic-carrageenan rats with insulin prevented the loss of skeletal muscle mass mainly via atrogin-1 inhibition. Under the condition of DM and inflammation, muscles with the prevalence of slow-twitch, type 1 fibers were more responsive to insulin treatment, recovering the ability to grow.
Collapse
Affiliation(s)
- Clara Maria Pinheiro-Dardis
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Department of Clinical Analysis, Araraquara, São Paulo, Brazil
| | - Vânia Ortega Gutierres
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Department of Clinical Analysis, Araraquara, São Paulo, Brazil
| | - Renata Pires Assis
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Department of Clinical Analysis, Araraquara, São Paulo, Brazil
| | - Sabrina Messa Peviani
- Federal University of São Carlos (UFSCar), Department of Physical Therapy, São Carlos, São Paulo, Brazil
| | - Gabriel Borges Delfino
- Federal University of São Carlos (UFSCar), Department of Physical Therapy, São Carlos, São Paulo, Brazil
| | | | - Tania de Fátima Salvini
- Federal University of São Carlos (UFSCar), Department of Physical Therapy, São Carlos, São Paulo, Brazil
| | - Amanda Martins Baviera
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Department of Clinical Analysis, Araraquara, São Paulo, Brazil
| | - Iguatemy Lourenço Brunetti
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Department of Clinical Analysis, Araraquara, São Paulo, Brazil
| |
Collapse
|
37
|
The Role of IGF-1 Signaling in Skeletal Muscle Atrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:109-137. [PMID: 30390250 DOI: 10.1007/978-981-13-1435-3_6] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Insulin-like growth factor 1 (IGF-1) is a key anabolic growth factor stimulating phosphatidylinositol 3-kinase (PI3K)/Akt signaling which is well known for regulating muscle hypertrophy. However, the role of IGF-1 in muscle atrophy is less clear. This review provides an overview of the mechanisms via which IGF-1 signaling is implicated in several conditions of muscle atrophy and via which mechanisms protein turnover is altered. IGF-1/PI3K/Akt signaling stimulates the rate of protein synthesis via p70S6Kinase and p90 ribosomal S6 kinase and negatively regulates protein degradation, predominantly by its inhibiting effect on proteasomal and lysosomal protein degradation. Caspase-dependent protein degradation is also attenuated by IGF/PI3K/Akt signaling, whereas evidence for an effect on calpain-dependent protein degradation is inconclusive. IGF-1/PI3K/Akt signaling reduces during denervation-, unloading-, and joint immobilization-induced muscle atrophy, whereas IGF-1/PI3K/Akt signaling seems unaltered during aging-associated muscle atrophy. During denervation and aging, IGF-1 overexpression or injection counteracts denervation- and aging-associated muscle atrophy, despite enhanced anabolic resistance with regard to IGF-1 signaling with aging. It remains unclear whether pharmacological stimulation of IGF-1/PI3K/Akt signaling attenuates immobilization- or unloading-induced muscle atrophy. Exploration of the possibilities to interfere with IGF-1/PI3K/Akt signaling reveals that microRNAs targeting IGF-1 signaling components are promising targets to counterbalance muscle atrophy. Overall, the findings summarized in this review show that in disuse conditions, but not with aging, IGF-1/PI3K/Akt signaling is attenuated and that in some conditions stimulation of this pathway may alleviate skeletal muscle atrophy.
Collapse
|
38
|
Sipos F, Székely H, Kis ID, Tulassay Z, Műzes G. Relation of the IGF/IGF1R system to autophagy in colitis and colorectal cancer. World J Gastroenterol 2017; 23:8109-8119. [PMID: 29290648 PMCID: PMC5739918 DOI: 10.3748/wjg.v23.i46.8109] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 10/28/2017] [Accepted: 12/04/2017] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome (MetS), as a chronic inflammatory disorder has a potential role in the development of inflammatory and cancerous complications of the colonic tissue. The interaction of DNA damage and inflammation is affected by the insulin-like growth factor 1 receptor (IGF1R) signaling pathway. The IGF1R pathway has been reported to regulate autophagy, as well, but sometimes through a bidirectional context. Targeting the IGF1R-autophagy crosstalk could represent a promising strategy for the development of new antiinflammatory and anticancer therapies, and may help for subjects suffering from MetS who are at increased risk of colorectal cancer. However, therapeutic responses to targeted therapies are often shortlived, since a signaling crosstalk of IGF1R with other receptor tyrosine kinases or autophagy exists, leading to acquired cellular resistance to therapy. From a pharmacological point of view, it is attractive to speculate that synergistic benefits could be achieved by inhibition of one of the key effectors of the IGF1R pathway, in parallel with the pharmacological stimulation of the autophagy machinery, but cautiousness is also required, because pharmacologic IGF1R modulation can initiate additional, sometimes unfavorable biologic effects.
Collapse
Affiliation(s)
- Ferenc Sipos
- 2nd Department of Internal Medicine, Semmelweis University, Budapest 1088, Hungary
| | - Hajnal Székely
- 2nd Department of Internal Medicine, Semmelweis University, Budapest 1088, Hungary
| | - Imre Dániel Kis
- Faculty of Medicine, Semmelweis University, Budapest 1088, Hungary
| | - Zsolt Tulassay
- Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest 1088, Hungary
| | - Györgyi Műzes
- 2nd Department of Internal Medicine, Semmelweis University, Budapest 1088, Hungary
| |
Collapse
|
39
|
Dalle S, Rossmeislova L, Koppo K. The Role of Inflammation in Age-Related Sarcopenia. Front Physiol 2017; 8:1045. [PMID: 29311975 PMCID: PMC5733049 DOI: 10.3389/fphys.2017.01045] [Citation(s) in RCA: 390] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/29/2017] [Indexed: 12/25/2022] Open
Abstract
Many physiological changes occur with aging. These changes often, directly or indirectly, result in a deterioration of the quality of life and even in a shortening of life expectancy. Besides increased levels of reactive oxygen species, DNA damage and cell apoptosis, another important factor affecting the aging process involves a systemic chronic low-grade inflammation. This condition has already been shown to be interrelated with several (sub)clinical conditions, such as insulin resistance, atherosclerosis and Alzheimer's disease. Recent evidence, however, shows that chronic low-grade inflammation also contributes to the loss of muscle mass, strength and functionality, referred to as sarcopenia, as it affects both muscle protein breakdown and synthesis through several signaling pathways. Classic interventions to counteract age-related muscle wasting mainly focus on resistance training and/or protein supplementation to overcome the anabolic inflexibility from which elderly suffer. Although the elderly benefit from these classic interventions, the therapeutic potential of anti-inflammatory strategies is of great interest, as these might add up to/support the anabolic effect of resistance exercise and/or protein supplementation. In this review, the molecular interaction between inflammation, anabolic sensitivity and muscle protein metabolism in sarcopenic elderly will be addressed.
Collapse
Affiliation(s)
- Sebastiaan Dalle
- Exercise Physiology Research Group, Department of Kinesiology, KU Leuven, Leuven, Belgium
| | - Lenka Rossmeislova
- Department for the Study of Obesity and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Katrien Koppo
- Exercise Physiology Research Group, Department of Kinesiology, KU Leuven, Leuven, Belgium
| |
Collapse
|
40
|
Verzola D, Bonanni A, Sofia A, Montecucco F, D'Amato E, Cademartori V, Parodi EL, Viazzi F, Venturelli C, Brunori G, Garibotto G. Toll-like receptor 4 signalling mediates inflammation in skeletal muscle of patients with chronic kidney disease. J Cachexia Sarcopenia Muscle 2017; 8:131-144. [PMID: 27897392 PMCID: PMC5326826 DOI: 10.1002/jcsm.12129] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 05/16/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Inflammation in skeletal muscle is implicated in the pathogenesis of insulin resistance and cachexia but why uremia up-regulates pro-inflammatory cytokines is unknown. Toll-like receptors (TLRs) regulate locally the innate immune responses, but it is unknown whether in chronic kidney disease (CKD) TLR4 muscle signalling is altered. The aim of the study is to investigate whether in CKD muscle, TLRs had abnormal function and may be involved in transcription of pro-inflammatory cytokine. METHODS TLR4, phospho-p65, phospho-ikBα, tumour necrosis factor (TNF)-α, phospho p38, Murf 1, and atrogin were studied in skeletal muscle from nondiabetic CKD stage 5 patients (n = 29) and controls (n = 14) by immunohistochemistry, western blot, and RT-PCR. Muscle cell cultures (C2C12) exposed to uremic serum were employed to study TLR4 expression (western blot and RT-PCR) and TLR-driven signalling. TLR4 signalling was abrogated by a small molecule chemical inhibitor or TLR4 siRNA. Phospho AKT and phospho p38 were evaluated by western blot. RESULTS CKD subjects had elevated TLR4 gene and protein expression. Also expression of NFkB, p38 MAPK and the NFkB-regulated gene TNF-α was increased. At multivariate analysis, TLR4 protein content was predicted by eGFR and Subjective Global Assessment, suggesting that the progressive decline in renal function and wasting mediate TLR4 activation. In C2C12, uremic serum increased TLR4 as well as TNF-α and down-regulated pAkt. These effects were prevented by blockade of TLR4. CONCLUSIONS CKD promotes muscle inflammation through an up-regulation of TLR4, which may activate downward inflammatory signals such as TNF-α and NFkB-regulated genes.
Collapse
Affiliation(s)
- Daniela Verzola
- Department of Internal Medicine, Nephrology Division and First Clinic of Internal Medicine, University of Genova and IRCCS AOU San Martino-IST, and Ospedale Santa ChiaraI, Trento, Italy
| | - Alice Bonanni
- Department of Internal Medicine, Nephrology Division and First Clinic of Internal Medicine, University of Genova and IRCCS AOU San Martino-IST, and Ospedale Santa ChiaraI, Trento, Italy
| | - Antonella Sofia
- Department of Internal Medicine, Nephrology Division and First Clinic of Internal Medicine, University of Genova and IRCCS AOU San Martino-IST, and Ospedale Santa ChiaraI, Trento, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, Nephrology Division and First Clinic of Internal Medicine, University of Genova and IRCCS AOU San Martino-IST, and Ospedale Santa ChiaraI, Trento, Italy
| | - Elena D'Amato
- Department of Internal Medicine, Nephrology Division and First Clinic of Internal Medicine, University of Genova and IRCCS AOU San Martino-IST, and Ospedale Santa ChiaraI, Trento, Italy
| | - Valeria Cademartori
- Department of Internal Medicine, Nephrology Division and First Clinic of Internal Medicine, University of Genova and IRCCS AOU San Martino-IST, and Ospedale Santa ChiaraI, Trento, Italy
| | - Emanuele Luigi Parodi
- Department of Internal Medicine, Nephrology Division and First Clinic of Internal Medicine, University of Genova and IRCCS AOU San Martino-IST, and Ospedale Santa ChiaraI, Trento, Italy
| | - Francesca Viazzi
- Department of Internal Medicine, Nephrology Division and First Clinic of Internal Medicine, University of Genova and IRCCS AOU San Martino-IST, and Ospedale Santa ChiaraI, Trento, Italy
| | - Chiara Venturelli
- Department of Internal Medicine, Nephrology Division and First Clinic of Internal Medicine, University of Genova and IRCCS AOU San Martino-IST, and Ospedale Santa ChiaraI, Trento, Italy
| | - Giuliano Brunori
- Department of Internal Medicine, Nephrology Division and First Clinic of Internal Medicine, University of Genova and IRCCS AOU San Martino-IST, and Ospedale Santa ChiaraI, Trento, Italy
| | - Giacomo Garibotto
- Department of Internal Medicine, Nephrology Division and First Clinic of Internal Medicine, University of Genova and IRCCS AOU San Martino-IST, and Ospedale Santa ChiaraI, Trento, Italy
| |
Collapse
|
41
|
Jeong J, Park CH, Kim I, Kim YH, Yoon JM, Kim KS, Kim JB. Korean mistletoe (Viscum album coloratum) extract regulates gene expression related to muscle atrophy and muscle hypertrophy. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:68. [PMID: 28109285 PMCID: PMC5251312 DOI: 10.1186/s12906-017-1575-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 01/11/2017] [Indexed: 02/05/2023]
Abstract
Background Korean mistletoe (Viscum album coloratum) is a semi-parasitic plant that grows on various trees and has a diverse range of effects on biological functions, being implicated in having anti-tumor, immunostimulatory, anti-diabetic, and anti-obesity properties. Recently, we also reported that Korean mistletoe extract (KME) improves endurance exercise in mice, suggesting its beneficial roles in enhancing the capacity of skeletal muscle. Methods We examined the expression pattern of several genes concerned with muscle physiology in C2C12 myotubes cells to identify whether KME inhibits muscle atrophy or promotes muscle hypertrophy. We also investigated these effects of KME in denervated mice model. Results Interestingly, KME induced the mRNA expression of SREBP-1c, PGC-1α, and GLUT4, known positive regulators of muscle hypertrophy, in C2C12 cells. On the contrary, KME reduced the expression of Atrogin-1, which is directly involved in the induction of muscle atrophy. In animal models, KME mitigated the decrease of muscle weight in denervated mice. The expression of Atrogin-1 was also diminished in those mice. Moreover, KME enhanced the grip strength and muscle weight in long-term feeding mice. Conclusions Our results suggest that KME has beneficial effects on muscle atrophy and muscle hypertrophy.
Collapse
|
42
|
Liu Y, Wang X, Leng W, Pi D, Tu Z, Zhu H, Shi H, Li S, Hou Y, Hu CAA. Aspartate inhibits LPS-induced MAFbx and MuRF1 expression in skeletal muscle in weaned pigs by regulating Akt, AMPKα and FOXO1. Innate Immun 2016; 23:34-43. [PMID: 28064564 DOI: 10.1177/1753425916673443] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Infection and inflammation can result in the rapid loss of muscle mass and myofibrillar proteins (muscle atrophy). In addition, aspartate (Asp) is necessary for protein synthesis in mammalian cells. We hypothesized that Asp could attenuate LPS-induced muscle atrophy in a piglet model. Twenty-four weaning piglets were allotted to four treatments, including non-challenged control, LPS challenged control, LPS+0.5% Asp and LPS+1.0% Asp. On d 21, the piglets were injected with i.p. injection of LPS (100 ug/kg BM) or saline. At 4 h post-injection, blood, gastrocnemius and longissimus dorsi muscles samples were collected for biochemical and molecular analyses. Asp decreased the concentrations of cortisol and glucagon in plasma. In addition, Asp increased protein and RNA contents in muscles, and decreased mRNA expression of muscle atrophy F-box (MAFbx) and muscle RING finger 1 (MuRF1). Moreover, Asp decreased phosphorylation of AMPKα but increased phosphorylation of Akt and Forkhead Box O (FOXO) 1 in the muscles. Our results indicate that Asp suppresses LPS-induced MAFbx and MuRF1 expression via activation of Akt signaling, and inhibition of AMPKα and FOXO1 signaling.
Collapse
Affiliation(s)
- Yulan Liu
- 1 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People's Republic of China
| | - Xiuying Wang
- 1 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People's Republic of China
| | - Weibo Leng
- 1 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People's Republic of China
| | - Dingan Pi
- 1 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People's Republic of China
| | - Zhixiao Tu
- 1 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People's Republic of China
| | - Huiling Zhu
- 1 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People's Republic of China
| | - Haifeng Shi
- 1 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People's Republic of China
| | - Shuang Li
- 1 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People's Republic of China
| | - Yongqing Hou
- 1 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People's Republic of China
| | - Chien-An Andy Hu
- 1 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People's Republic of China
- 2 Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| |
Collapse
|
43
|
Ham DJ, Caldow MK, Chhen V, Chee A, Wang X, Proud CG, Lynch GS, Koopman R. Glycine restores the anabolic response to leucine in a mouse model of acute inflammation. Am J Physiol Endocrinol Metab 2016; 310:E970-81. [PMID: 27094036 DOI: 10.1152/ajpendo.00468.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 04/18/2016] [Indexed: 12/23/2022]
Abstract
Amino acids, especially leucine, potently stimulate protein synthesis and reduce protein breakdown in healthy skeletal muscle and as a result have received considerable attention as potential treatments for muscle wasting. However, the normal anabolic response to amino acids is impaired during muscle-wasting conditions. Although the exact mechanisms of this anabolic resistance are unclear, inflammation and ROS are believed to play a central role. The nonessential amino acid glycine has anti-inflammatory and antioxidant properties and preserves muscle mass in calorie-restricted and tumor-bearing mice. We hypothesized that glycine would restore the normal muscle anabolic response to amino acids under inflammatory conditions. Relative rates of basal and leucine-stimulated protein synthesis were measured using SUnSET methodology 4 h after an injection of 1 mg/kg lipopolysaccharide (LPS). Whereas leucine failed to stimulate muscle protein synthesis in LPS-treated mice pretreated with l-alanine (isonitrogenous control), leucine robustly stimulated protein synthesis (+51%) in mice pretreated with 1 g/kg glycine. The improvement in leucine-stimulated protein synthesis was accompanied by a higher phosphorylation status of mTOR, S6, and 4E-BP1 compared with l-alanine-treated controls. Despite its known anti-inflammatory action in inflammatory cells, glycine did not alter the skeletal muscle inflammatory response to LPS in vivo or in vitro but markedly reduced DHE staining intensity, a marker of oxidative stress, in muscle cross-sections and attenuated LPS-induced wasting in C2C12 myotubes. Our observations in male C57BL/6 mice suggest that glycine may represent a promising nutritional intervention for the attenuation of skeletal muscle wasting.
Collapse
Affiliation(s)
- Daniel J Ham
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Marissa K Caldow
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Victoria Chhen
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Annabel Chee
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Xuemin Wang
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, South Austrailia, Australia; and School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Christopher G Proud
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, South Austrailia, Australia; and School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Gordon S Lynch
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - René Koopman
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia;
| |
Collapse
|
44
|
Liu Y, Wang X, Wu H, Chen S, Zhu H, Zhang J, Hou Y, Hu CAA, Zhang G. Glycine enhances muscle protein mass associated with maintaining Akt-mTOR-FOXO1 signaling and suppressing TLR4 and NOD2 signaling in piglets challenged with LPS. Am J Physiol Regul Integr Comp Physiol 2016; 311:R365-73. [PMID: 27225947 DOI: 10.1152/ajpregu.00043.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/16/2016] [Indexed: 12/25/2022]
Abstract
Pro-inflammatory cytokines play a critical role in the pathophysiology of muscle atrophy. We hypothesized that glycine exerted an anti-inflammatory effect and alleviated lipopolysaccharide (LPS)-induced muscle atrophy in piglets. Pigs were assigned to four treatments including the following: 1) nonchallenged control, 2) LPS-challenged control, 3) LPS+1.0% glycine, and 4) LPS+2.0% glycine. After receiving the control, 1.0 or 2.0% glycine-supplemented diets, piglets were treated with either saline or LPS. At 4 h after treatment with saline or LPS, blood and muscle samples were harvested. We found that 1.0 or 2.0% glycine increased protein/DNA ratio, protein content, and RNA/DNA ratio in gastrocnemius or longissimus dorsi (LD) muscles. Glycine also resulted in decreased mRNA expression of muscle atrophy F-box (MAFbx) and muscle RING finger 1 (MuRF1) in gastrocnemius muscle. In addition, glycine restored the phosphorylation of Akt, mammalian target of rapamycin (mTOR), eukaryotic initiation factor 4E binding protein 1 (4E-BP1), and Forkhead Box O 1 (FOXO1) in gastrocnemius or LD muscles. Furthermore, glycine resulted in decreased plasma tumor necrosis factor-α (TNF-α) concentration and muscle TNF-α mRNA abundance. Moreover, glycine resulted in decreased mRNA expresson of Toll-like receptor 4 (TLR4), nucleotide-binding oligomerization domain protein 2 (NOD2), and their respective downstream molecules in gastrocnemius or LD muscles. These results indicate glycine enhances muscle protein mass under an inflammatory condition. The beneficial roles of glycine on the muscle are closely associated with maintaining Akt-mTOR-FOXO1 signaling and suppressing the activation of TLR4 and/or NOD2 signaling pathways.
Collapse
Affiliation(s)
- Yulan Liu
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China;
| | - Xiuying Wang
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Huanting Wu
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Shaokui Chen
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Huiling Zhu
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Jing Zhang
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Yongqing Hou
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Chien-An Andy Hu
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China; Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico; and
| | - Guolong Zhang
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China; Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma
| |
Collapse
|
45
|
Gómez-SanMiguel AB, Villanúa MÁ, Martín AI, López-Calderón A. D-TRP(8)-γMSH Prevents the Effects of Endotoxin in Rat Skeletal Muscle Cells through TNFα/NF-KB Signalling Pathway. PLoS One 2016; 11:e0155645. [PMID: 27177152 PMCID: PMC4866687 DOI: 10.1371/journal.pone.0155645] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 05/02/2016] [Indexed: 02/07/2023] Open
Abstract
Sepsis induces anorexia and muscle wasting secondary to an increase in muscle proteolysis. Melanocyte stimulating hormones (MSH) is a family of peptides that have potent anti-inflammatory effects. Melanocortin receptor-3 (MC3-R) has been reported as the predominant anti-inflammatory receptor for melanocortins. The aim of this work was to analyse whether activation of MC3-R, by administration of its agonist D-Trp(8)-γMSH, is able to modify the response of skeletal muscle to inflammation induced by lipopolysaccharide endotoxin (LPS) or TNFα. Adult male rats were injected with 250 μg/kg LPS and/or 500 μg/kg D-Trp(8)-γMSH 17:00 h and at 8:00 h the following day, and euthanized 4 hours afterwards. D-Trp(8)-γMSH decreased LPS-induced anorexia and prevented the stimulatory effect of LPS on hypothalamic IL-1β, COX-2 and CRH as well as on serum ACTH and corticosterone. Serum IGF-I and its expression in liver and gastrocnemius were decreased in rats injected with LPS, but not in those that also received D-Trp(8)-γMSH. However, D-Trp(8)-γMSH was unable to modify the effect of LPS on IGFBP-3. In the gastrocnemius D-Trp(8)-γMSH blocked LPS-induced decrease in pAkt, pmTOR, MHC I and MCH II, as well as the increase in pNF-κB(p65), FoxO1, FoxO3, LC3b, Bnip-3, Gabarap1, atrogin-1, MuRF1 and in LC3a/b lipidation. In L6 myotube cultures, D-Trp(8)-γMSH was able to prevent TNFα-induced increase of NF-κB(p65) phosphorylation and decrease of Akt phosphorylation as well as of IGF-I and MHC I expression. These data suggest that MC3-R activation prevents the effect of endotoxin on skeletal wasting by modifying inflammation, corticosterone and IGF-I responses and also by directly acting on muscle cells through the TNFα/NF-κB(p65) pathway.
Collapse
Affiliation(s)
- Ana Belén Gómez-SanMiguel
- Department of Physiology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - María Ángeles Villanúa
- Department of Physiology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Ana Isabel Martín
- Department of Physiology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Asunción López-Calderón
- Department of Physiology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- * E-mail:
| |
Collapse
|
46
|
Rivas DA, McDonald DJ, Rice NP, Haran PH, Dolnikowski GG, Fielding RA. Diminished anabolic signaling response to insulin induced by intramuscular lipid accumulation is associated with inflammation in aging but not obesity. Am J Physiol Regul Integr Comp Physiol 2016; 310:R561-9. [PMID: 26764052 DOI: 10.1152/ajpregu.00198.2015] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 01/11/2016] [Indexed: 12/24/2022]
Abstract
The loss of skeletal muscle mass is observed in many pathophysiological conditions, including aging and obesity. The loss of muscle mass and function with aging is defined as sarcopenia and is characterized by a mismatch between skeletal muscle protein synthesis and breakdown. Characteristic metabolic features of both aging and obesity are increases in intramyocellular lipid (IMCL) content in muscle. IMCL accumulation may play a mechanistic role in the development of anabolic resistance and the progression of muscle atrophy in aging and obesity. In the present study, aged and high-fat fed mice were used to determine mechanisms leading to muscle loss. We hypothesized the accumulation of bioactive lipids in skeletal muscle, such as ceramide or diacylglycerols, leads to insulin resistance with aging and obesity and the inability to activate protein synthesis, contributing to skeletal muscle loss. We report a positive association between bioactive lipid accumulation and the loss of lean mass and muscle strength. Obese and aged animals had significantly higher storage of ceramide and diacylglycerol compared with young. Furthermore, there was an attenuated insulin response in components of the mTOR anabolic signaling pathway. We also observed differential increases in the expression of inflammatory cytokines and the phosphorylation of IκBα with aging and obesity. These data challenge the accepted role of increased inflammation in obesity-induced insulin resistance in skeletal muscle. Furthermore, we have now established IκBα with a novel function in aging-associated muscle loss that may be independent of its previously understood role as an NF-κB inhibitor.
Collapse
Affiliation(s)
- Donato A Rivas
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Tufts University, Boston, Massachusetts; and
| | - Devin J McDonald
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Tufts University, Boston, Massachusetts; and
| | - Nicholas P Rice
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Tufts University, Boston, Massachusetts; and
| | - Prashanth H Haran
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Tufts University, Boston, Massachusetts; and
| | - Gregory G Dolnikowski
- Mass Spectrometry Unit; Jean Mayer U.S. Department of Agriculture, Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts
| | - Roger A Fielding
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Tufts University, Boston, Massachusetts; and
| |
Collapse
|
47
|
Asano S, Arvapalli R, Manne NDPK, Maheshwari M, Ma B, Rice KM, Selvaraj V, Blough ER. Cerium oxide nanoparticle treatment ameliorates peritonitis-induced diaphragm dysfunction. Int J Nanomedicine 2015; 10:6215-25. [PMID: 26491293 PMCID: PMC4599716 DOI: 10.2147/ijn.s89783] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The severe inflammation observed during sepsis is thought to cause diaphragm dysfunction, which is associated with poor patient prognosis. Cerium oxide (CeO2) nanoparticles have been posited to exhibit anti-inflammatory and antioxidative activities suggesting that these particles may be of potential use for the treatment of inflammatory disorders. To investigate this possibility, Sprague Dawley rats were randomly assigned to the following groups: sham control, CeO2 nanoparticle treatment only (0.5 mg/kg iv), sepsis, and sepsis+CeO2 nanoparticles. Sepsis was induced by the introduction of cecal material (600 mg/kg) directly into the peritoneal cavity. Nanoparticle treatment decreased sepsis-associated impairments in diaphragmatic contractile (P(o)) function (sham: 25.6±1.6 N/cm(2) vs CeO2: 23.4±0.8 N/cm(2) vs Sep: 15.9±1.0 N/cm(2) vs Sep+CeO2: 20.0±1.0 N/cm(2), P<0.05). These improvements in diaphragm contractile function were accompanied by a normalization of protein translation signaling (Akt, FOXO-1, and 4EBP1), diminished proteolysis (caspase 8 and ubiquitin levels), and decreased inflammatory signaling (Stat3 and iNOS). Histological analysis suggested that nanoparticle treatment was associated with diminished sarcolemma damage and diminished inflammatory cell infiltration. These data indicate CeO2 nanoparticles may improve diaphragmatic function in the septic laboratory rat.
Collapse
Affiliation(s)
- Shinichi Asano
- Center for Diagnostic Nanosystems, Marshall University, Huntington, WV, USA ; Department of Pharmacology, Pharmaceutical Sciences and Research, School of Pharmacy, Marshall University, Huntington, WV, USA
| | | | - Nandini D P K Manne
- Center for Diagnostic Nanosystems, Marshall University, Huntington, WV, USA ; Department of Pharmacology, Pharmaceutical Sciences and Research, School of Pharmacy, Marshall University, Huntington, WV, USA
| | - Mani Maheshwari
- Center for Diagnostic Nanosystems, Marshall University, Huntington, WV, USA ; Department of Pharmacology, Physiology and Toxicology, Joan C Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Bing Ma
- Center for Diagnostic Nanosystems, Marshall University, Huntington, WV, USA ; Department of Pharmacology, Pharmaceutical Sciences and Research, School of Pharmacy, Marshall University, Huntington, WV, USA
| | - Kevin M Rice
- Center for Diagnostic Nanosystems, Marshall University, Huntington, WV, USA
| | - Vellaisamy Selvaraj
- Center for Diagnostic Nanosystems, Marshall University, Huntington, WV, USA ; Department of Pharmacology, Pharmaceutical Sciences and Research, School of Pharmacy, Marshall University, Huntington, WV, USA
| | - Eric R Blough
- Center for Diagnostic Nanosystems, Marshall University, Huntington, WV, USA ; Department of Pharmacology, Pharmaceutical Sciences and Research, School of Pharmacy, Marshall University, Huntington, WV, USA ; Department of Pharmacology, Physiology and Toxicology, Joan C Edwards School of Medicine, Marshall University, Huntington, WV, USA
| |
Collapse
|
48
|
Gallo D, Gesmundo I, Trovato L, Pera G, Gargantini E, Minetto MA, Ghigo E, Granata R. GH-Releasing Hormone Promotes Survival and Prevents TNF-α-Induced Apoptosis and Atrophy in C2C12 Myotubes. Endocrinology 2015; 156:3239-52. [PMID: 26110916 DOI: 10.1210/en.2015-1098] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Skeletal muscle atrophy is a consequence of different chronic diseases, including cancer, heart failure, and diabetes, and also occurs in aging and genetic myopathies. It results from an imbalance between anabolic and catabolic processes, and inflammatory cytokines, such as TNF-α, have been found elevated in muscle atrophy and implicated in its pathogenesis. GHRH, in addition to stimulating GH secretion from the pituitary, exerts survival and antiapoptotic effects in different cell types. Moreover, we and others have recently shown that GHRH displays antiapoptotic effects in isolated cardiac myocytes and protects the isolated heart from ischemia/reperfusion injury and myocardial infarction in vivo. On these bases, we investigated the effects of GHRH on survival and apoptosis of TNF-α-treated C2C12 myotubes along with the underlying mechanisms. GHRH increased myotube survival and prevented TNF-α-induced apoptosis through GHRH receptor-mediated mechanisms. These effects involved activation of phosphoinositide 3-kinase/Akt pathway and inactivation of glycogen synthase kinase-3β, whereas mammalian target of rapamycin was unaffected. GHRH also increased the expression of myosin heavy chain and the myogenic transcription factor myogenin, which were both reduced by the cytokine. Furthermore, GHRH inhibited TNF-α-induced expression of nuclear factor-κB, calpain, and muscle ring finger1, which are all involved in muscle protein degradation. In summary, these results indicate that GHRH exerts survival and antiapoptotic effects in skeletal muscle cells through the activation of anabolic pathways and the inhibition of proteolytic routes. Overall, our findings suggest a novel therapeutic role for GHRH in the treatment of muscle atrophy-associated diseases.
Collapse
Affiliation(s)
- Davide Gallo
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Iacopo Gesmundo
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Letizia Trovato
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Giulia Pera
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Eleonora Gargantini
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Marco Alessandro Minetto
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Ezio Ghigo
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Riccarda Granata
- Laboratory of Molecular and Cellular Endocrinology (D.G., I.G., L.T., G.P., E.Ga., R.G.), and Division of Endocrinology, Diabetes, and Metabolism (D.G., I.G., L.T., G.P., E.Ga., M.A.M., E.Gh., R.G.), Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| |
Collapse
|
49
|
Systemic inflammation in chronic obstructive pulmonary disease and lung cancer: common driver of pulmonary cachexia? Curr Opin Support Palliat Care 2015; 8:339-45. [PMID: 25158627 DOI: 10.1097/spc.0000000000000088] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW In this article, a putative role of systemic inflammation as a driver of pulmonary cachexia induced by either chronic obstructive pulmonary disease or nonsmall cell lung cancer is reviewed. Gaps in current translational research approaches are discussed and alternative strategies are proposed to provide new insights. RECENT FINDINGS Activation of the ubiquitin proteasome system has generally been considered a cause of pulmonary cachexia, but current animal models lack specificity and evidence is lacking in nonsmall cell lung cancer and conflicting in chronic obstructive pulmonary disease patients. Recent studies have shown activation of the autophagy-lysosome pathway in both nonsmall cell lung cancer and chronic obstructive pulmonary disease. Myonuclear loss, as a consequence of increased apoptotic events in myofibers, has been suggested in cancer-cachexia-associated muscle atrophy. Plasma transfer on myotube cultures can be used to detect early inflammatory signals in patients and presence of atrophy-inducing activity within the circulation. SUMMARY Comparative clinical research between nonsmall cell lung cancer and chronic obstructive pulmonary disease in different disease stages is useful to unravel disease-specific versus common denominators of pulmonary cachexia.
Collapse
|
50
|
Friedrich O, Reid MB, Van den Berghe G, Vanhorebeek I, Hermans G, Rich MM, Larsson L. The Sick and the Weak: Neuropathies/Myopathies in the Critically Ill. Physiol Rev 2015; 95:1025-109. [PMID: 26133937 PMCID: PMC4491544 DOI: 10.1152/physrev.00028.2014] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Critical illness polyneuropathies (CIP) and myopathies (CIM) are common complications of critical illness. Several weakness syndromes are summarized under the term intensive care unit-acquired weakness (ICUAW). We propose a classification of different ICUAW forms (CIM, CIP, sepsis-induced, steroid-denervation myopathy) and pathophysiological mechanisms from clinical and animal model data. Triggers include sepsis, mechanical ventilation, muscle unloading, steroid treatment, or denervation. Some ICUAW forms require stringent diagnostic features; CIM is marked by membrane hypoexcitability, severe atrophy, preferential myosin loss, ultrastructural alterations, and inadequate autophagy activation while myopathies in pure sepsis do not reproduce marked myosin loss. Reduced membrane excitability results from depolarization and ion channel dysfunction. Mitochondrial dysfunction contributes to energy-dependent processes. Ubiquitin proteasome and calpain activation trigger muscle proteolysis and atrophy while protein synthesis is impaired. Myosin loss is more pronounced than actin loss in CIM. Protein quality control is altered by inadequate autophagy. Ca(2+) dysregulation is present through altered Ca(2+) homeostasis. We highlight clinical hallmarks, trigger factors, and potential mechanisms from human studies and animal models that allow separation of risk factors that may trigger distinct mechanisms contributing to weakness. During critical illness, altered inflammatory (cytokines) and metabolic pathways deteriorate muscle function. ICUAW prevention/treatment is limited, e.g., tight glycemic control, delaying nutrition, and early mobilization. Future challenges include identification of primary/secondary events during the time course of critical illness, the interplay between membrane excitability, bioenergetic failure and differential proteolysis, and finding new therapeutic targets by help of tailored animal models.
Collapse
Affiliation(s)
- O Friedrich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| | - M B Reid
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| | - G Van den Berghe
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| | - I Vanhorebeek
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| | - G Hermans
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| | - M M Rich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| | - L Larsson
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|