1
|
Giovarelli M, Mocciaro E, Carnovale C, Cervia D, Perrotta C, Clementi E. Immunosenescence in skeletal muscle: The role-play in cancer cachexia chessboard. Semin Cancer Biol 2025; 111:48-59. [PMID: 40020976 DOI: 10.1016/j.semcancer.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/03/2025]
Abstract
With the increase in life expectancy, age-related conditions and diseases have become a widespread and relevant social burden. Among these, immunosenescence and cancer cachexia play a significant often intertwined role. Immunosenescence is the progressive aging decline of both the innate and adaptive immune systems leading to increased infection susceptibility, poor vaccination efficacy, autoimmune disease, and malignancies. Cancer cachexia affects elderly patients with cancer causing severe weight loss, muscle wasting, inflammation, and reduced response to therapies. Whereas the connections between immunosenescence and cancer cachexia have been raising attention, the molecular mechanisms still need to be completely elucidated. This review aims at providing the current knowledge about the interplay between immunosenescence, skeletal muscle, and cancer cachexia, analyzing the molecular pathways known so far to be involved. Finally, we highlight potential therapeutic strategies suited for elderly population aimed to block immunosenescence and to preserve muscle mass in cachexia, also presenting the analysis of the current state-of-the-art of related clinical trials.
Collapse
Affiliation(s)
- Matteo Giovarelli
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy.
| | - Emanuele Mocciaro
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy
| | - Carla Carnovale
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Viterbo 01100, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy.
| |
Collapse
|
2
|
Bartlett ST, Santi I, Hachaj G, Wilund KR, Tzvetanov I, Spaggiari M, Almario J, Di Cocco P, Bianco F, Hajjiri Z, Gallon L, Rana A, Kumar S, Benedetti E, Motl RW. Resistance-Based Muscle Therapy, Frailty, and Muscle Biopsy Findings in Kidney Transplant Candidates: A Clinical Trial. Kidney Med 2025; 7:100978. [PMID: 40201398 PMCID: PMC11978330 DOI: 10.1016/j.xkme.2025.100978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025] Open
Abstract
Rationale & Objective Frailty is associated with increased morbidity and mortality in kidney transplant recipients. We hypothesized that frailty may be attributable to diminished muscle function associated with muscle morphologic changes. This trial in kidney transplant candidates tested the reversibility of frailty by specifically targeting the affected major muscle groups. Study Design Randomized clinical trial. Setting & Participants Kidney transplant candidates. Exposure Supervised, resistance-based muscle therapy program delivered for 1 hour, 2 times per week for 1 year. Outcomes Baseline, 6-month, and 12-month Short Physical Performance Battery, gait speed, grip strength, sit-to-stand in 30 s, 36-item Short Form Survey, Patient-Reported Outcomes Measurement Information System-29, and muscle biopsy light and electron microscopy and immunohistochemistry. Analytic Approach Paired 2-tailed t test, 1-way repeated measures analysis of variance. Results Twenty-nine participants (mean age, 55 years; female, 55%; African American, 65%) were analyzed: 23 intervention and 6 control. Exercise intervention participants had significant improvements in Short Physical Performance Battery, baseline 5.2 (95% CI, 3.6-6.7) versus 6 months, 6.9 (95% CI, 5.2-8.5; P < 0.001) and 12 months, 7.2 (95% CI, 5.6-8.8; P < 0.001); baseline hand grip, 14.3 kg (95% CI, 10.3-18.4) versus 6 months, 16.9 kg (95% CI, 13.1-20.8; P < 0.05) and 12 months, 17.4 kg (95% CI, 13.9-21.0; P < 0.05); and baseline sit-to-stand in 30 s, 8.0 (95% CI, 3.8-12.2) versus 6 months, 12.7 (95% CI, 8.2-17.1; P < 0.001) and 12 months, 16.2 (95% CI, 10.7-21.7; P < 0.001). The exercise group 12-month muscle fiber diameter increased by 18.6 μm (95% CI, 8.4-28.5; P = 0.003). Expression of immunohistology markers of muscle atrophy decreased significantly. The mean difference in immunohistology score of mitochondrial oxidative function improved for cytochrome c oxidase complex IV, 1.00 (95% CI, 0.71-1.29; P < 0.001) and ATP5I increased by 0.74 (95% CI, 0.49-0.99; P < 0.001). Increased mitochondrial count did not achieve statistical significance (P = 0.096). Controls showed no improvement in either physical performance or histology. Limitations Significant under-enrollment in the control group required a paired t test analysis of experimental participants. Conclusions One year of muscle rehabilitation therapy resulted in significant improvements in physical performance metrics accompanied by significant improvements in muscle morphology.
Collapse
Affiliation(s)
| | | | | | - Kenneth R. Wilund
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, Arizona
| | | | | | | | | | | | | | | | - Ajay Rana
- Department of Surgery, College of Medicine
| | | | | | - Robert W. Motl
- Department of Kinesiology and Nutrition, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
3
|
Faiad J, Andrade MF, de Castro G, de Resende J, Coêlho M, Aquino G, Seelaender M. Muscle loss in cancer cachexia: what is the basis for nutritional support? Front Pharmacol 2025; 16:1519278. [PMID: 40078277 PMCID: PMC11897308 DOI: 10.3389/fphar.2025.1519278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/27/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer cachexia (CC) is characterized by significant skeletal muscle wasting, and contributes to diminished quality of life, while being associated with poorer response to treatment and with reduced survival. Chronic inflammation plays a central role in driving CC progression, within a complex interplay favoring catabolism. Although cachexia cannot be fully reversed by conventional nutritional support, nutritional intervention shows promise for the prevention and treatment of the syndrome. Of special interest are nutrients with antioxidant and anti-inflammatory potential and those that activate pathways involved in muscle mass synthesis and/or in the inhibition of muscle wasting. Extensive research has been carried out on novel nutritional supplements' power to mitigate CC impact, while the mechanisms through which some nutrients or bioactive compounds exert beneficial effects on muscle mass are still not totally clear. Here, we discuss the most studied supplements and nutritional strategies for dealing with muscle loss in CC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marilia Seelaender
- Cancer Metabolism Research Group, Faculdade de Medicina da Universidade de São Paulo, Departamento de Cirurgia, LIM 26-HC-USP, São Paulo, Brazil
| |
Collapse
|
4
|
Wang T, Zhou D, Hong Z. Sarcopenia and cachexia: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2025; 6:e70030. [PMID: 39764565 PMCID: PMC11702502 DOI: 10.1002/mco2.70030] [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: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 03/17/2025] Open
Abstract
Sarcopenia is defined as a muscle-wasting syndrome that occurs with accelerated aging, while cachexia is a severe wasting syndrome associated with conditions such as cancer and immunodeficiency disorders, which cannot be fully addressed through conventional nutritional supplementation. Sarcopenia can be considered a component of cachexia, with the bidirectional interplay between adipose tissue and skeletal muscle potentially serving as a molecular mechanism for both conditions. However, the underlying mechanisms differ. Recognizing the interplay and distinctions between these disorders is essential for advancing both basic and translational research in this area, enhancing diagnostic accuracy and ultimately achieving effective therapeutic solutions for affected patients. This review discusses the muscle microenvironment's changes contributing to these conditions, recent therapeutic approaches like lifestyle modifications, small molecules, and nutritional interventions, and emerging strategies such as gene editing, stem cell therapy, and gut microbiome modulation. We also address the challenges and opportunities of multimodal interventions, aiming to provide insights into the pathogenesis and molecular mechanisms of sarcopenia and cachexia, ultimately aiding in innovative strategy development and improved treatments.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Dong Zhou
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Zhen Hong
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| |
Collapse
|
5
|
Pranzini E, Muccillo L, Nesi I, Santi A, Mancini C, Lori G, Genovese M, Lottini T, Comito G, Caselli A, Arcangeli A, Sabatino L, Colantuoni V, Taddei ML, Cirri P, Paoli P. Limiting serine availability during tumor progression promotes muscle wasting in cancer cachexia. Cell Death Discov 2024; 10:510. [PMID: 39706853 DOI: 10.1038/s41420-024-02271-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/06/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024] Open
Abstract
Cancer cachexia is a multifactorial syndrome characterized by a progressive loss of body weight occurring in about 80% of cancer patients, frequently representing the leading cause of death. Dietary intervention is emerging as a promising therapeutic strategy to counteract cancer-induced wasting. Serine is the second most-consumed amino acid (AA) by cancer cells and has emerged to be strictly necessary to preserve skeletal muscle structure and functionality. Here, we demonstrate that decreased serine availability during tumor progression promotes myotubes diameter reduction in vitro and induces muscle wasting in in vivo mice models. By investigating the metabolic crosstalk between colorectal cancer cells and muscle cells, we found that incubating myotubes with conditioned media from tumor cells relying on exogenous serine consumption triggers pronounced myotubes diameter reduction. Accordingly, culturing myotubes in a serine-free medium induces fibers width reduction and suppresses the activation of the AKT-mTORC1 pathway with consequent impairment in protein synthesis, increased protein degradation, and enhanced expression of the muscle atrophy-related genes Atrogin1 and MuRF1. In addition, serine-starved conditions affect myoblast differentiation and mitochondrial oxidative metabolism, finally inducing oxidative stress in myotubes. Consistently, serine dietary deprivation strongly strengthens cancer-associated weight loss and muscle atrophy in mice models. These findings uncover serine consumption by tumor cells as a previously undisclosed driver in cancer cachexia, opening new routes for possible therapeutic approaches.
Collapse
Affiliation(s)
- Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Livio Muccillo
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
- AREA Science Park, Padriciano, 99, Trieste, Italy
| | - Ilaria Nesi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Alice Santi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Caterina Mancini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Giulia Lori
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Massimo Genovese
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Tiziano Lottini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giuseppina Comito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Anna Caselli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lina Sabatino
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Vittorio Colantuoni
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Paolo Cirri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.
| |
Collapse
|
6
|
Fornelli C, Beltrà M, Zorzano A, Costelli P, Sebastian D, Penna F. BNIP3 Downregulation Ameliorates Muscle Atrophy in Cancer Cachexia. Cancers (Basel) 2024; 16:4133. [PMID: 39766033 PMCID: PMC11674865 DOI: 10.3390/cancers16244133] [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: 11/08/2024] [Revised: 12/06/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND AND AIMS Cancer cachexia is a complex syndrome affecting most cancer patients and is directly responsible for about 20% of cancer-related deaths. Previous studies showed muscle proteolysis hyper-activation and mitophagy induction in tumor-bearing animals. While basal mitophagy is required for maintaining muscle mass and quality, excessive mitophagy promotes uncontrolled protein degradation, muscle loss and impaired function. BNIP3, a key mitophagy-related protein, is significantly increased in the muscles of both mice and human cancer hosts. This study aimed to define the potential of mitigating mitophagy via BNIP3 downregulation in preserving mitochondrial integrity, counteracting skeletal muscle loss in experimental cancer cachexia. METHODS Two in vivo gene delivery methods were performed to knock down muscle BNIP3: electroporation of a BNIP3-specific shRNA expression vector or adenovirus injection. RESULTS The electroporation effectively reduced muscle BNIP3 in healthy mice but was ineffective in C26 tumor-bearing mice. In contrast, adenovirus-mediated BNIP3 knockdown successfully decreased BNIP3 levels also in tumor hosts. Although BNIP3 knockdown did not impact overall on body or muscle mass, it improved muscle fiber size in C26-bearing miceh2, suggesting partial prevention of muscle atrophy. Mitochondrial respiratory chain complexes (OxPhos) and TOM20 protein levels were consistently rescued, indicating improvements in mitochondrial mass, while H2O2 levels were unchanged among the groups, suggesting that BNIP3 downregulation does not impair the endogenous control of oxidative balance. CONCLUSIONS These findings suggest that a fine balance between mitochondrial disposal and biogenesis is fundamental for preserving muscle homeostasis and highlight a potential role for BNIP3 modulation against cancer-induced muscle wasting.
Collapse
Affiliation(s)
- Claudia Fornelli
- Department of Clinical and Biological Sciences, University of Torino, 10043 Turin, Italy; (C.F.); (P.C.)
| | - Marc Beltrà
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (M.B.); (A.Z.)
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (M.B.); (A.Z.)
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Torino, 10043 Turin, Italy; (C.F.); (P.C.)
| | - David Sebastian
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain;
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Torino, 10043 Turin, Italy; (C.F.); (P.C.)
| |
Collapse
|
7
|
Berriel Diaz M, Rohm M, Herzig S. Cancer cachexia: multilevel metabolic dysfunction. Nat Metab 2024; 6:2222-2245. [PMID: 39578650 DOI: 10.1038/s42255-024-01167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024]
Abstract
Cancer cachexia is a complex metabolic disorder marked by unintentional body weight loss or 'wasting' of body mass, driven by multiple aetiological factors operating at various levels. It is associated with many malignancies and significantly contributes to cancer-related morbidity and mortality. With emerging recognition of cancer as a systemic disease, there is increasing awareness that understanding and treatment of cancer cachexia may represent a crucial cornerstone for improved management of cancer. Here, we describe the metabolic changes contributing to body wasting in cachexia and explain how the entangled action of both tumour-derived and host-amplified processes induces these metabolic changes. We discuss energy homeostasis and possible ways that the presence of a tumour interferes with or hijacks physiological energy conservation pathways. In that context, we highlight the role played by metabolic cross-talk mechanisms in cachexia pathogenesis. Lastly, we elaborate on the challenges and opportunities in the treatment of this devastating paraneoplastic phenomenon that arise from the complex and multifaceted metabolic cross-talk mechanisms and provide a status on current and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Chair Molecular Metabolic Control, Technical University of Munich, Munich, Germany.
| |
Collapse
|
8
|
Mandic M, Paunovic V, Vucicevic L, Kosic M, Mijatovic S, Trajkovic V, Harhaji-Trajkovic L. No energy, no autophagy-Mechanisms and therapeutic implications of autophagic response energy requirements. J Cell Physiol 2024; 239:e31366. [PMID: 38958520 DOI: 10.1002/jcp.31366] [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: 02/01/2024] [Revised: 05/29/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024]
Abstract
Autophagy is a lysosome-mediated self-degradation process of central importance for cellular quality control. It also provides macromolecule building blocks and substrates for energy metabolism during nutrient or energy deficiency, which are the main stimuli for autophagy induction. However, like most biological processes, autophagy itself requires ATP, and there is an energy threshold for its initiation and execution. We here present the first comprehensive review of this often-overlooked aspect of autophagy research. The studies in which ATP deficiency suppressed autophagy in vitro and in vivo were classified according to the energy pathway involved (oxidative phosphorylation or glycolysis). A mechanistic insight was provided by pinpointing the critical ATP-consuming autophagic events, including transcription/translation/interaction of autophagy-related molecules, autophagosome formation/elongation, autophagosome fusion with the lysosome, and lysosome acidification. The significance of energy-dependent fine-tuning of autophagic response for preserving the cell homeostasis, and potential implications for the therapy of cancer, autoimmunity, metabolic disorders, and neurodegeneration are discussed.
Collapse
Affiliation(s)
- Milos Mandic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Verica Paunovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ljubica Vucicevic
- Department of Neurophysiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milica Kosic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Srdjan Mijatovic
- Clinic for Emergency Surgery, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Vladimir Trajkovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ljubica Harhaji-Trajkovic
- Department of Neurophysiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
9
|
Molfino A, Ambrosani F, Udali S, Imbimbo G, Moruzzi S, Castagna A, Pattini P, Tambaro F, Ramaccini C, Muscaritoli M, Friso S. DNA Methylation Signatures Characterize Gene Expression Modulation in Lung Cancer Patients Affected by Anorexia. Nutrients 2024; 16:3721. [PMID: 39519555 PMCID: PMC11547925 DOI: 10.3390/nu16213721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES The pathophysiology of cancer anorexia is multifactorial and unclear. Transcriptomic analysis from PBMCs RNA showed diverse patterns of gene expression pathways in anorexic cancer patients. We assessed whether the different transcriptomic signatures are modulated by DNA methylation in lung cancer patients presenting with poor appetite. METHODS Lung cancer patients and controls were enrolled, and anorexia was assessed by the FAACT-score questionnaire. Genome-wide DNA methylation was determined by Human Infinium MethylationEPIC BeadChip Kit. Data from genome-wide methylation analysis were merged with those from gene expression analysis, previously obtained by RNA sequencing (NGS). Four groups of genes were identified for each comparison: hypermethylated repressed, hypermethylated induced, hypomethylated repressed, and hypomethylated induced. RESULTS Cancer patients (n = 16) showed 382 differentially methylated genes when compared with controls (n = 8). Anorexic patients (n = 8) presented 586 hypomethylated and 174 hypermethylated genes compared with controls. In anorexic patients vs. non-anorexic (n = 8), 211 genes were identified as hypomethylated and 90 hypermethylated. When microarray methylation data were merged with transcriptomic data by RNA sequencing, we observed significant differences in anorexic patients vs. controls; a total of 42 genes resulted as hypomethylated and induced, 5 hypermethylated repressed, 10 hypermethylated induced, and 15 hypomethylated repressed. The CG sites analyzed by targeted bisulfite NGS in four genes of interest (FLNA, PGRMC1, GNL3L, and FHL1) resulting as hypomethylated in anorexic vs. controls allowed the validation of the data obtained from DNA methylation. Interestingly, the four genes resulted as hypomethylated in anorexic patients vs. non-anorexic patients and vs. controls (p < 0.0001). CONCLUSIONS Our data support that methylation is implicated in cancer-associated anorexia and nutritional derangements among lung cancer patients.
Collapse
Affiliation(s)
- Alessio Molfino
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (G.I.); (F.T.); (C.R.); (M.M.)
| | - Francesca Ambrosani
- Unit of Internal Medicine B, Department of Medicine, School of Medicine, University of Verona, 37129 Verona, Italy; (F.A.); (S.U.); (S.M.); (A.C.); (P.P.)
| | - Silvia Udali
- Unit of Internal Medicine B, Department of Medicine, School of Medicine, University of Verona, 37129 Verona, Italy; (F.A.); (S.U.); (S.M.); (A.C.); (P.P.)
| | - Giovanni Imbimbo
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (G.I.); (F.T.); (C.R.); (M.M.)
| | - Sara Moruzzi
- Unit of Internal Medicine B, Department of Medicine, School of Medicine, University of Verona, 37129 Verona, Italy; (F.A.); (S.U.); (S.M.); (A.C.); (P.P.)
| | - Annalisa Castagna
- Unit of Internal Medicine B, Department of Medicine, School of Medicine, University of Verona, 37129 Verona, Italy; (F.A.); (S.U.); (S.M.); (A.C.); (P.P.)
| | - Patrizia Pattini
- Unit of Internal Medicine B, Department of Medicine, School of Medicine, University of Verona, 37129 Verona, Italy; (F.A.); (S.U.); (S.M.); (A.C.); (P.P.)
| | - Federica Tambaro
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (G.I.); (F.T.); (C.R.); (M.M.)
| | - Cesarina Ramaccini
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (G.I.); (F.T.); (C.R.); (M.M.)
| | - Maurizio Muscaritoli
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (G.I.); (F.T.); (C.R.); (M.M.)
| | - Simonetta Friso
- Unit of Internal Medicine B, Department of Medicine, School of Medicine, University of Verona, 37129 Verona, Italy; (F.A.); (S.U.); (S.M.); (A.C.); (P.P.)
| |
Collapse
|
10
|
Rahman FA, Baechler BL, Quadrilatero J. Key considerations for investigating and interpreting autophagy in skeletal muscle. Autophagy 2024; 20:2121-2132. [PMID: 39007805 PMCID: PMC11423691 DOI: 10.1080/15548627.2024.2373676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Skeletal muscle plays a crucial role in generating force to facilitate movement. Skeletal muscle is a heterogenous tissue composed of diverse fibers with distinct contractile and metabolic profiles. The intricate classification of skeletal muscle fibers exists on a continuum ranging from type I (slow-twitch, oxidative) to type II (fast-twitch, glycolytic). The heterogenous distribution and characteristics of fibers within and between skeletal muscles profoundly influences cellular signaling; however, this has not been broadly discussed as it relates to macroautophagy/autophagy. The growing interest in skeletal muscle autophagy research underscores the necessity of comprehending the interplay between autophagic responses among skeletal muscles and fibers with different contractile properties, metabolic profiles, and other related signaling processes. We recommend approaching the interpretation of autophagy findings with careful consideration for two key reasons: 1) the distinct behaviors and responses of different skeletal muscles or fibers to various perturbations, and 2) the potential impact of alterations in skeletal muscle fiber type or metabolic profile on observed autophagic outcomes. This review provides an overview of the autophagic profile and response in skeletal muscles/fibers of different types and metabolic profiles. Further, this review discusses autophagic findings in various conditions and diseases that may differentially affect skeletal muscle. Finally, we provide key points of consideration to better enable researchers to fine-tune the design and interpretation of skeletal muscle autophagy experiments.Abbreviation: AKT1: AKT serine/threonine kinase 1; AMPK: AMP-activated protein kinase; ATG: autophagy related; ATG4: autophagy related 4 cysteine peptidase; ATG5: autophagy related 5; ATG7: autophagy related 7; ATG12: autophagy related 12; BECN1: beclin 1; BNIP3: BCL2 interacting protein 3; CKD: chronic kidney disease; COPD: chronic obstructive pulmonary disease; CS: citrate synthase; DIA: diaphragm; EDL: extensor digitorum longus; FOXO3/FOXO3A: forkhead box O3; GAS; gastrocnemius; GP: gastrocnemius-plantaris complex; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MAPK: mitogen-activated protein kinase; MYH: myosin heavy chain; PINK1: PTEN induced kinase 1; PLANT: plantaris; PRKN: parkin RBR E3 ubiquitin protein ligase; QUAD: quadriceps; RA: rectus abdominis; RG: red gastrocnemius; RQ: red quadriceps; SOL: soleus; SQSTM1: sequestosome 1; TA: tibialis anterior; WG: white gastrocnemius; WQ: white quadriceps; WVL: white vastus lateralis; VL: vastus lateralis; ULK1: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Fasih A. Rahman
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Brittany L. Baechler
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Joe Quadrilatero
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
11
|
Geppert J, Rohm M. Cancer cachexia: biomarkers and the influence of age. Mol Oncol 2024; 18:2070-2086. [PMID: 38414161 PMCID: PMC11467804 DOI: 10.1002/1878-0261.13590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 12/01/2023] [Accepted: 01/15/2024] [Indexed: 02/29/2024] Open
Abstract
Cancer cachexia (Ccx) is a complex metabolic condition characterized by pronounced muscle and fat wasting, systemic inflammation, weakness and fatigue. Up to 30% of cancer patients succumb directly to Ccx, yet therapies that effectively address this perturbed metabolic state are rare. In recent decades, several characteristics of Ccx have been established in mice and humans, of which we here highlight adipose tissue dysfunction, muscle wasting and systemic inflammation, as they are directly linked to biomarker discovery. To counteract cachexia pathogenesis as early as possible and mitigate its detrimental impact on anti-cancer treatments, identification and validation of clinically endorsed biomarkers assume paramount importance. Ageing was recently shown to affect both the validity of Ccx biomarkers and Ccx development, but the underlying mechanisms are still unknown. Thus, unravelling the intricate interplay between ageing and Ccx can help to counteract Ccx pathogenesis and tailor diagnostic and treatment strategies to individual needs.
Collapse
Affiliation(s)
- Julia Geppert
- Institute for Diabetes and CancerHelmholtz MunichNeuherbergGermany
- Joint Heidelberg‐IDC Translational Diabetes Program, Inner Medicine 1Heidelberg University HospitalGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| | - Maria Rohm
- Institute for Diabetes and CancerHelmholtz MunichNeuherbergGermany
- Joint Heidelberg‐IDC Translational Diabetes Program, Inner Medicine 1Heidelberg University HospitalGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| |
Collapse
|
12
|
Ryan PJ, Uranga S, Stanelle ST, Lewis MH, O'Reilly CL, Cardin JM, Deaver JW, Morton AB, Fluckey JD. The autophagy inhibitor NSC185058 suppresses mTORC1-mediated protein anabolism in cultured skeletal muscle. Sci Rep 2024; 14:8094. [PMID: 38582781 PMCID: PMC10998866 DOI: 10.1038/s41598-024-58716-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/02/2024] [Indexed: 04/08/2024] Open
Abstract
The mammalian target of rapamycin (mTOR), and specifically the mTOR complex 1 (mTORC1) is the central regulator of anabolism in skeletal muscle. Among the many functions of this kinase complex is the inhibition of the catabolic process of autophagy; however, less work has been done in investigating the role of autophagy in regulating mTORC1 signaling. Using an in vitro model to better understand the pathways involved, we activated mTORC1 by several different means (growth factors, leucine supplementation, or muscle contraction), alone or with the autophagy inhibitor NSC185058. We found that inhibiting autophagy with NSC185058 suppresses mTORC1 activity, preventing any increase in cellular protein anabolism. These decrements were the direct result of action on the mTORC1 kinase, which we demonstrate, for the first time, cannot function when autophagy is inhibited by NSC185058. Our results indicate that, far from being a matter of unidirectional action, the relationship between mTORC1 and the autophagic cascade is more nuanced, with autophagy serving as an mTORC1 input, and mTORC1 inhibition of autophagy as a form of homeostatic feedback to regulate anabolic signaling. Future studies of cellular metabolism will have to consider this fundamental intertwining of protein anabolism and catabolism, and how it ultimately serves to regulate muscle proteostasis.
Collapse
Affiliation(s)
- Patrick J Ryan
- Muscle Biology Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, Gilchrist Building, 2929 Research Parkway, College Station, TX, 77843-4243, USA
| | - Selina Uranga
- Muscle Biology Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, Gilchrist Building, 2929 Research Parkway, College Station, TX, 77843-4243, USA
| | - Sean T Stanelle
- Muscle Biology Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, Gilchrist Building, 2929 Research Parkway, College Station, TX, 77843-4243, USA
| | - Megan H Lewis
- Muscle Biology Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, Gilchrist Building, 2929 Research Parkway, College Station, TX, 77843-4243, USA
| | - Colleen L O'Reilly
- Muscle Biology Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, Gilchrist Building, 2929 Research Parkway, College Station, TX, 77843-4243, USA
| | - Jessica M Cardin
- Muscle Biology Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, Gilchrist Building, 2929 Research Parkway, College Station, TX, 77843-4243, USA
| | - J William Deaver
- Muscle Biology Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, Gilchrist Building, 2929 Research Parkway, College Station, TX, 77843-4243, USA
| | - Aaron B Morton
- Soft Tissue Regeneration and Applied Biomaterials Laboratory, Texas A&M University, Gilchrist Building, 2929 Research Parkway, College Station, TX, 77843-4243, USA
| | - James D Fluckey
- Muscle Biology Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, Gilchrist Building, 2929 Research Parkway, College Station, TX, 77843-4243, USA.
| |
Collapse
|
13
|
Dave S, Patel BM. Deliberation on debilitating condition of cancer cachexia: Skeletal muscle wasting. Fundam Clin Pharmacol 2023; 37:1079-1091. [PMID: 37474262 DOI: 10.1111/fcp.12931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/18/2023] [Accepted: 06/08/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Cancer cachexia is a debilitating syndrome associated with marked body loss because of muscular atrophy and fat loss. There are several mechanisms contributing to the pathogenesis of cachexia. The presence of the tumor releases cytokines from inflammatory and immune cells, which play a significant role in activating and deactivating certain pathways associated with protein, carbohydrate, and lipid metabolism. This review focuses on various cascades involving an imbalance between protein synthesis and degradation in the skeletal muscles. OBJECTIVES This study aimed to elucidate the mechanisms involved in skeletal muscle wasting phenomenon over the last few years. METHODS This article briefly overviews different pathways responsible for muscle atrophy in cancer cachexia. Studies published up to April 2023 were included. Important findings and study contributions were chosen and compiled using several databases including PubMed, Google Scholar, Science Direct, and ClinicalTrials.gov using relevant keywords. RESULTS Cancer cachexia is a complex disease involving multiple factors resulting in atrophy of skeletal muscles. Systemic inflammation, altered energy balance and carbohydrate metabolism, altered lipid and protein metabolism, and adipose tissue browning are some of the major culprits in cancer cachexia. Increased protein degradation and decreased protein synthesis lead to muscle atrophy. Changes in signaling pathway like ubiquitin-proteasome, autophagy, mTOR, AMPK, and IGF-1 also lead to muscle wasting. Physical exercise, nutritional supplementation, steroids, myostatin inhibitors, and interventions targeting on inflammation have been investigated to treat cancer cachexia. Some therapy showed positive results in preclinical and clinical settings, although more research on the efficacy and safety of the treatment should be done. CONCLUSION Muscle atrophy in cancer cachexia is the result of multiple complex mechanisms; as a result, a lot more research has been done to describe the pathophysiology of the disease. Targeted therapy and multimodal interventions can improve clinical outcomes for patients.
Collapse
Affiliation(s)
- Srusti Dave
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Bhoomika M Patel
- School of Medico-legal Studies, National Forensic Sciences University, Gandhinagar, India
| |
Collapse
|
14
|
Conti F, Di Martino S, Drago F, Bucolo C, Micale V, Montano V, Siciliano G, Mancuso M, Lopriore P. Red Flags in Primary Mitochondrial Diseases: What Should We Recognize? Int J Mol Sci 2023; 24:16746. [PMID: 38069070 PMCID: PMC10706469 DOI: 10.3390/ijms242316746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Primary mitochondrial diseases (PMDs) are complex group of metabolic disorders caused by genetically determined impairment of the mitochondrial oxidative phosphorylation (OXPHOS). The unique features of mitochondrial genetics and the pivotal role of mitochondria in cell biology explain the phenotypical heterogeneity of primary mitochondrial diseases and the resulting diagnostic challenges that follow. Some peculiar features ("red flags") may indicate a primary mitochondrial disease, helping the physician to orient in this diagnostic maze. In this narrative review, we aimed to outline the features of the most common mitochondrial red flags offering a general overview on the topic that could help physicians to untangle mitochondrial medicine complexity.
Collapse
Affiliation(s)
- Federica Conti
- Department of Biomedical and Biotechnological Science, School of Medicine, University of Catania, 95123 Catania, Italy; (F.C.); (S.D.M.); (C.B.); (V.M.)
| | - Serena Di Martino
- Department of Biomedical and Biotechnological Science, School of Medicine, University of Catania, 95123 Catania, Italy; (F.C.); (S.D.M.); (C.B.); (V.M.)
| | - Filippo Drago
- Department of Biomedical and Biotechnological Science, School of Medicine, University of Catania, 95123 Catania, Italy; (F.C.); (S.D.M.); (C.B.); (V.M.)
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Science, School of Medicine, University of Catania, 95123 Catania, Italy; (F.C.); (S.D.M.); (C.B.); (V.M.)
- Center for Research in Ocular Pharmacology-CERFO, University of Catania, 95213 Catania, Italy
| | - Vincenzo Micale
- Department of Biomedical and Biotechnological Science, School of Medicine, University of Catania, 95123 Catania, Italy; (F.C.); (S.D.M.); (C.B.); (V.M.)
| | - Vincenzo Montano
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy (P.L.)
| | - Gabriele Siciliano
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy (P.L.)
| | - Michelangelo Mancuso
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy (P.L.)
| | - Piervito Lopriore
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy (P.L.)
| |
Collapse
|
15
|
Kubat GB, Bouhamida E, Ulger O, Turkel I, Pedriali G, Ramaccini D, Ekinci O, Ozerklig B, Atalay O, Patergnani S, Nur Sahin B, Morciano G, Tuncer M, Tremoli E, Pinton P. Mitochondrial dysfunction and skeletal muscle atrophy: Causes, mechanisms, and treatment strategies. Mitochondrion 2023; 72:33-58. [PMID: 37451353 DOI: 10.1016/j.mito.2023.07.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Skeletal muscle, which accounts for approximately 40% of total body weight, is one of the most dynamic and plastic tissues in the human body and plays a vital role in movement, posture and force production. More than just a component of the locomotor system, skeletal muscle functions as an endocrine organ capable of producing and secreting hundreds of bioactive molecules. Therefore, maintaining healthy skeletal muscles is crucial for supporting overall body health. Various pathological conditions, such as prolonged immobilization, cachexia, aging, drug-induced toxicity, and cardiovascular diseases (CVDs), can disrupt the balance between muscle protein synthesis and degradation, leading to skeletal muscle atrophy. Mitochondrial dysfunction is a major contributing mechanism to skeletal muscle atrophy, as it plays crucial roles in various biological processes, including energy production, metabolic flexibility, maintenance of redox homeostasis, and regulation of apoptosis. In this review, we critically examine recent knowledge regarding the causes of muscle atrophy (disuse, cachexia, aging, etc.) and its contribution to CVDs. Additionally, we highlight the mitochondrial signaling pathways involvement to skeletal muscle atrophy, such as the ubiquitin-proteasome system, autophagy and mitophagy, mitochondrial fission-fusion, and mitochondrial biogenesis. Furthermore, we discuss current strategies, including exercise, mitochondria-targeted antioxidants, in vivo transfection of PGC-1α, and the potential use of mitochondrial transplantation as a possible therapeutic approach.
Collapse
Affiliation(s)
- Gokhan Burcin Kubat
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey.
| | - Esmaa Bouhamida
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Gaia Pedriali
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Daniela Ramaccini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Ozgur Ekinci
- Department of Pathology, Gazi University, 06500 Ankara, Turkey
| | - Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Ozbeyen Atalay
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Simone Patergnani
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Beyza Nur Sahin
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Giampaolo Morciano
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Elena Tremoli
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Paolo Pinton
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
16
|
F AR, Quadrilatero J. Emerging role of mitophagy in myoblast differentiation and skeletal muscle remodeling. Semin Cell Dev Biol 2023; 143:54-65. [PMID: 34924331 DOI: 10.1016/j.semcdb.2021.11.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 12/17/2022]
Abstract
Mitochondrial turnover in the form of mitophagy is emerging as a central process in maintaining cellular function. The degradation of damaged mitochondria through mitophagy is particularly important in cells/tissues that exhibit high energy demands. Skeletal muscle is one such tissue that requires precise turnover of mitochondria in several conditions in order to optimize energy production and prevent bioenergetic crisis. For instance, the formation of skeletal muscle (i.e., myogenesis) is accompanied by robust turnover of low-functioning mitochondria to eventually allow the formation of high-functioning mitochondria. In mature skeletal muscle, alterations in mitophagy-related signaling occur during exercise, aging, and various disease states. Nonetheless, several questions regarding the direct role of mitophagy in various skeletal muscle conditions remain unknown. Furthermore, given the heterogenous nature of skeletal muscle with respect to various cellular and molecular properties, and the plasticity in these properties in various conditions, the involvement and characterization of mitophagy requires more careful consideration in this tissue. Therefore, this review will highlight the known mechanisms of mitophagy in skeletal muscle, and discuss their involvement during myogenesis and various skeletal muscle conditions. This review also provides important considerations for the accurate measurement of mitophagy and interpretation of data in skeletal muscle.
Collapse
Affiliation(s)
- Ahmad Rahman F
- Department of Kinesiology & Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Joe Quadrilatero
- Department of Kinesiology & Health Sciences, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
17
|
Zhang Z, Tan S, Li S, Cheng Y, Wang J, Liu H, Yan M, Wu G. Mitophagy-mediated inflammation and oxidative stress contribute to muscle wasting in cancer cachexia. J Clin Biochem Nutr 2023; 73:34-42. [PMID: 37534096 PMCID: PMC10390805 DOI: 10.3164/jcbn.23-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/12/2023] [Indexed: 08/04/2023] Open
Abstract
Cancer cachexia is commonly seen in patients with malignant tumors, which usually leads to poor life quality and negatively affects long-term prognosis and survival. Mitochondria dysfunction and enhanced autophagy are well-established to play an important role in skeletal muscle wasting. However, whether mitophagy is engaged in the pathogenesis of cancer cachexia requires further investigation. This study comprised a clinical study and animal experimentation. Clinical data such as CT images and laboratory results were obtained and analyzed. Then mice model of cancer cachexia and mitophagy inhibition were established. Data including skeletal muscle mass and function, mitochondria structure and function, inflammatory factors as well as ROS concentration. Mitophagy was enhanced in cancer cachexia patients with increased inflammatory factors. Greater disruption of skeletal muscle fiber and mitochondria structure were seen in cancer cachexia, with a higher level of inflammatory factors and ROS expression in skeletal muscle. Meanwhile, ATP production was undermined, indicating a close relationship with mitophagy, inflammation, and oxidative stress in the skeletal muscle of cancer cachexia mice models. In conclusion, mitophagy is activated in cancer cachexia and may play a role in skeletal muscle atrophy, and inflammation and oxidative stress might participate in mitophagy-related skeletal muscle injury.
Collapse
Affiliation(s)
- Zhige Zhang
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Shanjun Tan
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Shuhao Li
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Yuxi Cheng
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Junjie Wang
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Hao Liu
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Mingyue Yan
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Guohao Wu
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| |
Collapse
|
18
|
Martin A, Gallot YS, Freyssenet D. Molecular mechanisms of cancer cachexia-related loss of skeletal muscle mass: data analysis from preclinical and clinical studies. J Cachexia Sarcopenia Muscle 2023; 14:1150-1167. [PMID: 36864755 PMCID: PMC10235899 DOI: 10.1002/jcsm.13073] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 06/15/2022] [Accepted: 08/14/2022] [Indexed: 03/04/2023] Open
Abstract
Cancer cachexia is a systemic hypoanabolic and catabolic syndrome that diminishes the quality of life of cancer patients, decreases the efficiency of therapeutic strategies and ultimately contributes to decrease their lifespan. The depletion of skeletal muscle compartment, which represents the primary site of protein loss during cancer cachexia, is of very poor prognostic in cancer patients. In this review, we provide an extensive and comparative analysis of the molecular mechanisms involved in the regulation of skeletal muscle mass in human cachectic cancer patients and in animal models of cancer cachexia. We summarize data from preclinical and clinical studies investigating how the protein turnover is regulated in cachectic skeletal muscle and question to what extent the transcriptional and translational capacities, as well as the proteolytic capacity (ubiquitin-proteasome system, autophagy-lysosome system and calpains) of skeletal muscle are involved in the cachectic syndrome in human and animals. We also wonder how regulatory mechanisms such as insulin/IGF1-AKT-mTOR pathway, endoplasmic reticulum stress and unfolded protein response, oxidative stress, inflammation (cytokines and downstream IL1ß/TNFα-NF-κB and IL6-JAK-STAT3 pathways), TGF-ß signalling pathways (myostatin/activin A-SMAD2/3 and BMP-SMAD1/5/8 pathways), as well as glucocorticoid signalling, modulate skeletal muscle proteostasis in cachectic cancer patients and animals. Finally, a brief description of the effects of various therapeutic strategies in preclinical models is also provided. Differences in the molecular and biochemical responses of skeletal muscle to cancer cachexia between human and animals (protein turnover rates, regulation of ubiquitin-proteasome system and myostatin/activin A-SMAD2/3 signalling pathways) are highlighted and discussed. Identifying the various and intertwined mechanisms that are deregulated during cancer cachexia and understanding why they are decontrolled will provide therapeutic targets for the treatment of skeletal muscle wasting in cancer patients.
Collapse
Affiliation(s)
- Agnès Martin
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| | - Yann S. Gallot
- LBEPS, Univ Evry, IRBA, Université Paris SaclayEvryFrance
| | - Damien Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| |
Collapse
|
19
|
van de Worp WR, Theys J, González AS, van der Heyden B, Verhaegen F, Hauser D, Caiment F, Smeets HJ, Schols AM, van Helvoort A, Langen RC. A novel orthotopic mouse model replicates human lung cancer cachexia. J Cachexia Sarcopenia Muscle 2023; 14:1410-1423. [PMID: 37025071 PMCID: PMC10235890 DOI: 10.1002/jcsm.13222] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 02/17/2023] [Accepted: 02/28/2023] [Indexed: 04/08/2023] Open
Abstract
INTRODUCTION Cancer cachexia, highly prevalent in lung cancer, is a debilitating syndrome characterized by involuntary loss of skeletal muscle mass and is associated with poor clinical outcome, decreased survival and negative impact on tumour therapy. Various lung tumour-bearing animal models have been used to explore underlying mechanisms of cancer cachexia. However, these models do not simulate anatomical and immunological features key to lung cancer and associated muscle wasting. Overcoming these shortcomings is essential to translate experimental findings into the clinic. We therefore evaluated whether a syngeneic, orthotopic lung cancer mouse model replicates systemic and muscle-specific alterations associated with human lung cancer cachexia. METHODS Immune competent, 11 weeks old male 129S2/Sv mice, were randomly allocated to either (1) sham control group or (2) tumour-bearing group. Syngeneic lung epithelium-derived adenocarcinoma cells (K-rasG12D ; p53R172HΔG ) were inoculated intrapulmonary into the left lung lobe of the mice. Body weight and food intake were measured daily. At baseline and weekly after surgery, grip strength was measured and tumour growth and muscle volume were assessed using micro cone beam CT imaging. After reaching predefined surrogate survival endpoint, animals were euthanized, and skeletal muscles of the lower hind limbs were collected for biochemical analysis. RESULTS Two-third of the tumour-bearing mice developed cachexia based on predefined criteria. Final body weight (-13.7 ± 5.7%; P < 0.01), muscle mass (-13.8 ± 8.1%; P < 0.01) and muscle strength (-25.5 ± 10.5%; P < 0.001) were reduced in cachectic mice compared with sham controls and median survival time post-surgery was 33.5 days until humane endpoint. Markers for proteolysis, both ubiquitin proteasome system (Fbxo32 and Trim63) and autophagy-lysosomal pathway (Gabarapl1 and Bnip3), were significantly upregulated, whereas markers for protein synthesis (relative phosphorylation of Akt, S6 and 4E-BP1) were significantly decreased in the skeletal muscle of cachectic mice compared with control. The cachectic mice exhibited increased pentraxin-2 (P < 0.001) and CXCL1/KC (P < 0.01) expression levels in blood plasma and increased mRNA expression of IκBα (P < 0.05) in skeletal muscle, indicative for the presence of systemic inflammation. Strikingly, RNA sequencing, pathway enrichment and miRNA expression analyses of mouse skeletal muscle strongly mirrored alterations observed in muscle biopsies of patients with lung cancer cachexia. CONCLUSIONS We developed an orthotopic model of lung cancer cachexia in immune competent mice. Because this model simulates key aspects specific to cachexia in lung cancer patients, it is highly suitable to further investigate the underlying mechanisms of lung cancer cachexia and to test the efficacy of novel intervention strategies.
Collapse
Affiliation(s)
- Wouter R.P.H. van de Worp
- Department of Respiratory Medicine, NUTRIM – School of Nutrition and Translational Research in MetabolismMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Jan Theys
- Department of Precision Medicine, GROW – School for Oncology and Developmental BiologyMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Alba Sanz González
- Department of Respiratory Medicine, NUTRIM – School of Nutrition and Translational Research in MetabolismMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Brent van der Heyden
- Department of Radiation Oncology (MAASTRO), GROW – School for Oncology and Developmental BiologyMaastricht University Medical Centre+MaastrichtThe Netherlands
| | - Frank Verhaegen
- Department of Radiation Oncology (MAASTRO), GROW – School for Oncology and Developmental BiologyMaastricht University Medical Centre+MaastrichtThe Netherlands
| | - Duncan Hauser
- Department of Toxicogenomics, GROW – School for Oncology and Developmental Biology, MHeNs – School for Mental Health and NeurosciencesMaastricht UniversityMaastrichtThe Netherlands
| | - Florian Caiment
- Department of Toxicogenomics, GROW – School for Oncology and Developmental Biology, MHeNs – School for Mental Health and NeurosciencesMaastricht UniversityMaastrichtThe Netherlands
| | - Hubertus J.M. Smeets
- Department of Toxicogenomics, GROW – School for Oncology and Developmental Biology, MHeNs – School for Mental Health and NeurosciencesMaastricht UniversityMaastrichtThe Netherlands
| | - Annemie M.W.J. Schols
- Department of Respiratory Medicine, NUTRIM – School of Nutrition and Translational Research in MetabolismMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Ardy van Helvoort
- Department of Respiratory Medicine, NUTRIM – School of Nutrition and Translational Research in MetabolismMaastricht University Medical Center+MaastrichtThe Netherlands
- Danone Nutricia ResearchUtrechtThe Netherlands
| | - Ramon C.J. Langen
- Department of Respiratory Medicine, NUTRIM – School of Nutrition and Translational Research in MetabolismMaastricht University Medical Center+MaastrichtThe Netherlands
| |
Collapse
|
20
|
Simões E Silva AC, Oliveira EA, Cheung WW, Mak RH. Redox Signaling in Chronic Kidney Disease-Associated Cachexia. Antioxidants (Basel) 2023; 12:antiox12040945. [PMID: 37107320 PMCID: PMC10136196 DOI: 10.3390/antiox12040945] [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: 02/21/2023] [Revised: 04/14/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Redox signaling alterations contribute to chronic kidney disease (CKD)-associated cachexia. This review aims to summarize studies about redox pathophysiology in CKD-associated cachexia and muscle wasting and to discuss potential therapeutic approaches based on antioxidant and anti-inflammatory molecules to restore redox homeostasis. Enzymatic and non-enzymatic systems of antioxidant molecules have been studied in experimental models of kidney diseases and patients with CKD. Oxidative stress is increased by several factors present in CKD, including uremic toxins, inflammation, and metabolic and hormone alterations, leading to muscle wasting. Rehabilitative nutritional and physical exercises have shown beneficial effects for CKD-associated cachexia. Anti-inflammatory molecules have also been tested in experimental models of CKD. The importance of oxidative stress has been shown by experimental studies in which antioxidant therapies ameliorated CKD and its associated complications in the 5/6 nephrectomy model. Treatment of CKD-associated cachexia is a challenge and further studies are necessary to investigate potential therapies involving antioxidant therapy.
Collapse
Affiliation(s)
- Ana Cristina Simões E Silva
- Department of Pediatrics, Division of Pediatric Nephrology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte 30130-100, MG, Brazil
| | - Eduardo A Oliveira
- Department of Pediatrics, Division of Pediatric Nephrology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte 30130-100, MG, Brazil
| | - Wai W Cheung
- Department of Pediatrics, Rady Children's Hospital San Diego, University of California San Diego, La Jolla, CA 92093, USA
| | - Robert H Mak
- Department of Pediatrics, Rady Children's Hospital San Diego, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
21
|
Saroul N, Tardif N, Pereira B, Dissard A, Montrieul L, Sanchez P, Salles J, Petersen JE, Jakobson T, Gilain L, Mom T, Boirie Y, Rooyakers O, Walrand S. Conditioned Media from Head and Neck Cancer Cell Lines and Serum Samples from Head and Neck Cancer Patients Drive Catabolic Pathways in Cultured Muscle Cells. Cancers (Basel) 2023; 15:cancers15061843. [PMID: 36980729 PMCID: PMC10047086 DOI: 10.3390/cancers15061843] [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: 01/26/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND The role of secreted factors from the tumor cells in driving cancer cachexia and especially muscle loss is unknown. We wanted to study both the action of secreted factors from head and neck cancer (HNC) cell lines and circulating factors in HNC patients on skeletal muscle protein catabolism. METHODS Conditioned media (CM) made from head and neck cancer cell lines and mix of sera from head and neck cancer (HNC) patients were incubated for 48 h with human myotubes. The atrophy and the catabolic pathway were monitored in myotubes. The patients were classified regarding their skeletal muscle loss observed at the outset of management. RESULTS Tumor CM (TCM) was able to produce atrophy on myotubes as compared with control CM (CCM). However, a mix of sera from HNC patients was not able to produce atrophy in myotubes. Despite this discrepancy on atrophy, we observed a similar regulation of the catabolic pathways by the tumor-conditioned media and mix of sera from cancer patients. The catabolic response after incubation with the mix of sera seemed to depend on the muscle loss seen in patients. CONCLUSION This study found evidence that the atrophy observed in HNC patients cannot be solely explained by a deficit in food intake.
Collapse
Affiliation(s)
- Nicolas Saroul
- Otolaryngology Head and Neck Surgery Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
- Biostatistics Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
| | - Nicolas Tardif
- Anesthesiology and Intensive Care, Department of Clinical Science Intervention and Technology, CLINTEC, Karolinska Institutet, 141 86 Huddinge, Sweden
- Division of Perioperative Medicine and Intensive Care, Karolinska University Hospital, 171 77 Huddinge, Sweden
| | - Bruno Pereira
- Biostatistics Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
| | - Alexis Dissard
- Otolaryngology Head and Neck Surgery Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
| | - Laura Montrieul
- Otolaryngology Head and Neck Surgery Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
| | - Phelipe Sanchez
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
| | - Jérôme Salles
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
| | - Jens Erik Petersen
- Otolaryngology Head and Neck Surgery Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
| | - Towe Jakobson
- Anesthesiology and Intensive Care, Department of Clinical Science Intervention and Technology, CLINTEC, Karolinska Institutet, 141 86 Huddinge, Sweden
| | - Laurent Gilain
- Otolaryngology Head and Neck Surgery Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
| | - Thierry Mom
- Otolaryngology Head and Neck Surgery Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
| | - Yves Boirie
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
- Clinical Nutrition Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
| | - Olav Rooyakers
- Anesthesiology and Intensive Care, Department of Clinical Science Intervention and Technology, CLINTEC, Karolinska Institutet, 141 86 Huddinge, Sweden
- Division of Perioperative Medicine and Intensive Care, Karolinska University Hospital, 171 77 Huddinge, Sweden
| | - Stéphane Walrand
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
- Clinical Nutrition Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
| |
Collapse
|
22
|
Della Peruta C, Lozanoska-Ochser B, Renzini A, Moresi V, Sanchez Riera C, Bouché M, Coletti D. Sex Differences in Inflammation and Muscle Wasting in Aging and Disease. Int J Mol Sci 2023; 24:ijms24054651. [PMID: 36902081 PMCID: PMC10003083 DOI: 10.3390/ijms24054651] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Only in recent years, thanks to a precision medicine-based approach, have treatments tailored to the sex of each patient emerged in clinical trials. In this regard, both striated muscle tissues present significant differences between the two sexes, which may have important consequences for diagnosis and therapy in aging and chronic illness. In fact, preservation of muscle mass in disease conditions correlates with survival; however, sex should be considered when protocols for the maintenance of muscle mass are designed. One obvious difference is that men have more muscle than women. Moreover, the two sexes differ in inflammation parameters, particularly in response to infection and disease. Therefore, unsurprisingly, men and women respond differently to therapies. In this review, we present an up-to-date overview on what is known about sex differences in skeletal muscle physiology and disfunction, such as disuse atrophy, age-related sarcopenia, and cachexia. In addition, we summarize sex differences in inflammation which may underly the aforementioned conditions because pro-inflammatory cytokines deeply affect muscle homeostasis. The comparison of these three conditions and their sex-related bases is interesting because different forms of muscle atrophy share common mechanisms; for instance, those responsible for protein dismantling are similar although differing in terms of kinetics, severity, and regulatory mechanisms. In pre-clinical research, exploring sexual dimorphism in disease conditions could highlight new efficacious treatments or recommend implementation of an existing one. Any protective factors discovered in one sex could be exploited to achieve lower morbidity, reduce the severity of the disease, or avoid mortality in the opposite sex. Thus, the understanding of sex-dependent responses to different forms of muscle atrophy and inflammation is of pivotal importance to design innovative, tailored, and efficient interventions.
Collapse
Affiliation(s)
- Chiara Della Peruta
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00161 Roma, Italy
| | - Biliana Lozanoska-Ochser
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00161 Roma, Italy
- Department of Medicine and Surgery, LUM University, 70010 Bari, Italy
| | - Alessandra Renzini
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00161 Roma, Italy
| | - Viviana Moresi
- Institute of Nanotechnology (Nanotec), National Research Council (CNR), c/o Sapienza University of Rome, 00185 Roma, Italy
| | - Carles Sanchez Riera
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00161 Roma, Italy
| | - Marina Bouché
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00161 Roma, Italy
- Correspondence:
| | - Dario Coletti
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00161 Roma, Italy
- Biological Adaptation and Ageing (B2A), Institut de Biologie Paris-Seine, Sorbonne Université, CNRS UMR 8256, Inserm U1164, 75005 Paris, France
| |
Collapse
|
23
|
Gorski PP, Raastad T, Ullrich M, Turner DC, Hallén J, Savari SI, Nilsen TS, Sharples AP. Aerobic exercise training resets the human skeletal muscle methylome 10 years after breast cancer treatment and survival. FASEB J 2023; 37:e22720. [PMID: 36542473 DOI: 10.1096/fj.202201510rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
Cancer survivors suffer impairments in skeletal muscle in terms of reduced mass and function. Interestingly, human skeletal muscle possesses an epigenetic memory of earlier stimuli, such as exercise. Long-term retention of epigenetic changes in skeletal muscle following cancer survival and/or exercise training has not yet been studied. We, therefore, investigated genome-wide DNA methylation (methylome) in skeletal muscle following a 5-month, 3/week aerobic-training intervention in breast cancer survivors 10-14 years after diagnosis and treatment. These results were compared to breast cancer survivors who remained untrained and to age-matched controls with no history of cancer, who undertook the same training intervention. Skeletal muscle biopsies were obtained from 23 females before(pre) and after(post) the 5-month training period. InfiniumEPIC 850K DNA methylation arrays and RT-PCR for gene expression were performed. The breast cancer survivors displayed a significant retention of increased DNA methylation (i.e., hypermethylation) at a larger number of differentially methylated positions (DMPs) compared with healthy age-matched controls pre training. Training in cancer survivors led to an exaggerated number of DMPs with a hypermethylated signature occurring at non-regulatory regions compared with training in healthy age-matched controls. However, the opposite occurred in important gene regulatory regions, where training in cancer survivors elicited a considerable reduction in methylation (i.e., hypomethylation) in 99% of the DMPs located in CpG islands within promoter regions. Importantly, training was able to reverse the hypermethylation identified in cancer survivors back toward a hypomethylated signature that was observed pre training in healthy age-matched controls at 300 (out of 881) of these island/promoter-associated CpGs. Pathway enrichment analysis identified training in cancer survivors evoked a predominantly hypomethylated signature in pathways associated with cell cycle, DNA replication/repair, transcription, translation, mTOR signaling, and the proteosome. Differentially methylated region (DMR) analysis also identified genes: BAG1, BTG2, CHP1, KIFC1, MKL2, MTR, PEX11B, POLD2, S100A6, SNORD104, and SPG7 as hypermethylated in breast cancer survivors, with training reversing these CpG island/promoter-associated DMRs toward a hypomethylated signature. Training also elicited a largely different epigenetic response in healthy individuals than that observed in cancer survivors, with very few overlapping changes. Only one gene, SIRT2, was identified as having altered methylation in cancer survivors at baseline and after training in both the cancer survivors and healthy controls. Overall, human skeletal muscle may retain a hypermethylated signature as long as 10-14 years after breast cancer treatment/survival. Five months of aerobic training reset the skeletal muscle methylome toward signatures identified in healthy age-matched individuals in gene regulatory regions.
Collapse
Affiliation(s)
- Piotr P Gorski
- Institute for Physical Performance (IFP), Norwegian School of Sport Sciences, Oslo, Norway
| | - Truls Raastad
- Institute for Physical Performance (IFP), Norwegian School of Sport Sciences, Oslo, Norway
| | - Max Ullrich
- Institute for Physical Performance (IFP), Norwegian School of Sport Sciences, Oslo, Norway
| | - Daniel C Turner
- Institute for Physical Performance (IFP), Norwegian School of Sport Sciences, Oslo, Norway
| | - Jostein Hallén
- Institute for Physical Performance (IFP), Norwegian School of Sport Sciences, Oslo, Norway
| | - Sebastian Imre Savari
- Department of Cardiology, Oslo University Hospital, Oslo, Norway.,Precision Health Center for Optimized Cardiac Care, Oslo University Hospital, Oslo, Norway
| | - Tormod S Nilsen
- Institute for Physical Performance (IFP), Norwegian School of Sport Sciences, Oslo, Norway
| | - Adam P Sharples
- Institute for Physical Performance (IFP), Norwegian School of Sport Sciences, Oslo, Norway
| |
Collapse
|
24
|
Di Girolamo D, Tajbakhsh S. Pathological features of tissues and cell populations during cancer cachexia. CELL REGENERATION 2022; 11:15. [PMID: 35441960 PMCID: PMC9021355 DOI: 10.1186/s13619-022-00108-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 12/28/2021] [Indexed: 11/10/2022]
Abstract
Cancers remain among the most devastating diseases in the human population in spite of considerable advances in limiting their impact on lifespan and healthspan. The multifactorial nature of cancers, as well as the number of tissues and organs that are affected, have exposed a considerable diversity in mechanistic features that are reflected in the wide array of therapeutic strategies that have been adopted. Cachexia is manifested in a number of diseases ranging from cancers to diabetes and ageing. In the context of cancers, a majority of patients experience cachexia and succumb to death due to the indirect effects of tumorigenesis that drain the energy reserves of different organs. Considerable information is available on the pathophysiological features of cancer cachexia, however limited knowledge has been acquired on the resident stem cell populations, and their function in the context of these diseases. Here we review current knowledge on cancer cachexia and focus on how tissues and their resident stem and progenitor cell populations are individually affected.
Collapse
|
25
|
Parry TL, Tichy L, Brantley JT. Cardioprotective effects of preconditioning exercise in the female tumor bearing mouse. Front Cell Dev Biol 2022; 10:950479. [DOI: 10.3389/fcell.2022.950479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 11/11/2022] [Indexed: 12/04/2022] Open
Abstract
Cancer cachexia, a metabolic wasting syndrome, affects up to 80% of cancer patients and leads to the death in up to 20% of cancer patients. While research is growing in the field, there are still no clear diagnostic criteria and cancer cachexia remains an untreated condition. Aerobic exercise has been shown to positively impact cachexia by slowing its development and attenuating muscle loss. The most effective timing, duration, and intensity of exercise as a preventative and protective measure against cancer cachexia remains questionable. Therefore, the purpose of this study was to examine the effects of preconditioning exercise as a protective measure for tumor-mediated muscle wasting. Female LC3 Tg+ and wildtype mice were randomly separated into four groups, sedentary non-tumor bearing (SED + NT), sedentary tumor bearing (SED + T), treadmill exercise non-tumor bearing (TM + NT), and treadmill exercise tumor bearing (TM + T). Mice underwent an 8-week treadmill exercise training protocol (TM) or remained sedentary (SED). Next, mice were implanted with tumor cells (T group; 5 × 105 Lewis Lung Carcinoma cells in flank) or remained non-tumor (NT) for 4 weeks. Tumor bearing resulted in a significant decline in cardiac function. SED + T showed a significant decrease in fractional shortening (p < 0.05) when compared to the other groups. This coincided with an increase in beclin-1 and MyD88 protein expression and decrease in p-FOXO1 (inactivated) protein expression in SED + T mice. Interestingly, preconditioning exercise (exercise prior to tumor bearing) appeared to preserve cardiac function (TM + T not significantly different than SED + NT). Exercise-mediated cardioprotection also coincided with abolished beclin-1 and MyD88 signaling that was not significantly elevated in TM + T mice. Additionally, TM resulted in a 22-fold decrease in estimated tumor volume (p < 0.05) and a 45% decrease in tumor mass (p < 0.05) compared to SED tumors. The data indicate potential cardioprotective effects of preconditioning exercise on preserving cardiac structure and function, as well as regulating autophagic (beclin-1), inflammatory (TGF-β and MyD88), and atrophy (p-FOXO1) pathways during tumor bearing. Preconditioning exercise may be an effective and accessible treatment intervention for early-stage cancer survivors. This data is crucial in identifying the significance of exercise and the timing of exercise as a protective measure against the detrimental effects of cancer cachexia.
Collapse
|
26
|
Testa MTDJ, Cella PS, Marinello PC, Frajacomo FTT, Padilha CDS, Perandini PC, Moura FA, Duarte JA, Cecchini R, Guarnier FA, Deminice R. Resistance Training Attenuates Activation of STAT3 and Muscle Atrophy in Tumor-Bearing Mice. Front Oncol 2022; 12:880787. [PMID: 35847939 PMCID: PMC9283857 DOI: 10.3389/fonc.2022.880787] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose Although the role of signal transducers and activators of transcription (STAT3) in cachexia due to the association of circulating IL-6 and muscle wasting has been extensively demonstrated, the effect of resistance training on STAT3 in mediating muscle atrophy in tumor-bearing mice is unknown. The aim of this study is to investigate the effects of resistance exercise training on inflammatory cytokines and oxidative-mediated STAT3 activation and muscle loss prevention in tumor-bearing mice. Methods Male Swiss mice were inoculated with Ehrlich tumor cells and exposed or not exposed to resistance exercise protocol of ladder climbing. Skeletal muscle STAT3 protein content was measured, compared between groups, and tested for possible association with plasma interleukins and local oxidative stress markers. Components of the ubiquitin-proteasome and autophagy pathways were assessed by real-time PCR or immunoblotting. Results Resistance training prevented STAT3 excessive activation in skeletal muscle mediated by the overabundance of plasma IL-6 and muscle oxidative stress. These mechanisms contributed to preventing the increased key genes and proteins of ubiquitin-proteasome and autophagy pathways in tumor-bearing mice, such as Atrogin-1, LC3B-II, and Beclin-1. Beyond preventing muscle atrophy, RT also prevented strength loss and impaired locomotor capacity, hallmarks of sarcopenia. Conclusion Our results suggest that STAT3 inhibition is central in resistance exercise protective effects against cancer-induced muscle atrophy and strength loss.
Collapse
Affiliation(s)
| | - Paola Sanches Cella
- Department of Physical Education, State University of Londrina, Londrina, Brazil
| | - Poliana Camila Marinello
- Department of Physical Education, State University of Londrina, Londrina, Brazil
- Department of General Pathology, State University of Londrina, Londrina, Brazil
| | | | - Camila de Souza Padilha
- Department of Physical Education, State University of Londrina, Londrina, Brazil
- Department of Physical Education, State University of São Paulo (UNESP), Presidente Prudente, Brazil
| | | | - Felipe Arruda Moura
- Department of Physical Education, State University of Londrina, Londrina, Brazil
| | | | - Rubens Cecchini
- Department of General Pathology, State University of Londrina, Londrina, Brazil
| | | | - Rafael Deminice
- Department of Physical Education, State University of Londrina, Londrina, Brazil
- *Correspondence: Rafael Deminice, ; orcid.org/0000-0002-9246-1079
| |
Collapse
|
27
|
Morena da Silva F, Rosa-Caldwell ME, Schrems ER, Martinez L, Amos MG, Lim S, Cabrera AR, Brown JL, Washington TA, Greene NP. PGC-1α overexpression is not sufficient to mitigate cancer cachexia in either male or female mice. Appl Physiol Nutr Metab 2022; 47:933-948. [PMID: 35700525 DOI: 10.1139/apnm-2022-0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cancer-cachexia accounts for 20-40% of cancer-related deaths. Mitochondrial aberrations have been shown to precede muscle atrophy in different atrophy models, including cancer. Therefore, this study investigated potential protection from the cachectic phenotype through overexpression of PGC-1α. First, to establish potential of mitochondria-based approaches we showed that the mitochondrial antioxidant mitoTEMPO attenuates myotube atrophy induced by Lewis Lung Carcinoma (LLC) cell conditioned media. Next, cachexia was induced in muscle specific PGC-1α overexpressing (MCK-PCG1α) or wildtype (WT) littermate mice by LLC implantation. MCK-PCG1α did not protect LLC-induced muscle mass loss. In plantaris, Atrogin mRNA content was 6.2-fold and ~11-fold greater in WT-LLC vs. WT-PBS for males and females, respectively (p<0.05). MitoTimer red:green ratio for male PGC was ~65% higher than WT groups (p<0.05), with ~3-fold more red puncta in LLC than PBS (p<0.05). Red:green ratio was ~56% lower in females WT-LLC vs. PGC-LLC (p<0.05). In females, no change in red puncta was noted across conditions. Lc3 mRNA content was ~ 73% and 2-fold higher in male and female LLC mice respectively vs. PBS (p<0.05). While MitoTEMPO could mitigate cancer-induced atrophy in vitro, PGC1α overexpression was insufficient to protect muscle mass and mitochondrial health in vivo despite mitigation of cachexia-associated signaling pathways.
Collapse
Affiliation(s)
| | | | - Eleanor R Schrems
- University of Arkansas Fayetteville, 3341, Fayetteville, Arkansas, United States;
| | - Lauren Martinez
- University of Arkansas Fayetteville, 3341, HHPR, Fayetteville, Arkansas, United States;
| | - Madeline G Amos
- University of Arkansas Fayetteville, 3341, HHPR, Fayetteville, Arkansas, United States;
| | - Seongkyun Lim
- University of Arkansas Fayetteville, 3341, HHPR, Fayetteville, Arkansas, United States;
| | - Ana Regina Cabrera
- University of Arkansas Fayetteville, 3341, HHPR, Fayetteville, Arkansas, United States;
| | - Jacob L Brown
- University of Arkansas Fayetteville, 3341, Health, Human Performance and Recreation, Fayetteville, Arkansas, United States;
| | - Tyrone A Washington
- University of Arkansas Fayetteville, 3341, Health, Human Performance, and Recreation, Fayetteville, Arkansas, United States;
| | - Nicholas P Greene
- University of Arkansas Fayetteville, 3341, Health, Human Performance and Recreation, Fayetteville, Arkansas, United States;
| |
Collapse
|
28
|
Cancer Cachexia and Antitumor Immunity: Common Mediators and Potential Targets for New Therapies. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060880. [PMID: 35743911 PMCID: PMC9225288 DOI: 10.3390/life12060880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 12/23/2022]
Abstract
Cancer cachexia syndrome (CCS) is a multifactorial metabolic syndrome affecting a significant proportion of patients. CCS is characterized by progressive weight loss, alterations of body composition and a systemic inflammatory status, which exerts a major impact on the host’s innate and adaptive immunity. Over the last few years, the development of immune checkpoint inhibitors (ICIs) transformed the treatment landscape for a wide spectrum of malignancies, creating an unprecedented opportunity for long term remissions in a significant subset of patients. Early clinical data indicate that CCS adversely impairs treatment outcomes of patients receiving ICIs. We herein reviewed existing evidence on the potential links between the mechanisms that promote the catabolic state in CCS and those that impair the antitumor immune response. We show that the biological mediators and processes leading to the development of CCS may also participate in the modulation and the sustainment of an immune suppressive tumor microenvironment and impaired anti-tumor immunity. Moreover, we demonstrate that the deregulation of the host’s metabolic homeostasis in cancer cachexia is associated with resistance to ICIs. Further research on the interrelation between cancer cachexia and anti-tumor immunity is required for the effective management of resistance to immunotherapy in this specific but large subgroup of ICI treated individuals.
Collapse
|
29
|
Mallard J, Hucteau E, Charles AL, Bender L, Baeza C, Pélissie M, Trensz P, Pflumio C, Kalish-Weindling M, Gény B, Schott R, Favret F, Pivot X, Hureau TJ, Pagano AF. Chemotherapy impairs skeletal muscle mitochondrial homeostasis in early breast cancer patients. J Cachexia Sarcopenia Muscle 2022; 13:1896-1907. [PMID: 35373507 PMCID: PMC9178151 DOI: 10.1002/jcsm.12991] [Citation(s) in RCA: 22] [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: 10/13/2021] [Revised: 02/22/2022] [Accepted: 03/07/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Chemotherapy is extensively used to treat breast cancer and is associated with skeletal muscle deconditioning, which is known to reduce patients' quality of life, treatment efficiency, and overall survival. To date, skeletal muscle mitochondrial alterations represent a major aspect explored in breast cancer patients; nevertheless, the cellular mechanisms remain relatively unknown. This study was dedicated to investigating overall skeletal muscle mitochondrial homeostasis in early breast cancer patients undergoing chemotherapy, including mitochondrial quantity, function, and dynamics. METHODS Women undergoing (neo)adjuvant anthracycline-cyclophosphamide and taxane-based chemotherapy participated in this study (56 ± 12 years). Two muscle biopsies were collected from the vastus lateralis muscle before the first and after the last chemotherapy administration. Mitochondrial respiratory capacity, reactive oxygen species production, and western blotting analyses were performed. RESULTS Among the 11 patients, we found a decrease in key markers of mitochondrial quantity, reaching -52.0% for citrate synthase protein levels (P = 0.02) and -38.2% for VDAC protein levels (P = 0.04). This mitochondrial content loss is likely explained by reduced mitochondrial biogenesis, as evidenced by a decrease in PGC-1α1 protein levels (-29.5%; P = 0.04). Mitochondrial dynamics were altered, as documented by a decrease in MFN2 protein expression (-33.4%; P = 0.01), a key marker of mitochondrial outer membrane fusion. Mitochondrial fission is a prerequisite for mitophagy activation, and no variation was found in either key markers of mitochondrial fission (Fis1 and DRP1) or mitophagy (Parkin, PINK1, and Mul1). Two contradictory hypotheses arise from these results: defective mitophagy, which probably increases the number of damaged and fragmented mitochondria, or a relative increase in mitophagy through elevated mitophagic potential (Parkin/VDAC ratio; +176.4%; P < 0.02). Despite no change in mitochondrial respiratory capacity and COX IV protein levels, we found an elevation in H2 O2 production (P < 0.05 for all substrate additions) without change in antioxidant enzymes. We investigated the apoptosis pathway and found an increase in the protein expression of the apoptosis initiation marker Bax (+72.0%; P = 0.04), without variation in the anti-apoptotic protein Bcl-2. CONCLUSIONS This study demonstrated major mitochondrial alterations subsequent to chemotherapy in early breast cancer patients: (i) a striking reduction in mitochondrial biogenesis, (ii) altered mitochondrial dynamics and potential mitophagy defects, (iii) exacerbated H2 O2 production, and (iv) increased initiation of apoptosis. All of these alterations likely explain, at least in part, the high prevalence of skeletal muscle and cardiorespiratory deconditioning classically observed in breast cancer patients.
Collapse
Affiliation(s)
- Joris Mallard
- Faculté de médecine, maïeutique et sciences de la santé, "Mitochondrie, Stress oxydant, Protection musculaire", Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France.,Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Elyse Hucteau
- Faculté de médecine, maïeutique et sciences de la santé, "Mitochondrie, Stress oxydant, Protection musculaire", Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France.,Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Anne-Laure Charles
- Faculté de médecine, maïeutique et sciences de la santé, "Mitochondrie, Stress oxydant, Protection musculaire", Université de Strasbourg, Strasbourg, France
| | - Laura Bender
- Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Claire Baeza
- Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Mathilde Pélissie
- Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Philippe Trensz
- Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Carole Pflumio
- Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | | | - Bernard Gény
- Faculté de médecine, maïeutique et sciences de la santé, "Mitochondrie, Stress oxydant, Protection musculaire", Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France
| | - Roland Schott
- Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Fabrice Favret
- Faculté de médecine, maïeutique et sciences de la santé, "Mitochondrie, Stress oxydant, Protection musculaire", Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France
| | - Xavier Pivot
- Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Thomas J Hureau
- Faculté de médecine, maïeutique et sciences de la santé, "Mitochondrie, Stress oxydant, Protection musculaire", Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France
| | - Allan F Pagano
- Faculté de médecine, maïeutique et sciences de la santé, "Mitochondrie, Stress oxydant, Protection musculaire", Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France
| |
Collapse
|
30
|
Nunes EA, Stokes T, McKendry J, Currier BS, Phillips SM. Disuse-induced skeletal muscle atrophy in disease and non-disease states in humans: mechanisms, prevention, and recovery strategies. Am J Physiol Cell Physiol 2022; 322:C1068-C1084. [PMID: 35476500 DOI: 10.1152/ajpcell.00425.2021] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Decreased skeletal muscle contractile activity (disuse) or unloading leads to muscle mass loss, also known as muscle atrophy. The balance between muscle protein synthesis (MPS) and muscle protein breakdown (MPB) is the primary determinant of skeletal muscle mass. A reduced mechanical load on skeletal muscle is one of the main external factors leading to muscle atrophy. However, endocrine and inflammatory factors can act synergistically in catabolic states, amplifying the atrophy process and accelerating its progression. Additionally, older individuals display aging-induced anabolic resistance, which can predispose this population to more pronounced effects when exposed to periods of reduced physical activity or mechanical unloading. Different cellular mechanisms contribute to the regulation of muscle protein balance during skeletal muscle atrophy. This review summarizes the effects of muscle disuse on muscle protein balance and the molecular mechanisms involved in muscle atrophy in the absence or presence of disease. Finally, a discussion of the current literature describing efficient strategies to prevent or improve the recovery from muscle atrophy is also presented.
Collapse
Affiliation(s)
- Everson A Nunes
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada.,Laboratory of Investigation of Chronic Diseases, Department of Physiological Sciences, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Tanner Stokes
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - James McKendry
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Brad S Currier
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Stuart M Phillips
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
31
|
Abstract
Autophagy is an important life phenomenon in eukaryotic cells. Its main role is to remove and degrade its damaged organelles and excess biological macromolecules, and use degradation products to provide energy and rebuild the cell structure, playing an important role in maintaining cell homeostasis and cell life activities. Mitophagy is a form of macroautophagy. It has the beneficial effect of eliminating damaged mitochondria, thereby maintaining the integrity of the mitochondrial pool. Autophagy and mitophagy have a dual role in the development of cancer. On one hand, autophagy and mitophagy can maintain the normal physiological function of cells. On the other hand, excessive autophagy and mitophagy can lead to diseases. The present review introduces the mechanisms of autophagy and mitophagy, and the main related proteins, and introduce the correlation with cancers, providing a basis for the treatment of cancers through the understanding of these proteins.
Collapse
Affiliation(s)
- Hong-Ming Xu
- Department of Orthopaedic Surgery, Affiliated Cixi Hospital of Wenzhou Medical University, Cixi, Ningbo, People's Republic of China
| | - Fei Hu
- Diabetes Research Center, School of Medicine, Ningbo University, Ningbo, People's Republic of China
| |
Collapse
|
32
|
Pin F, Huot JR, Bonetto A. The Mitochondria-Targeting Agent MitoQ Improves Muscle Atrophy, Weakness and Oxidative Metabolism in C26 Tumor-Bearing Mice. Front Cell Dev Biol 2022; 10:861622. [PMID: 35392166 PMCID: PMC8980422 DOI: 10.3389/fcell.2022.861622] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/07/2022] [Indexed: 01/06/2023] Open
Abstract
Cancer cachexia is a debilitating syndrome characterized by skeletal muscle wasting, weakness and fatigue. Several pathogenetic mechanisms can contribute to these muscle derangements. Mitochondrial alterations, altered metabolism and increased oxidative stress are known to promote muscle weakness and muscle catabolism. To the extent of improving cachexia, several drugs have been tested to stimulate mitochondrial function and normalize the redox balance. The aim of this study was to test the potential beneficial anti-cachectic effects of Mitoquinone Q (MitoQ), one of the most widely-used mitochondria-targeting antioxidant. Here we show that MitoQ administration (25 mg/kg in drinking water, daily) in vivo was able to improve body weight loss in Colon-26 (C26) bearers, without affecting tumor size. Consistently, the C26 hosts displayed ameliorated skeletal muscle and strength upon treatment with MitoQ. In line with improved skeletal muscle mass, the treatment with MitoQ was able to partially correct the expression of the E3 ubiquitin ligases Atrogin-1 and Murf1. Contrarily, the anabolic signaling was not improved by the treatment, as showed by unchanged AKT, mTOR and 4EBP1 phosphorylation. Assessment of gene expression showed altered levels of markers of mitochondrial biogenesis and homeostasis in the tumor hosts, although only Mitofusin-2 levels were significantly affected by the treatment. Interestingly, the levels of Pdk4 and CytB, genes involved in the regulation of mitochondrial function and metabolism, were also partially increased by MitoQ, in line with the modulation of hexokinase (HK), pyruvate dehydrogenase (PDH) and succinate dehydrogenase (SDH) enzymatic activities. The improvement of the oxidative metabolism was associated with reduced myosteatosis (i.e., intramuscular fat infiltration) in the C26 bearers receiving MitoQ, despite unchanged muscle LDL receptor expression, therefore suggesting that MitoQ could boost β-oxidation in the muscle tissue and promote a glycolytic-to-oxidative shift in muscle metabolism and fiber composition. Overall, our data identify MitoQ as an effective treatment to improve skeletal muscle mass and function in tumor hosts and further support studies aimed at testing the anti-cachectic properties of mitochondria-targeting antioxidants also in combination with routinely administered chemotherapy agents.
Collapse
Affiliation(s)
- Fabrizio Pin
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Joshua R. Huot
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrea Bonetto
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- *Correspondence: Andrea Bonetto,
| |
Collapse
|
33
|
Quinlan JI, Dhaliwal A, Williams F, Allen SL, Breen L, Greig CA, Lord JM, Armstrong MJ, Elsharkawy AM. Feasibility, Efficacy, and Safety of Percutaneous Muscle Biopsies in Patients With Chronic Liver Disease. Front Physiol 2022; 12:817152. [PMID: 35242045 PMCID: PMC8886882 DOI: 10.3389/fphys.2021.817152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Sarcopenia is present in many chronic disease states including decompensated end stage liver disease (ESLD) and non-cirrhotic non-alcoholic fatty liver disease (NAFLD). Sarcopenia in ESLD can negatively impact quality of life and increase mortality. Despite this, very little is understood about the mechanisms of sarcopenia in these conditions. One key reason for this is the reluctance to undertake percutaneous muscle biopsies due to the perceived increased risks. ESLD can induce thrombocytopaenia and coagulopathy which significantly increases the risk of bleeding. In addition, patients with either NAFLD or ESLD often have co-morbidities that would require additional care and risk assessment. Thus, the aim of this study was to establish an effective and safe protocol for the implementation of percutaneous muscle biopsies in patients with NAFLD and ESLD. METHODS A total of 47 patients with ESLD and 9 patients with non-cirrhotic NAFLD were recruited from the Liver Unit, Queen Elizabeth Hospital (Birmingham, United Kingdom). A total of 71 percutaneous vastus lateralis biopsies were attempted over two study visits. A vigorous safety screening occurred prior to and during each visit and a strict protocol was followed to mitigate against complications and risk. RESULTS A total of 85% of patients consented to the muscle biopsy at either visit (48/56). A total of 9% of consented biopsies could not occur due to medical considerations, including high international normalised ratio (INR) (n = 3) and the use of aspirin (n = 4). Muscle tissue was obtained from 90% of attempts, with a mean average yield (wet weight tissue) of 98.1 ± 52.9 mg. CONCLUSION Percutaneous muscle biopsies are both feasible and yield sufficient tissue in an ESLD population. The procedure is effective for obtaining muscle tissue whilst also safe, with only one adverse event. This study provides evidence for the successful use of muscle biopsies in this population, even in consideration of disease specific complications, medications, and comorbidities.
Collapse
Affiliation(s)
- Jonathan I Quinlan
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Amritpal Dhaliwal
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Felicity Williams
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Sophie L Allen
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Leigh Breen
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom.,MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - Carolyn A Greig
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom.,MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - Janet M Lord
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - Matthew J Armstrong
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Ahmed M Elsharkawy
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| |
Collapse
|
34
|
Reyes-Castellanos G, Abdel Hadi N, Carrier A. Autophagy Contributes to Metabolic Reprogramming and Therapeutic Resistance in Pancreatic Tumors. Cells 2022; 11:426. [PMID: 35159234 PMCID: PMC8834004 DOI: 10.3390/cells11030426] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Metabolic reprogramming is a feature of cancers for which recent research has been particularly active, providing numerous insights into the mechanisms involved. It occurs across the entire cancer process, from development to resistance to therapies. Established tumors exhibit dependencies for metabolic pathways, constituting vulnerabilities that can be targeted in the clinic. This knowledge is of particular importance for cancers that are refractory to any therapeutic approach, such as Pancreatic Ductal Adenocarcinoma (PDAC). One of the metabolic pathways dysregulated in PDAC is autophagy, a survival process that feeds the tumor with recycled intracellular components, through both cell-autonomous (in tumor cells) and nonautonomous (from the local and distant environment) mechanisms. Autophagy is elevated in established PDAC tumors, contributing to aberrant proliferation and growth even in a nutrient-poor context. Critical elements link autophagy to PDAC including genetic alterations, mitochondrial metabolism, the tumor microenvironment (TME), and the immune system. Moreover, high autophagic activity in PDAC is markedly related to resistance to current therapies. In this context, combining autophagy inhibition with standard chemotherapy, and/or drugs targeting other vulnerabilities such as metabolic pathways or the immune response, is an ongoing clinical strategy for which there is still much to do through translational and multidisciplinary research.
Collapse
Affiliation(s)
| | | | - Alice Carrier
- Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS, INSERM, Institut Paoli-Calmettes, Aix Marseille Université, F-13009 Marseille, France; (G.R.-C.); (N.A.H.)
| |
Collapse
|
35
|
Bordignon C, dos Santos BS, Rosa DD. Impact of Cancer Cachexia on Cardiac and Skeletal Muscle: Role of Exercise Training. Cancers (Basel) 2022; 14:cancers14020342. [PMID: 35053505 PMCID: PMC8773522 DOI: 10.3390/cancers14020342] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/27/2021] [Accepted: 01/07/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cachexia is a syndrome that can be present in many patients diagnosed with cancer, especially in those with metastatic or very advanced tumors. The patient may present with weight loss, loss of muscle mass, and even cardiac dysfunction as a result of it. The aim of this review is to understand how cachexia manifests and whether physical exercise has any role in trying to prevent or reverse this syndrome in cancer patients. Abstract Cachexia is a multifactorial syndrome that presents with, among other characteristics, progressive loss of muscle mass and anti-cardiac remodeling effect that may lead to heart failure. This condition affects about 80% of patients with advanced cancer and contributes to worsening patients’ tolerance to anticancer treatments and to their premature death. Its pathogenesis involves an imbalance in metabolic homeostasis, with increased catabolism and inflammatory cytokines levels, leading to proteolysis and lipolysis, with insufficient food intake. A multimodal approach is indicated for patients with cachexia, with the aim of reducing the speed of muscle wasting and improving their quality of life, which may include nutritional, physical, pharmacologic, and psychological support. This review aims to outline the mechanisms of muscle loss, as well as to evaluate the current clinical evidence of the use of physical exercise in patients with cachexia.
Collapse
Affiliation(s)
- Cláudia Bordignon
- Oncology Center, Hospital Moinhos de Vento, Porto Alegre 90560-030, Brazil;
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-070, Brazil
| | - Bethânia S. dos Santos
- Department of Clinical Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20560-121, Brazil;
- Rede D’Or São Luiz, Rio de Janeiro 22271-110, Brazil
| | - Daniela D. Rosa
- Oncology Center, Hospital Moinhos de Vento, Porto Alegre 90560-030, Brazil;
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-070, Brazil
- Brazilian Breast Cancer Study Group (GBECAM), Porto Alegre 90619-900, Brazil
- Correspondence:
| |
Collapse
|
36
|
Abstract
Diverse inflammatory diseases, infections and malignancies are associated with wasting syndromes. In many of these conditions, the standards for diagnosis and treatment are lacking due to our limited understanding of the causative molecular mechanisms. Here, we discuss the complex immunological context of cachexia, a systemic catabolic syndrome that depletes both fat and muscle mass with profound consequences for patient prognosis. We highlight the main cytokine and immune cell-driven pathways that have been linked to weight loss and tissue wasting in the context of cancer-associated and infection-associated cachexia. Moreover, we discuss the potential immunometabolic consequences of cachexia on the basis of newly identified pathways and explore the multilayered area of immunometabolic crosstalk both upstream and downstream of tissue catabolism. Collectively, this Review highlights the intricate relationship of the immune system with cachexia in the context of malignant and infectious diseases.
Collapse
|
37
|
Esposito P, Picciotto D, Battaglia Y, Costigliolo F, Viazzi F, Verzola D. Myostatin: Basic biology to clinical application. Adv Clin Chem 2021; 106:181-234. [PMID: 35152972 DOI: 10.1016/bs.acc.2021.09.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Myostatin is a member of the transforming growth factor (TGF)-β superfamily. It is expressed by animal and human skeletal muscle cells where it limits muscle growth and promotes protein breakdown. Its effects are influenced by complex mechanisms including transcriptional and epigenetic regulation and modulation by extracellular binding proteins. Due to its actions in promoting muscle atrophy and cachexia, myostatin has been investigated as a promising therapeutic target to counteract muscle mass loss in experimental models and patients affected by different muscle-wasting conditions. Moreover, growing evidence indicates that myostatin, beyond to regulate skeletal muscle growth, may have a role in many physiologic and pathologic processes, such as obesity, insulin resistance, cardiovascular and chronic kidney disease. In this chapter, we review myostatin biology, including intracellular and extracellular regulatory pathways, and the role of myostatin in modulating physiologic processes, such as muscle growth and aging. Moreover, we discuss the most relevant experimental and clinical evidence supporting the extra-muscle effects of myostatin. Finally, we consider the main strategies developed and tested to inhibit myostatin in clinical trials and discuss the limits and future perspectives of the research on myostatin.
Collapse
Affiliation(s)
- Pasquale Esposito
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Daniela Picciotto
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Yuri Battaglia
- Nephrology and Dialysis Unit, St. Anna University Hospital, Ferrara, Italy
| | - Francesca Costigliolo
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesca Viazzi
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Daniela Verzola
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
38
|
A Blueprint for Cancer-Related Inflammation and Host Innate Immunity. Cells 2021; 10:cells10113211. [PMID: 34831432 PMCID: PMC8623541 DOI: 10.3390/cells10113211] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/27/2021] [Accepted: 11/10/2021] [Indexed: 12/30/2022] Open
Abstract
Both in situ and allograft models of cancer in juvenile and adult Drosophila melanogaster fruit flies offer a powerful means for unravelling cancer gene networks and cancer-host interactions. They can also be used as tools for cost-effective drug discovery and repurposing. Moreover, in situ modeling of emerging tumors makes it possible to address cancer initiating events-a black box in cancer research, tackle the innate antitumor immune responses to incipient preneoplastic cells and recurrent growing tumors, and decipher the initiation and evolution of inflammation. These studies in Drosophila melanogaster can serve as a blueprint for studies in more complex organisms and help in the design of mechanism-based therapies for the individualized treatment of cancer diseases in humans. This review focuses on new discoveries in Drosophila related to the diverse innate immune responses to cancer-related inflammation and the systemic effects that are so detrimental to the host.
Collapse
|
39
|
Bae JH, Seo DY, Lee SH, Shin C, Jamrasi P, Han J, Song W. Effects of exercise on AKT/PGC1-α/FOXO3a pathway and muscle atrophy in cisplatin-administered rat skeletal muscle. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2021; 25:585-592. [PMID: 34697269 PMCID: PMC8552830 DOI: 10.4196/kjpp.2021.25.6.585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 01/06/2023]
Abstract
Cisplatin has been reported to cause side effects such as muscle wasting in
humans and rodents. The physiological mechanisms involved in preventing muscle
wasting, such as the regulation of AKT, PGC1-α, and autophagy-related
factor FOXO3a by MuRF 1 and Atrogin-1, remain unclear following different types
of exercise and in various skeletal muscle types. Eight-week-old male Wistar
rats (n = 34) were assigned to one of four groups: control (CON, n = 6),
cisplatin injection (1 mg/kg) without exercise (CC, n = 8), cisplatin (1 mg/kg)
+ resistance exercise (CRE, n = 9) group, and cisplatin (1 mg/kg) + aerobic
exercise (CAE, n = 11). The CRE group performed progressive ladder exercise
(starting with 10% of body weight on a 1-m ladder with 2-cm-interval grids, at
85°) for 8 weeks. The CAE group exercised by treadmill running (20 m/min
for 60 min daily, 4 times/week) for 8 weeks. Compared with the CC group, the
levels of the autophagy-related factors BNIP3, Beclin 1, LC3-II/I ratio, p62,
and FOXO3a in the gastrocnemius and soleus muscles were significantly decreased
in the CRE and CAE groups. The CRE and CAE groups further showed significantly
decreased MuRF 1 and Atrogin-1 levels and increased phosphorylation of AKT,
FOXO3a, and PGC1-α. These results suggest that both ladder and aerobic
exercise directly affected muscle wasting by modulating the
AKT/PGC1-α/FOXO3a signaling pathways regardless of the skeletal muscle
type.
Collapse
Affiliation(s)
- Jun Hyun Bae
- Health and Exercise Science Laboratory, Institute of Sports Science, Seoul National University, Seoul 08826, Korea
| | - Dae Yun Seo
- National ResearchLaboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Smart Marine Therapeutics Center, Cardiovascular and Metabolic Disease Center, Inje University
| | - Sang Ho Lee
- Department of Taekwondo, Dong-A University, Busan 49315, Korea
| | - Chaeyoung Shin
- Health and Exercise Science Laboratory, Institute of Sports Science, Seoul National University, Seoul 08826, Korea
| | - Parivash Jamrasi
- Health and Exercise Science Laboratory, Institute of Sports Science, Seoul National University, Seoul 08826, Korea
| | - Jin Han
- National ResearchLaboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Smart Marine Therapeutics Center, Cardiovascular and Metabolic Disease Center, Inje University
| | - Wook Song
- Health and Exercise Science Laboratory, Institute of Sports Science, Seoul National University, Seoul 08826, Korea.,Institute of Aging, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
40
|
Hain BA, Xu H, VanCleave AM, Gordon BS, Kimball SR, Waning DL. REDD1 deletion attenuates cancer cachexia in mice. J Appl Physiol (1985) 2021; 131:1718-1730. [PMID: 34672766 PMCID: PMC10392697 DOI: 10.1152/japplphysiol.00536.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cancer cachexia is a wasting disorder associated with advanced cancer that contributes to mortality. Cachexia is characterized by involuntary loss of body weight and muscle weakness that affects physical function. Regulated in DNA damage and development 1 (REDD1) is a stress-response protein that is transcriptionally upregulated in muscle during wasting conditions and inhibits mechanistic target of rapamycin complex 1 (mTORC1). C2C12 myotubes treated with Lewis lung carcinoma (LLC)-conditioned media increased REDD1 mRNA expression and decreased myotube diameter. To investigate the role of REDD1 in cancer cachexia, we inoculated 12-week old male wild-type or global REDD1 knockout (REDD1 KO) mice with LLC cells and euthanized 28-days later. Wild-type mice had increased skeletal muscle REDD1 expression, and REDD1 deletion prevented loss of body weight and lean tissue mass, but not fat mass. We found that REDD1 deletion attenuated loss of individual muscle weights and loss of myofiber cross sectional area. We measured markers of the Akt/mTORC1 pathway and found that, unlike wild-type mice, phosphorylation of both Akt and 4E-BP1 was maintained in the muscle of REDD1 KO mice after LLC inoculation, suggesting that loss of REDD1 is beneficial in maintaining mTORC1 activity in mice with cancer cachexia. We measured Foxo3a phosphorylation as a marker of the ubiquitin proteasome pathway and autophagy and found that REDD1 deletion prevented dephosphorylation of Foxo3a in muscles from cachectic mice. Our data provides evidence that REDD1 plays an important role in cancer cachexia through the regulation of both protein synthesis and protein degradation pathways.
Collapse
Affiliation(s)
- Brian A Hain
- The Penn State College of Medicine, Dept. of Cellular and Molecular Physiology, Hershey, PA, United States.,Penn State Cancer Institute, Hershey, PA, United States
| | - Haifang Xu
- The Penn State College of Medicine, Dept. of Cellular and Molecular Physiology, Hershey, PA, United States
| | - Ashley M VanCleave
- The Penn State College of Medicine, Dept. of Cellular and Molecular Physiology, Hershey, PA, United States
| | - Bradley S Gordon
- Florida State University, Dept. of Nutrition and Integrative Physiology, Tallahassee, FL, United States
| | - Scot R Kimball
- The Penn State College of Medicine, Dept. of Cellular and Molecular Physiology, Hershey, PA, United States
| | - David L Waning
- The Penn State College of Medicine, Dept. of Cellular and Molecular Physiology, Hershey, PA, United States.,Penn State Cancer Institute, Hershey, PA, United States
| |
Collapse
|
41
|
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
|
42
|
Mallard J, Hucteau E, Hureau TJ, Pagano AF. Skeletal Muscle Deconditioning in Breast Cancer Patients Undergoing Chemotherapy: Current Knowledge and Insights From Other Cancers. Front Cell Dev Biol 2021; 9:719643. [PMID: 34595171 PMCID: PMC8476809 DOI: 10.3389/fcell.2021.719643] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/10/2021] [Indexed: 01/18/2023] Open
Abstract
Breast cancer represents the most commonly diagnosed cancer while neoadjuvant and adjuvant chemotherapies are extensively used in order to reduce tumor development and improve disease-free survival. However, chemotherapy also leads to severe off-target side-effects resulting, together with the tumor itself, in major skeletal muscle deconditioning. This review first focuses on recent advances in both macroscopic changes and cellular mechanisms implicated in skeletal muscle deconditioning of breast cancer patients, particularly as a consequence of the chemotherapy treatment. To date, only six clinical studies used muscle biopsies in breast cancer patients and highlighted several important aspects of muscle deconditioning such as a decrease in muscle fibers cross-sectional area, a dysregulation of protein turnover balance and mitochondrial alterations. However, in comparison with the knowledge accumulated through decades of intensive research with many different animal and human models of muscle atrophy, more studies are necessary to obtain a comprehensive understanding of the cellular processes implicated in breast cancer-mediated muscle deconditioning. This understanding is indeed essential to ultimately lead to the implementation of efficient preventive strategies such as exercise, nutrition or pharmacological treatments. We therefore also discuss potential mechanisms implicated in muscle deconditioning by drawing a parallel with other cancer cachexia models of muscle wasting, both at the pre-clinical and clinical levels.
Collapse
Affiliation(s)
- Joris Mallard
- Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France.,Centre de Recherche en Biomédecine de Strasbourg (CRBS), Fédération de Médecine Translationnelle, UR 3072, Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France
| | - Elyse Hucteau
- Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France.,Centre de Recherche en Biomédecine de Strasbourg (CRBS), Fédération de Médecine Translationnelle, UR 3072, Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France
| | - Thomas J Hureau
- Centre de Recherche en Biomédecine de Strasbourg (CRBS), Fédération de Médecine Translationnelle, UR 3072, Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France
| | - Allan F Pagano
- Centre de Recherche en Biomédecine de Strasbourg (CRBS), Fédération de Médecine Translationnelle, UR 3072, Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France
| |
Collapse
|
43
|
Liu H, Zang P, Lee I(I, Anderson B, Christiani A, Strait‐Bodey L, Breckheimer BA, Storie M, Tewnion A, Krumm K, Li T, Irwin B, Garcia JM. Growth hormone secretagogue receptor-1a mediates ghrelin's effects on attenuating tumour-induced loss of muscle strength but not muscle mass. J Cachexia Sarcopenia Muscle 2021; 12:1280-1295. [PMID: 34264027 PMCID: PMC8517358 DOI: 10.1002/jcsm.12743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 05/11/2021] [Accepted: 06/08/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Ghrelin may ameliorate cancer cachexia (CC) by preventing anorexia, muscle, and fat loss. However, the mechanisms mediating these effects are not fully understood. This study characterizes the pathways involved in muscle mass and strength loss in the Lewis lung carcinoma (LLC)-induced cachexia model, and the effects of ghrelin in mice with or without its only known receptor: the growth hormone secretagogue receptor-1a ((GHSR-1a), Ghsr+/+ and Ghsr-/- ). METHODS Five to 7-month-old male C57BL/6J Ghsr+/+ and Ghsr-/- mice were inoculated with 1 × 106 heat-killed (HK) or live LLC cells (tumour implantation, TI). When tumours were palpable (7 days after TI), tumour-bearing mice were injected with vehicle (T + V) or ghrelin twice/day for 14 days (T + G, 0.8 mg/kg), while HK-treated mice were given vehicle (HK + V). Body weight and grip strength were evaluated before TI and at termination (21 days after TI). Hindlimb muscles were collected for analysis. RESULTS Less pronounced body weight (BW) loss (87.70 ± 0.98% vs. 83.92 ± 1.23%, percentage of baseline BW in tumour-bearing Ghsr+/+ vs. Ghsr-/- , P = 0.008), and lower upregulation of ubiquitin-proteasome system (UPS, MuRF1/Trim63, 5.71 ± 1.53-fold vs. 9.22 ± 1.94-fold-change from Ghsr+/+ HK + V in tumour-bearing Ghsr+/+ vs. Ghsr-/- , P = 0.036) and autophagy markers (Becn1, Atg5, Atg7, tumour-bearing Ghsr+/+ < Ghsr-/- , all P < 0.02) were found in T + V Ghsr+/+ vs. Ghsr-/- mice. Ghrelin attenuated LLC-induced UPS marker upregulation in both genotypes, [Trim63 was decreased from 5.71 ± 1.53-fold to 1.96 ± 0.47-fold in Ghsr+/+ (T + V vs. T + G: P = 0.032) and 9.22 ± 1.94-fold to 4.72 ± 1.06-fold in Ghsr-/- (T + V vs. T + G: P = 0.008)]. Only in Ghsr+/+ mice ghrelin ameliorated LLC-induced grip strength loss [improved from 89.24 ± 3.48% to 97.80 ± 2.31% of baseline (T + V vs. T + G: P = 0.042)], mitophagy markers [Bnip3 was decreased from 2.28 ± 0.56 to 1.38 ± 0.14-fold (T + V vs. T + G: P ≤ 0.05)], and impaired mitochondrial respiration [State 3u improved from 698.23 ± 73.96 to 934.37 ± 95.21 pmol/min (T + V vs. T + G: P ≤ 0.05)], whereas these markers were not improved by ghrelin Ghsr-/- . Compared with Ghsr+/+ , Ghsr-/- tumour-bearing mice also showed decreased response to ghrelin in BW [T + G-treated Ghsr+/+ vs. Ghsr -/- : 91.75 ± 1.05% vs. 86.18 ± 1.13% of baseline BW, P < 0.001)], gastrocnemius (T + G-treated Ghsr+/+ vs. Ghsr-/- : 96.9 ± 2.08% vs. 88.15 ± 1.78% of Ghsr+/+ HK + V, P < 0.001) and quadriceps muscle mass (T + G-treated Ghsr+/+ vs. Ghsr-/- : 96.12 ± 2.31% vs. 88.36 ± 1.94% of Ghsr+/+ HK + V, P = 0.01), and gastrocnemius type IIA (T + G-treated Ghsr+/+ vs. Ghsr-/- : 1250.49 ± 31.72 vs. 1017.62 ± 70.99 μm2 , P = 0.027) and IIB fibre cross-sectional area (T + G-treated Ghsr+/+ vs. Ghsr-/- : 2496.48 ± 116.88 vs. 2183.04 ± 103.43 μm2 , P = 0.024). CONCLUSIONS Growth hormone secretagogue receptor-1a mediates ghrelin's effects on attenuating LLC-induced weakness but not muscle mass loss by modulating the autophagy-lysosome pathway, mitophagy, and mitochondrial respiration.
Collapse
Affiliation(s)
- Haiming Liu
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Pu Zang
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
- Department of EndocrinologyNanjing Jinling HospitalNanjingChina
| | - Ian (In‐gi) Lee
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Barbara Anderson
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Anthony Christiani
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Lena Strait‐Bodey
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Beatrice A. Breckheimer
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Mackenzie Storie
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Alison Tewnion
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Kora Krumm
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Theresa Li
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Brynn Irwin
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Jose M. Garcia
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| |
Collapse
|
44
|
Mao X, Gu Y, Sui X, Shen L, Han J, Wang H, Xi Q, Zhuang Q, Meng Q, Wu G. Phosphorylation of Dynamin-Related Protein 1 (DRP1) Regulates Mitochondrial Dynamics and Skeletal Muscle Wasting in Cancer Cachexia. Front Cell Dev Biol 2021; 9:673618. [PMID: 34422804 PMCID: PMC8375307 DOI: 10.3389/fcell.2021.673618] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Background Cancer-associated cachexia (CAC) is a syndrome characterized by skeletal muscle atrophy, and the underlying mechanisms are still unclear. Recent research studies have shed light on a noteworthy link between mitochondrial dynamics and muscle physiology. In the present study, we investigate the role of dynamin-related protein 1 (DRP1), a pivotal factor of mitochondrial dynamics, in myotube atrophy during cancer-associated cachexia. Methods Seventy-six surgical patients, including gastrointestinal tumor and benign disease, were enrolled in the study and divided to three groups: control, non-cachexia, and cancer-associated cachexia. Demographic data were collected. Their rectus abdominis samples were acquired intraoperatively. Muscle fiber size, markers of ubiquitin proteasome system (UPS), mitochondrial ultrastructure, and markers of mitochondrial function and dynamics were assayed. A cachexia model in vitro was established via coculturing a C2C12 myotube with media from C26 colon cancer cells. A specific DRP1 inhibitor, Mdivi-1, and a lentivirus of DRP1 knockdown/overexpression were used to regulate the expression of DRP1. Muscle diameter, mitochondrial morphology, mass, reactive oxygen species (ROS), membrane potential, and markers of UPS, mitochondrial function, and dynamics were determined. Results Patients of cachexia suffered from a conspicuous worsened nutrition status and muscle loss compared to patients of other groups. Severe mitochondrial swelling and enlarged area were observed, and partial alterations in mitochondrial function were found in muscle. Analysis of mitochondrial dynamics indicated an upregulation of phosphorylated DRP1 at the ser616 site. In vitro, cancer media resulted in the atrophy of myotube. This was accompanied with a prominent unbalance of mitochondrial dynamics, as well as enhanced mitochondrial ROS and decreased mitochondrial function and membrane potential. However, certain concentrations of Mdivi-1 and DRP1 knockdown rebalanced the mitochondrial dynamics, mitigating this negative phenotype caused by cachexia. Moreover, overexpression of DRP1 aggravated these phenomena. Conclusion In clinical patients, cachexia induces abnormal mitochondrial changes and possible fission activation for the atrophied muscle. Our cachexia model in vitro further demonstrates that unbalanced mitochondrial dynamics contributes to this atrophy and mitochondrial impairment, and rebuilding the balance by regulating of DRP1 could ameliorate these alterations.
Collapse
Affiliation(s)
- Xiangyu Mao
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yihua Gu
- Shanghai Institute of Planned Parenthood Research, Shanghai, China
| | - Xiangyu Sui
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Shen
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun Han
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haiyu Wang
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiulei Xi
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiulin Zhuang
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qingyang Meng
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guohao Wu
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
45
|
Singh A, Yadav A, Phogat J, Dabur R. Dynamics of autophagy and ubiquitin proteasome system coordination and interplay in skeletal muscle atrophy. Curr Mol Pharmacol 2021; 15:475-486. [PMID: 34365963 DOI: 10.2174/1874467214666210806163851] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/26/2021] [Accepted: 04/05/2021] [Indexed: 11/22/2022]
Abstract
Skeletal muscles are considered the largest reservoirs of the protein pool in the body and are critical for the maintenances of body homeostasis. Skeletal muscle atrophy is supported by various physiopathological conditions that lead to loss of muscle mass and contractile capacity of the skeletal muscle. Lysosomal mediated autophagy and ubiquitin-proteasomal system (UPS) concede the major intracellular systems of muscle protein degradation that result in the loss of mass and strength. Both systems recognize ubiquitination as a signal of degradation through different mechanisms, a sign of dynamic interplay between systems. Hence, growing shreds of evidence suggest the interdependency of autophagy and UPS in the progression of skeletal muscle atrophy under various pathological conditions. Therefore, understanding the molecular dynamics as well associated factors responsible for their interdependency is a necessity for the new therapeutic insights to counteract the muscle loss. Based on current literature, the present review summarizes the factors interplay in between the autophagy and UPS in favor of enhanced proteolysis of skeletal muscle and how they affect the anabolic signaling pathways under various conditions of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Ajay Singh
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| | - Aarti Yadav
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| | - Jatin Phogat
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| | - Rajesh Dabur
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| |
Collapse
|
46
|
Renzini A, Riera CS, Minic I, D’Ercole C, Lozanoska-Ochser B, Cedola A, Gigli G, Moresi V, Madaro L. Metabolic Remodeling in Skeletal Muscle Atrophy as a Therapeutic Target. Metabolites 2021; 11:517. [PMID: 34436458 PMCID: PMC8398298 DOI: 10.3390/metabo11080517] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle is a highly responsive tissue, able to remodel its size and metabolism in response to external demand. Muscle fibers can vary from fast glycolytic to slow oxidative, and their frequency in a specific muscle is tightly regulated by fiber maturation, innervation, or external causes. Atrophic conditions, including aging, amyotrophic lateral sclerosis, and cancer-induced cachexia, differ in the causative factors and molecular signaling leading to muscle wasting; nevertheless, all of these conditions are characterized by metabolic remodeling, which contributes to the pathological progression of muscle atrophy. Here, we discuss how changes in muscle metabolism can be used as a therapeutic target and review the evidence in support of nutritional interventions and/or physical exercise as tools for counteracting muscle wasting in atrophic conditions.
Collapse
Affiliation(s)
- Alessandra Renzini
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
| | - Carles Sánchez Riera
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
| | - Isidora Minic
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
| | - Chiara D’Ercole
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
| | - Biliana Lozanoska-Ochser
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
| | - Alessia Cedola
- Institute of Nanotechnology, c/o Dipartimento di Fisica, National Research Council (CNR-NANOTEC), Sapienza University of Rome, 00185 Rome, Italy;
| | - Giuseppe Gigli
- Institute of Nanotechnology, c/o Campus Ecotekne, National Research Council (CNR-NANOTEC), Monteroni, 73100 Lecce, Italy;
| | - Viviana Moresi
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
- Institute of Nanotechnology, c/o Dipartimento di Fisica, National Research Council (CNR-NANOTEC), Sapienza University of Rome, 00185 Rome, Italy;
| | - Luca Madaro
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
| |
Collapse
|
47
|
Halle JL, Counts-Franch BR, Prince RM, Carson JA. The Effect of Mechanical Stretch on Myotube Growth Suppression by Colon-26 Tumor-Derived Factors. Front Cell Dev Biol 2021; 9:690452. [PMID: 34395422 PMCID: PMC8363303 DOI: 10.3389/fcell.2021.690452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022] Open
Abstract
Preclinical models and in vitro experiments have provided valuable insight into the regulation of cancer-induced muscle wasting. Colon-26 (C26) tumor cells induce cachexia in mice, and conditioned media (CM) from these cells promotes myotube atrophy and catabolic signaling. While mechanical stimuli can prevent some effects of tumor-derived factors on myotubes, the impact of mechanical signaling on tumor-derived factor regulation of myosin heavy chain (MyHC) expression is not well understood. Therefore, we examined the effects of stretch-induced mechanical signaling on C2C12 myotube growth and MyHC expression after C26 CM exposure. C26 CM was administered to myotubes on day 5 of differentiation for 48 h. During the last 4 or 24 h of C26 CM exposure, 5% static uniaxial stretch was administered. C26 CM suppressed myotube growth and MyHC protein and mRNA expression. Stretch for 24 h increased myotube size and prevented the C26 CM suppression of MyHC-Fast protein expression. Stretch did not change suppressed MyHC mRNA expression. Stretch for 24 h reduced Atrogin-1/MAFbx, MuRF-1, and LC3B II/I ratio and increased integrin β1D protein expression and the myogenin-to-MyoD protein ratio. Stretch in the last 4 h of CM increased ERK1/2 phosphorylation but did not alter the CM induction of STAT3 or p38 phosphorylation. These results provide evidence that in myotubes pre-incubated with CM, the induction of mechanical signaling can still provide a growth stimulus and preserve MyHC-Fast protein expression independent of changes in mRNA expression.
Collapse
Affiliation(s)
| | | | | | - James A. Carson
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
48
|
Khezri R, Holland P, Schoborg TA, Abramovich I, Takáts S, Dillard C, Jain A, O'Farrell F, Schultz SW, Hagopian WM, Quintana EM, Ng R, Katheder NS, Rahman MM, Teles Reis JG, Brech A, Jasper H, Rusan NM, Jahren AH, Gottlieb E, Rusten TE. Host autophagy mediates organ wasting and nutrient mobilization for tumor growth. EMBO J 2021; 40:e107336. [PMID: 34309071 PMCID: PMC8441431 DOI: 10.15252/embj.2020107336] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 05/14/2021] [Accepted: 06/01/2021] [Indexed: 11/15/2022] Open
Abstract
During tumor growth—when nutrient and anabolic demands are high—autophagy supports tumor metabolism and growth through lysosomal organelle turnover and nutrient recycling. Ras‐driven tumors additionally invoke non‐autonomous autophagy in the microenvironment to support tumor growth, in part through transfer of amino acids. Here we uncover a third critical role of autophagy in mediating systemic organ wasting and nutrient mobilization for tumor growth using a well‐characterized malignant tumor model in Drosophila melanogaster. Micro‐computed X‐ray tomography and metabolic profiling reveal that RasV12; scrib−/− tumors grow 10‐fold in volume, while systemic organ wasting unfolds with progressive muscle atrophy, loss of body mass, ‐motility, ‐feeding, and eventually death. Tissue wasting is found to be mediated by autophagy and results in host mobilization of amino acids and sugars into circulation. Natural abundance Carbon 13 tracing demonstrates that tumor biomass is increasingly derived from host tissues as a nutrient source as wasting progresses. We conclude that host autophagy mediates organ wasting and nutrient mobilization that is utilized for tumor growth.
Collapse
Affiliation(s)
- Rojyar Khezri
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Petter Holland
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Todd Andrew Schoborg
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ifat Abramovich
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Bat Galim, Haifa, Israel
| | - Szabolcs Takáts
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Caroline Dillard
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Ashish Jain
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Fergal O'Farrell
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Sebastian Wolfgang Schultz
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - William M Hagopian
- Centre for Earth Evolution and Dynamics, University of Oslo, Oslo, Norway
| | - Eduardo Martin Quintana
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Rachel Ng
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nadja Sandra Katheder
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Mohammed Mahidur Rahman
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - José Gerardo Teles Reis
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Andreas Brech
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Heinrich Jasper
- Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA.,Buck Institute for Research on Aging, Novato, CA, USA
| | - Nasser M Rusan
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anne Hope Jahren
- Centre for Earth Evolution and Dynamics, University of Oslo, Oslo, Norway
| | - Eyal Gottlieb
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Bat Galim, Haifa, Israel
| | - Tor Erik Rusten
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
49
|
Mora S, Adegoke OAJ. The effect of a chemotherapy drug cocktail on myotube morphology, myofibrillar protein abundance, and substrate availability. Physiol Rep 2021; 9:e14927. [PMID: 34197700 PMCID: PMC8248921 DOI: 10.14814/phy2.14927] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/01/2021] [Accepted: 05/10/2021] [Indexed: 12/03/2022] Open
Abstract
Cachexia, a condition prevalent in many chronically ill patients, is characterized by weight loss, fatigue, and decreases in muscle mass and function. Cachexia is associated with tumor burden and disease-related malnutrition, but other studies implicate chemotherapy as being causative. We investigated the effects of a chemotherapy drug cocktail on myofibrillar protein abundance and synthesis, anabolic signaling mechanisms, and substrate availability. On day 4 of differentiation, L6 myotubes were treated with vehicle (1.4 μl/ml DMSO) or a chemotherapy drug cocktail (a mixture of cisplatin [20 μg/ml], leucovorin [10 μg/ml], and 5-fluorouracil [5-FLU; 50 μg/ml]) for 24-72 h. Compared to myotubes treated with vehicle, those treated with the drug cocktail showed 50%-80% reductions in the abundance of myofibrillar proteins, including myosin heavy chain-1, troponin, and tropomyosin (p < 0.05). Cells treated with only a mixture of cisplatin and 5-FLU had identical reductions in myofibrillar protein abundance. Myotubes treated with the drug cocktail also showed >50% reductions in the phosphorylation of AKTSer473 and of mTORC1 substrates ribosomal protein S6Ser235/236 , its kinase S6K1Thr389 and eukaryotic translation initiation factor 4E-binding protein 1 (all p < 0.05). Drug treatment impaired peptide chain initiation in myofibrillar protein fractions and insulin-stimulated glucose uptake (p = 0.06) but increased the expression of autophagy markers beclin-1 and microtubule-associated proteins 1A/1B light chain 3B (p < 0.05), and of apoptotic marker, cleaved caspase 3 (p < 0.05). Drug treatment reduced the expression of mitochondrial markers cytochrome oxidase and succinate dehydrogenase (p < 0.05). The observed profound negative effects of this chemotherapy drug cocktail on myotubes underlie a need for approaches that can reduce the negative effects of these drugs on muscle metabolism.
Collapse
Affiliation(s)
- Stephen Mora
- School of Kinesiology and Health Science and Muscle Health Research CentreYork UniversityTorontoOntarioCanada
| | - Olasunkanmi A. J. Adegoke
- School of Kinesiology and Health Science and Muscle Health Research CentreYork UniversityTorontoOntarioCanada
| |
Collapse
|
50
|
Fix DK, Counts BR, Smuder AJ, Sarzynski MA, Koh H, Carson JA. Wheel running improves fasting-induced AMPK signaling in skeletal muscle from tumor-bearing mice. Physiol Rep 2021; 9:e14924. [PMID: 34270178 PMCID: PMC8284248 DOI: 10.14814/phy2.14924] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022] Open
Abstract
Disruptions to muscle protein turnover and metabolic regulation contribute to muscle wasting during the progression of cancer cachexia. The initiation of cachexia is also associated with decreased physical activity. While chronic muscle AMPK activation occurs during cachexia progression in ApcMin/+ (MIN) mice, a preclinical cachexia model, the understanding of muscle AMPK's role during cachexia initiation is incomplete. Therefore, we examined if voluntary wheel exercise could improve skeletal muscle AMPK signaling in pre-cachectic MIN mice. Next, we examined muscle AMPK's role in aberrant catabolic signaling in response to a 12-h fast in mice initiating cachexia. Male C57BL/6 (B6: N = 26) and MIN (N = 29) mice were subjected to ad libitum feeding, 12-h fast, or 4 wks. of wheel access and then a 12-h fast during the initiation of cachexia. Male tamoxifen-inducible skeletal muscle AMPKα1 α2 (KO) knockout mice crossed with ApcMin/+ and floxed controls were examined (WT: N = 8, KO: N = 8, MIN: N = 10, MIN KO: N = 6). Male mice underwent a 12-h fast and the gastrocnemius muscle was analyzed. MIN gastrocnemius mass was reduced compared to B6 mice. A 12-h fast induced MIN muscle AMPKT172 , FOXOS413 , and ULK-1S555 phosphorylation compared to B6. Wheel running attenuated these inductions. A 12-h fast induced MIN muscle MuRF-1 protein expression compared to B6 and was suppressed by wheel running. Additionally, fasting induced muscle autophagy signaling and disrupted mitochondrial quality protein expression in the MIN, which was prevented in the MIN KO. We provide evidence that increased skeletal muscle AMPK sensitivity to a 12-h fast is an adverse event in pre-cachectic MIN mice, and exercise can improve this regulation.
Collapse
Affiliation(s)
- Dennis K. Fix
- Department of Exercise ScienceArnold School of Public HealthUniversity of South CarolinaColumbiaSCUSA
| | - Brittany R. Counts
- Integrative Muscle Biology LaboratoryDivision of Rehabilitation SciencesCollege of Health ProfessionsUniversity of Tennessee Health Science CenterMemphisTNUSA
| | - Ashley J. Smuder
- Department of Applied Physiology & KinesiologyCollege of Health & Human PerformanceUniversity of FloridaGainesvilleFLUSA
| | - Mark A. Sarzynski
- Department of Exercise ScienceArnold School of Public HealthUniversity of South CarolinaColumbiaSCUSA
| | - Ho‐Jin Koh
- Department of Exercise ScienceArnold School of Public HealthUniversity of South CarolinaColumbiaSCUSA
| | - James A. Carson
- Integrative Muscle Biology LaboratoryDivision of Rehabilitation SciencesCollege of Health ProfessionsUniversity of Tennessee Health Science CenterMemphisTNUSA
| |
Collapse
|