1
|
Polat K, Karadibak D, Güç ZGS, Yavuzşen T, Öztop İ. The Relationship between Exercise Capacity and Muscle Strength, Physical Activity, Fatigue and Quality of Life in Patients with Cancer Cachexia. Nutr Cancer 2023; 76:55-62. [PMID: 37917566 DOI: 10.1080/01635581.2023.2276486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Exercise capacity is a significant determinant of mortality for cancer patients, so knowing the possible determinants of exercise capacity will produce physical and psychological benefits for individuals with cancer cachexia. PURPOSE To investigate the relationship between exercise capacity on peripheric and respiratory muscle strength, physical activity, fatigue and quality of life in subjects with cancer cachexia. METHODS The study included 31 patients diagnosed with cancer cachexia. Functional capacity was assessed by 6-Minute Walk Test, hand grip strength and proximal muscle mass by hand dynamometer, respiratory muscle strength by the Maximum Expiratory Pressure and Maximum Inspiratory Pressure measurements, physical activity by International Physical Activity Questionnaire Short Form, fatigue by Brief Fatigue Inventory, and quality of life by EORT-QLQ-C30. The relationship between functional capacity and continuous independent variables was determined using Spearman's or Pearson's tests. RESULTS A strong positive correlation was observed between exercise capacity and expiratory muscle strength (r = 0.75, p < 0.001), activity level (r = 0.68, p < 0.001), and quality of life global health status (r = 0.74, p < 0.001). Conversely, a strong negative correlation was found between exercise capacity and fatigue severity (r = -0.64, p < 0.001). CONCLUSION Higher exercise capacity in cancer cachexia patients is linked to reduced fatigue, improved respiratory muscle strength, increased physical activity levels, and enhanced quality of life. When designing rehabilitation programs or exercise interventions for individuals with cancer cachexia, it is crucial to assess their exercise capacity and tailor the programs accordingly.
Collapse
Affiliation(s)
- Karya Polat
- Health Science Institute, Katip Celebi University, İzmir, Turkey
| | - Didem Karadibak
- School of Physical Therapy and Rehabilitation, Dokuz Eylul University, İzmir, Turkey
| | | | - Tuğba Yavuzşen
- Medicine Faculty Medical Oncology Subdivision, Dokuz Eylul University, İzmir, Turkey
| | - İlhan Öztop
- Medicine Faculty Medical Oncology Subdivision, Dokuz Eylul University, İzmir, Turkey
| |
Collapse
|
2
|
Sakuma K, Hamada K, Yamaguchi A, Aoi W. Current Nutritional and Pharmacological Approaches for Attenuating Sarcopenia. Cells 2023; 12:2422. [PMID: 37830636 PMCID: PMC10572610 DOI: 10.3390/cells12192422] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/27/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023] Open
Abstract
Sarcopenia is characterized by a gradual slowing of movement due to loss of muscle mass and quality, decreased power and strength, increased risk of injury from falls, and often weakness. This review will focus on recent research trends in nutritional and pharmacological approaches to controlling sarcopenia. Because nutritional studies in humans are fairly limited, this paper includes many results from nutritional studies in mammals. The combination of resistance training with supplements containing amino acids is the gold standard for preventing sarcopenia. Amino acid (HMB) supplementation alone has no significant effect on muscle strength or muscle mass in sarcopenia, but the combination of HMB and exercise (whole body vibration stimulation) is likely to be effective. Tea catechins, soy isoflavones, and ursolic acid are interesting candidates for reducing sarcopenia, but both more detailed basic research on this treatment and clinical studies in humans are needed. Vitamin D supplementation has been shown not to improve sarcopenia in elderly individuals who are not vitamin D-deficient. Myostatin inhibitory drugs have been tried in many neuromuscular diseases, but increases in muscle mass and strength are less likely to be expected. Validation of myostatin inhibitory antibodies in patients with sarcopenia has been positive, but excessive expectations are not warranted.
Collapse
Affiliation(s)
- Kunihiro Sakuma
- Institute for Liberal Arts, Environment and Society, Tokyo Institute of Technology, Meguro-ku, Tokyo 152-8550, Japan;
| | - Kento Hamada
- Institute for Liberal Arts, Environment and Society, Tokyo Institute of Technology, Meguro-ku, Tokyo 152-8550, Japan;
| | - Akihiko Yamaguchi
- Department of Physical Therapy, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan;
| | - Wataru Aoi
- Laboratory of Nutrition Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan;
| |
Collapse
|
3
|
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: 16] [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
|
4
|
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: 4.0] [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
|
5
|
Henrot P, Dupin I, Schilfarth P, Esteves P, Blervaque L, Zysman M, Gouzi F, Hayot M, Pomiès P, Berger P. Main Pathogenic Mechanisms and Recent Advances in COPD Peripheral Skeletal Muscle Wasting. Int J Mol Sci 2023; 24:ijms24076454. [PMID: 37047427 PMCID: PMC10095391 DOI: 10.3390/ijms24076454] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a worldwide prevalent respiratory disease mainly caused by tobacco smoke exposure. COPD is now considered as a systemic disease with several comorbidities. Among them, skeletal muscle dysfunction affects around 20% of COPD patients and is associated with higher morbidity and mortality. Although the histological alterations are well characterized, including myofiber atrophy, a decreased proportion of slow-twitch myofibers, and a decreased capillarization and oxidative phosphorylation capacity, the molecular basis for muscle atrophy is complex and remains partly unknown. Major difficulties lie in patient heterogeneity, accessing patients' samples, and complex multifactorial process including extrinsic mechanisms, such as tobacco smoke or disuse, and intrinsic mechanisms, such as oxidative stress, hypoxia, or systemic inflammation. Muscle wasting is also a highly dynamic process whose investigation is hampered by the differential protein regulation according to the stage of atrophy. In this review, we report and discuss recent data regarding the molecular alterations in COPD leading to impaired muscle mass, including inflammation, hypoxia and hypercapnia, mitochondrial dysfunction, diverse metabolic changes such as oxidative and nitrosative stress and genetic and epigenetic modifications, all leading to an impaired anabolic/catabolic balance in the myocyte. We recapitulate data concerning skeletal muscle dysfunction obtained in the different rodent models of COPD. Finally, we propose several pathways that should be investigated in COPD skeletal muscle dysfunction in the future.
Collapse
Affiliation(s)
- Pauline Henrot
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
- CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC 1401, Service de Pneumologie, F-33604 Pessac, France
| | - Isabelle Dupin
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
| | - Pierre Schilfarth
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
- CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC 1401, Service de Pneumologie, F-33604 Pessac, France
| | - Pauline Esteves
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
| | - Léo Blervaque
- PhyMedExp, INSERM-CNRS-Montpellier University, F-34090 Montpellier, France
| | - Maéva Zysman
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
- CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC 1401, Service de Pneumologie, F-33604 Pessac, France
| | - Fares Gouzi
- PhyMedExp, INSERM-CNRS-Montpellier University, CHRU Montpellier, F-34090 Montpellier, France
| | - Maurice Hayot
- PhyMedExp, INSERM-CNRS-Montpellier University, CHRU Montpellier, F-34090 Montpellier, France
| | - Pascal Pomiès
- PhyMedExp, INSERM-CNRS-Montpellier University, F-34090 Montpellier, France
| | - Patrick Berger
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
- CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC 1401, Service de Pneumologie, F-33604 Pessac, France
| |
Collapse
|
6
|
The Inhibition of Autophagy and Pyroptosis by an Ethanol Extract of Nelumbo nucifera Leaf Contributes to the Amelioration of Dexamethasone-Induced Muscle Atrophy. Nutrients 2023; 15:nu15040804. [PMID: 36839161 PMCID: PMC9965294 DOI: 10.3390/nu15040804] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Muscle atrophy is characterized by a decline in muscle mass and function. Excessive glucocorticoids in the body due to aging or drug treatment can promote muscle wasting. In this study, we investigated the preventive effect of Nelumbo nucifera leaf (NNL) ethanolic extract on muscle atrophy induced by dexamethasone (DEX), a synthetic glucocorticoid, in mice and its underlying mechanisms. The administration of NNL extract increased weight, cross-sectional area, and grip strength of quadriceps (QD) and gastrocnemius (GA) muscles in DEX-induced muscle atrophy in mice. The NNL extract administration decreased the expression of muscle atrophic factors, such as muscle RING-finger protein-1 and atrogin-1, and autophagy factors, such as Beclin-1, microtubule-associated protein 1A/1B-light chain 3 (LC3-I/II), and sequestosome 1 (p62/SQSTM1) in DEX-injected mice. DEX injection increased the protein expression levels of NOD-like receptor pyrin domain-containing protein 3 (NLRP3), cleaved-caspase-1, interleukin-1beta (IL-1β), and cleaved-gasdermin D (GSDMD), which were significantly reduced by NNL extract administration (500 mg/kg/day). In vitro studies using C2C12 myotubes also revealed that NNL extract treatment inhibited the DEX-induced increase in autophagy factors, pyroptosis-related factors, and NF-κB. Overall, the NNL extract prevented DEX-induced muscle atrophy by downregulating the ubiquitin-proteasome system, autophagy pathway, and GSDMD-mediated pyroptosis pathway, which are involved in muscle degradation.
Collapse
|
7
|
Murphy BT, Mackrill JJ, O'Halloran KD. Impact of cancer cachexia on respiratory muscle function and the therapeutic potential of exercise. J Physiol 2022; 600:4979-5004. [PMID: 36251564 PMCID: PMC10091733 DOI: 10.1113/jp283569] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/09/2022] [Indexed: 01/05/2023] Open
Abstract
Cancer cachexia is defined as a multi-factorial syndrome characterised by an ongoing loss of skeletal muscle mass and progressive functional impairment, estimated to affect 50-80% of patients and responsible for 20% of cancer deaths. Elevations in the morbidity and mortality rates of cachectic cancer patients has been linked to respiratory failure due to atrophy and dysfunction of the ventilatory muscles. Despite this, there is a distinct scarcity of research investigating the structural and functional condition of the respiratory musculature in cancer, with the majority of studies exclusively focusing on limb muscle. Treatment strategies are largely ineffective in mitigating the cachectic state. It is now widely accepted that an efficacious intervention will likely combine elements of pharmacology, nutrition and exercise. However, of these approaches, exercise has received comparatively little attention. Therefore, it is unlikely to be implemented optimally, whether in isolation or combination. In consideration of these limitations, the current review describes the mechanistic basis of cancer cachexia and subsequently explores the available respiratory- and exercise-focused literature within this context. The molecular basis of cachexia is thoroughly reviewed. The pivotal role of inflammatory mediators is described. Unravelling the mechanisms of exercise-induced support of muscle via antioxidant and anti-inflammatory effects in addition to promoting efficient energy metabolism via increased mitochondrial biogenesis, mitochondrial function and muscle glucose uptake provide avenues for interventional studies. Currently available pre-clinical mouse models including novel transgenic animals provide a platform for the development of multi-modal therapeutic strategies to protect respiratory muscles in people with cancer.
Collapse
Affiliation(s)
- Ben T. Murphy
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| | - John J. Mackrill
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| | - Ken D. O'Halloran
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| |
Collapse
|
8
|
Cancer Cachexia: Signaling and Transcriptional Regulation of Muscle Catabolic Genes. Cancers (Basel) 2022; 14:cancers14174258. [PMID: 36077789 PMCID: PMC9454911 DOI: 10.3390/cancers14174258] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 02/08/2023] Open
Abstract
Simple Summary An uncontrollable loss in the skeletal muscle of cancer patients which leads to a significant reduction in body weight is clinically referred to as cancer cachexia (CC). While factors derived from the tumor environment which trigger various signaling pathways have been identified, not much progress has been made clinically to effectively prevent muscle loss. Deeper insights into the transcriptional and epigenetic regulation of muscle catabolic genes may shed light on key regulators which can be targeted to develop new therapeutic avenues. Abstract Cancer cachexia (CC) is a multifactorial syndrome characterized by a significant reduction in body weight that is predominantly caused by the loss of skeletal muscle and adipose tissue. Although the ill effects of cachexia are well known, the condition has been largely overlooked, in part due to its complex etiology, heterogeneity in mediators, and the involvement of diverse signaling pathways. For a long time, inflammatory factors have been the focus when developing therapeutics for the treatment of CC. Despite promising pre-clinical results, they have not yet advanced to the clinic. Developing new therapies requires a comprehensive understanding of how deregulated signaling leads to catabolic gene expression that underlies muscle wasting. Here, we review CC-associated signaling pathways and the transcriptional cascade triggered by inflammatory cytokines. Further, we highlight epigenetic factors involved in the transcription of catabolic genes in muscle wasting. We conclude with reflections on the directions that might pave the way for new therapeutic approaches to treat CC.
Collapse
|
9
|
Graca FA, Rai M, Hunt LC, Stephan A, Wang YD, Gordon B, Wang R, Quarato G, Xu B, Fan Y, Labelle M, Demontis F. The myokine Fibcd1 is an endogenous determinant of myofiber size and mitigates cancer-induced myofiber atrophy. Nat Commun 2022; 13:2370. [PMID: 35501350 PMCID: PMC9061726 DOI: 10.1038/s41467-022-30120-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [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: 04/14/2022] [Indexed: 12/19/2022] Open
Abstract
Decline in skeletal muscle cell size (myofiber atrophy) is a key feature of cancer-induced wasting (cachexia). In particular, atrophy of the diaphragm, the major muscle responsible for breathing, is an important determinant of cancer-associated mortality. However, therapeutic options are limited. Here, we have used Drosophila transgenic screening to identify muscle-secreted factors (myokines) that act as paracrine regulators of myofiber growth. Subsequent testing in mouse myotubes revealed that mouse Fibcd1 is an evolutionary-conserved myokine that preserves myofiber size via ERK signaling. Local administration of recombinant Fibcd1 (rFibcd1) ameliorates cachexia-induced myofiber atrophy in the diaphragm of mice bearing patient-derived melanoma xenografts and LLC carcinomas. Moreover, rFibcd1 impedes cachexia-associated transcriptional changes in the diaphragm. Fibcd1-induced signaling appears to be muscle selective because rFibcd1 increases ERK activity in myotubes but not in several cancer cell lines tested. We propose that rFibcd1 may help reinstate myofiber size in the diaphragm of patients with cancer cachexia.
Collapse
Affiliation(s)
- Flavia A Graca
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Mamta Rai
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Liam C Hunt
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Anna Stephan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Yong-Dong Wang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Brittney Gordon
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
- Xenograft Core, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Ruishan Wang
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Giovanni Quarato
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Beisi Xu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Yiping Fan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Myriam Labelle
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States.
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States.
| |
Collapse
|
10
|
Talbert EE, Guttridge DC. Emerging signaling mediators in the anorexia-cachexia syndrome of cancer. Trends Cancer 2022; 8:397-403. [PMID: 35190301 PMCID: PMC9035074 DOI: 10.1016/j.trecan.2022.01.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 12/11/2022]
Abstract
The cachexia syndrome in cancer is characterized by weight loss resulting from the combination of anorexia and atrophy of adipose and skeletal muscle. For decades, inflammatory circulatory factors have been identified to regulate wasting, but inhibitors of these factors have not yielded the same clinical benefit as in animal models. Therefore, additional mediators of cachexia likely regulate this syndrome, and such factors might be more suitable for targeted intervention. We highlight several anorexia-cachexia signaling mediators, including activin A, myostatin, GDF15, and lipocalin-2. We discuss current evidence that these factors associate with cachexia in cancer patients, and summarize translational efforts including essential early-phase clinical trials. We conclude with thoughts on targeted and personalized approaches for future anti-cachexia treatments.
Collapse
Affiliation(s)
- Erin E Talbert
- Department of Health and Human Physiology, and the Holden Comprehensive Cancer Center, University Iowa, Iowa City, IA 52242, USA
| | - Denis C Guttridge
- Department of Pediatrics, Darby Children's Research Institute, and the Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
11
|
Hillege MMG, Shi A, Galli RA, Wu G, Bertolino P, Hoogaars WMH, Jaspers RT. Lack of Tgfbr1 and Acvr1b synergistically stimulates myofibre hypertrophy and accelerates muscle regeneration. eLife 2022; 11:77610. [PMID: 35323108 PMCID: PMC9005187 DOI: 10.7554/elife.77610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/05/2022] [Indexed: 12/02/2022] Open
Abstract
In skeletal muscle, transforming growth factor-β (TGF-β) family growth factors, TGF-β1 and myostatin, are involved in atrophy and muscle wasting disorders. Simultaneous interference with their signalling pathways may improve muscle function; however, little is known about their individual and combined receptor signalling. Here, we show that inhibition of TGF-β signalling by simultaneous muscle-specific knockout of TGF-β type I receptors Tgfbr1 and Acvr1b in mice, induces substantial hypertrophy, while such effect does not occur by single receptor knockout. Hypertrophy is induced by increased phosphorylation of Akt and p70S6K and reduced E3 ligases expression, while myonuclear number remains unaltered. Combined knockout of both TGF-β type I receptors increases the number of satellite cells, macrophages and improves regeneration post cardiotoxin-induced injury by stimulating myogenic differentiation. Extra cellular matrix gene expression is exclusively elevated in muscle with combined receptor knockout. Tgfbr1 and Acvr1b are synergistically involved in regulation of myofibre size, regeneration, and collagen deposition.
Collapse
Affiliation(s)
- Michèle M G Hillege
- Department of Human Movement, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Andi Shi
- Department of Human Movement, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ricardo A Galli
- Department of Human Movement, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Philippe Bertolino
- Centre de Recherche en Cancérologie de Lyon, Université de Lyon, UMR INSERM U1052, CNRS 5286, Lyon, France
| | - Willem M H Hoogaars
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Richard T Jaspers
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
12
|
Rodgers BD, Ward CW. Myostatin/Activin Receptor Ligands in Muscle and the Development Status of Attenuating Drugs. Endocr Rev 2022; 43:329-365. [PMID: 34520530 PMCID: PMC8905337 DOI: 10.1210/endrev/bnab030] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Indexed: 02/07/2023]
Abstract
Muscle wasting disease indications are among the most debilitating and often deadly noncommunicable disease states. As a comorbidity, muscle wasting is associated with different neuromuscular diseases and myopathies, cancer, heart failure, chronic pulmonary and renal diseases, peripheral neuropathies, inflammatory disorders, and, of course, musculoskeletal injuries. Current treatment strategies are relatively ineffective and can at best only limit the rate of muscle degeneration. This includes nutritional supplementation and appetite stimulants as well as immunosuppressants capable of exacerbating muscle loss. Arguably, the most promising treatments in development attempt to disrupt myostatin and activin receptor signaling because these circulating factors are potent inhibitors of muscle growth and regulators of muscle progenitor cell differentiation. Indeed, several studies demonstrated the clinical potential of "inhibiting the inhibitors," increasing muscle cell protein synthesis, decreasing degradation, enhancing mitochondrial biogenesis, and preserving muscle function. Such changes can prevent muscle wasting in various disease animal models yet many drugs targeting this pathway failed during clinical trials, some from serious treatment-related adverse events and off-target interactions. More often, however, failures resulted from the inability to improve muscle function despite preserving muscle mass. Drugs still in development include antibodies and gene therapeutics, all with different targets and thus, safety, efficacy, and proposed use profiles. Each is unique in design and, if successful, could revolutionize the treatment of both acute and chronic muscle wasting. They could also be used in combination with other developing therapeutics for related muscle pathologies or even metabolic diseases.
Collapse
Affiliation(s)
| | - Christopher W Ward
- Department of Orthopedics and Center for Biomedical Engineering and Technology (BioMET), University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
13
|
Li J, Xie Q, Liu L, Cheng Y, Han Y, Chen X, Lin J, Li Z, Liu H, Zhang X, Chen H, Peng J, Shen A. Swimming Attenuates Muscle Wasting and Mediates Multiple Signaling Pathways and Metabolites in CT-26 Bearing Mice. Front Mol Biosci 2022; 8:812681. [PMID: 35127824 PMCID: PMC8811507 DOI: 10.3389/fmolb.2021.812681] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Objectives: To investigate the effects of swimming on cancer induced muscle wasting and explore its underlying mechanism in CT-26 bearing mice.Methods: BALB/c mice (n = 16) injected with CT-26 cells were divided into two groups, including Tumor group (n = 8) and Swimming group (n = 8). Another 8 un-injected mice were set as Control group. Mice in Swimming group were subjected to physical training for swimming twice per day for 30 min intervals and 6 days per week for a total of 4 weeks. The tumor volume was monitored every 3 days and tumor weight was measured at the end of experiment. The changes of muscle function, pathological and cell apoptosis of quadriceps muscles were further assessed, and its underlying mechanisms were further explored using multiple biological technologies.Results: Swimming obviously alleviated tumor volume and weight in CT-26 bearing mice. Moreover, swimming attenuated the decrease of muscle tension, autonomic activities, and increase of muscle atrophy, pathological ultrastructure, as well as cell apoptosis of quadriceps muscles in CT-26 bearing mice. Furthermore, swimming significantly down-regulated the protein expression of NF-κB, p-NF-κB, TNF-α, IL-1β, IL-6 and Bax, while up-regulated the expression of Bcl-2. Further differential expressed metabolites (DEMs) analysis identified a total of 76 (in anion mode) and 330 (in cationic mode) DEMs in quadriceps muscles of CT-26 bearing mice after swimming, including taurochenodeoxycholic acid, taurocholic acid, ascorbic acid and eicosapentaenoic acid.Conclusion: Swimming attenuates tumor growth and muscle wasting, and by suppressing the activation of NF-κB signaling pathway mediated inflammation, reducing the level of Bax medicated cell apoptosis, as well as modulating multiple metabolites might be the importantly underlying mechanisms.
Collapse
Affiliation(s)
- Jiapeng Li
- The Department of Physical Education, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- School of Physical Education and Sport Science, Fujian Normal University, Fuzhou, China
| | - Qiurong Xie
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Liya Liu
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Ying Cheng
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Yuying Han
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Xiaoping Chen
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Jia Lin
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zuanfang Li
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Huixin Liu
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Xiuli Zhang
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Haichun Chen
- School of Physical Education and Sport Science, Fujian Normal University, Fuzhou, China
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
| | - Jun Peng
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
- *Correspondence: Jun Peng, ; Aling Shen,
| | - Aling Shen
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
- *Correspondence: Jun Peng, ; Aling Shen,
| |
Collapse
|
14
|
Counts BR, Halle JL, Carson JA. Early-Onset Physical Inactivity and Metabolic Dysfunction in Tumor-bearing Mice Is Associated with Accelerated Cachexia. Med Sci Sports Exerc 2022; 54:77-88. [PMID: 34431825 PMCID: PMC8678203 DOI: 10.1249/mss.0000000000002772] [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] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
METHODS Male C57BL/6J mice (12 wk of age) were injected with 1 × 106 LLC cells or phosphate-buffered saline (PBS) subcutaneously in the right flank, and tissue was collected 26-28 d after cell injection. Tumor volume was measured every 5 d throughout the study to calculate the tumor growth rate. Fifteen days after tumor inoculation, a subset of PBS (n = 11) and LLC (n = 16) mice were individually housed in metabolic Comprehensive Laboratory Animal Monitoring System cages for 5 d. RESULTS LLC mice exhibited greater body weight loss (-5.1%), decreased muscle mass (-7%), decreased fat mass (-22%), and increased plasma interleukin-6 (212%) compared with PBS mice. Before the onset of cachexia, total cage activity was decreased in tumor-bearing mice. Cage activity was negatively associated with tumor mass and positively associated with hindlimb muscle mass. In addition, LLC mice had greater lipid oxidation than PBS mice. CONCLUSIONS LLC mice exhibit early-onset physical inactivity and altered systemic lipid oxidation, which are associated with the eventual development of cachexia.
Collapse
Affiliation(s)
- Brittany R Counts
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis TN
| | | | | |
Collapse
|
15
|
Esposito P, Picciotto D, Battaglia Y, Costigliolo F, Viazzi F, Verzola D. Myostatin: Basic biology to clinical application. Adv Clin Chem 2022; 106:181-234. [PMID: 35152972 DOI: 10.1016/bs.acc.2021.09.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [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
|
16
|
Martin A, Freyssenet D. Phenotypic features of cancer cachexia-related loss of skeletal muscle mass and function: lessons from human and animal studies. J Cachexia Sarcopenia Muscle 2021; 12:252-273. [PMID: 33783983 PMCID: PMC8061402 DOI: 10.1002/jcsm.12678] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/22/2020] [Accepted: 12/30/2020] [Indexed: 12/18/2022] Open
Abstract
Cancer cachexia is a complex multi-organ catabolic syndrome that reduces mobility, increases fatigue, decreases the efficiency of therapeutic strategies, diminishes the quality of life, and increases the mortality of cancer patients. This review provides an exhaustive and comprehensive analysis of cancer cachexia-related phenotypic changes in skeletal muscle at both the cellular and subcellular levels in human cancer patients, as well as in animal models of cancer cachexia. Cancer cachexia is characterized by a major decrease in skeletal muscle mass in human and animals that depends on the severity of the disease/model and the localization of the tumour. It affects both type 1 and type 2 muscle fibres, even if some animal studies suggest that type 2 muscle fibres would be more prone to atrophy. Animal studies indicate an impairment in mitochondrial oxidative metabolism resulting from a decrease in mitochondrial content, an alteration in mitochondria morphology, and a reduction in mitochondrial metabolic fluxes. Immuno-histological analyses in human and animal models also suggest that a faulty mechanism of skeletal muscle repair would contribute to muscle mass loss. An increase in collagen deposit, an accumulation of fat depot outside and inside the muscle fibre, and a disrupted contractile machinery structure are also phenotypic features that have been consistently reported in cachectic skeletal muscle. Muscle function is also profoundly altered during cancer cachexia with a strong reduction in skeletal muscle force. Even though the loss of skeletal muscle mass largely contributes to the loss of muscle function, other factors such as muscle-nerve interaction and calcium handling are probably involved in the decrease in muscle force. Longitudinal analyses of skeletal muscle mass by imaging technics and skeletal muscle force in cancer patients, but also in animal models of cancer cachexia, are necessary to determine the respective kinetics and functional involvements of these factors. Our analysis also emphasizes that measuring skeletal muscle force through standardized tests could provide a simple and robust mean to early diagnose cachexia in cancer patients. That would be of great benefit to cancer patient's quality of life and health care systems.
Collapse
Affiliation(s)
- Agnès Martin
- Inter‐university Laboratory of Human Movement BiologyUniversité de Lyon, University Jean Monnet Saint‐EtienneSaint‐ÉtienneFrance
| | - Damien Freyssenet
- Inter‐university Laboratory of Human Movement BiologyUniversité de Lyon, University Jean Monnet Saint‐EtienneSaint‐ÉtienneFrance
| |
Collapse
|
17
|
Invernizzi M, de Sire A, Fusco N. Rethinking the clinical management of volumetric muscle loss in patients with spinal cord injury: Synergy among nutritional supplementation, pharmacotherapy, and rehabilitation. Curr Opin Pharmacol 2021; 57:132-139. [PMID: 33721616 DOI: 10.1016/j.coph.2021.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/30/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022]
Abstract
Spinal cord injury (SCI) is a condition defining the damage of the spinal cord that leads to musculoskeletal sequelae, including volumetric muscle loss (VML) in a significant proportion of patients. VML occurring after SCI is responsible for delayed recovery, with detrimental consequences in terms of functional outcomes and additional alterations of the muscle tissue. The treatment of muscle alterations in these patients usually relies on nutritional supplementation. However, rehabilitation therapy has a well-recognized role in improving muscle mass and function, even in subjects affected by SCI. Furthermore, novel medical therapies have been recently investigated, with positive results. In this scoping review, we portray the state-of-the-art treatment of muscle modifications after SCI, focusing on the multidisciplinary and multidimensional management of these patients.
Collapse
Affiliation(s)
- Marco Invernizzi
- Physical and Rehabilitative Medicine, Department of Health Sciences, University of Eastern Piedmont, Novara, Italy; Infrastruttura Ricerca Formazione Innovazione (IRFI), Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Alessandro de Sire
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Graecia", Catanzaro, Italy.
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
18
|
Antimyostatin Treatment in Health and Disease: The Story of Great Expectations and Limited Success. Cells 2021; 10:cells10030533. [PMID: 33802348 PMCID: PMC8001237 DOI: 10.3390/cells10030533] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 12/14/2022] Open
Abstract
In the past 20 years, myostatin, a negative regulator of muscle mass, has attracted attention as a potential therapeutic target in muscular dystrophies and other conditions. Preclinical studies have shown potential for increasing muscular mass and ameliorating the pathological features of dystrophic muscle by the inhibition of myostatin in various ways. However, hardly any clinical trials have proven to translate the promising results from the animal models into patient populations. We present the background for myostatin regulation, clinical and preclinical results and discuss why translation from animal models to patients is difficult. Based on this, we put the clinical relevance of future antimyostatin treatment into perspective.
Collapse
|
19
|
Targeting the Activin Receptor Signaling to Counteract the Multi-Systemic Complications of Cancer and Its Treatments. Cells 2021; 10:cells10030516. [PMID: 33671024 PMCID: PMC7997313 DOI: 10.3390/cells10030516] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Muscle wasting, i.e., cachexia, frequently occurs in cancer and associates with poor prognosis and increased morbidity and mortality. Anticancer treatments have also been shown to contribute to sustainment or exacerbation of cachexia, thus affecting quality of life and overall survival in cancer patients. Pre-clinical studies have shown that blocking activin receptor type 2 (ACVR2) or its ligands and their downstream signaling can preserve muscle mass in rodents bearing experimental cancers, as well as in chemotherapy-treated animals. In tumor-bearing mice, the prevention of skeletal and respiratory muscle wasting was also associated with improved survival. However, the definitive proof that improved survival directly results from muscle preservation following blockade of ACVR2 signaling is still lacking, especially considering that concurrent beneficial effects in organs other than skeletal muscle have also been described in the presence of cancer or following chemotherapy treatments paired with counteraction of ACVR2 signaling. Hence, here, we aim to provide an up-to-date literature review on the multifaceted anti-cachectic effects of ACVR2 blockade in preclinical models of cancer, as well as in combination with anticancer treatments.
Collapse
|
20
|
Rosa-Caldwell ME, Benson CA, Lee DE, Brown JL, Washington TA, Greene NP, Wiggs MP. Mitochondrial Function and Protein Turnover in the Diaphragm are Altered in LLC Tumor Model of Cancer Cachexia. Int J Mol Sci 2020; 21:E7841. [PMID: 33105841 PMCID: PMC7660065 DOI: 10.3390/ijms21217841] [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: 09/24/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023] Open
Abstract
It is established that cancer cachexia causes limb muscle atrophy and is strongly associated with morbidity and mortality; less is known about how the development of cachexia impacts the diaphragm. The purpose of this study was to investigate cellular signaling mechanisms related to mitochondrial function, reactive oxygen species (ROS) production, and protein synthesis during the development of cancer cachexia. C57BL/J6 mice developed Lewis Lung Carcinoma for either 0 weeks (Control), 1 week, 2 weeks, 3 weeks, or 4 weeks. At designated time points, diaphragms were harvested and analyzed. Mitochondrial respiratory control ratio was ~50% lower in experimental groups, which was significant by 2 weeks of cancer development, with no difference in mitochondrial content markers COXIV or VDAC. Compared to the controls, ROS was 4-fold elevated in 2-week animals but then was not different at later time points. Only one antioxidant protein, GPX3, was altered by cancer development (~70% lower in experimental groups). Protein synthesis, measured by a fractional synthesis rate, appeared to become progressively lower with the cancer duration, but the mean difference was not significant. The development and progression of cancer cachexia induces marked alterations to mitochondrial function and ROS production in the diaphragm and may contribute to increased cachexia-associated morbidity and mortality.
Collapse
Affiliation(s)
- Megan E. Rosa-Caldwell
- Exercise Science Research Center, Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, USA; (M.E.R.-C.); (D.E.L.); (J.L.B.); (N.P.G.)
| | - Conner A. Benson
- Integrative Physiology and Nutrition Laboratory Name, Department of Health and Kinesiology, University of Texas at Tyler, Tyler, TX 75799, USA;
| | - David E. Lee
- Exercise Science Research Center, Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, USA; (M.E.R.-C.); (D.E.L.); (J.L.B.); (N.P.G.)
| | - Jacob L. Brown
- Exercise Science Research Center, Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, USA; (M.E.R.-C.); (D.E.L.); (J.L.B.); (N.P.G.)
| | - Tyrone A. Washington
- Exercise Science Research Center, Exercise Muscle Biology Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Nicholas P. Greene
- Exercise Science Research Center, Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, USA; (M.E.R.-C.); (D.E.L.); (J.L.B.); (N.P.G.)
| | - Michael P. Wiggs
- Integrative Physiology and Nutrition Laboratory Name, Department of Health and Kinesiology, University of Texas at Tyler, Tyler, TX 75799, USA;
- Department of Health, Human Performance and Recreation, Baylor University, Waco, TX 76798, USA
| |
Collapse
|
21
|
Christian CJ, Benian GM. Animal models of sarcopenia. Aging Cell 2020; 19:e13223. [PMID: 32857472 PMCID: PMC7576270 DOI: 10.1111/acel.13223] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/23/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Sarcopenia is the age-related decline in muscle mass and function without any underlying disease. The exact molecular mechanisms responsible for this pathology remain unknown. The use of model organisms, such as mice, rats, flies, and worms, has advanced the field of sarcopenia research by identifying therapeutic strategies and genetic mutations that result in improved muscle mass and function of elderly animals. This review discusses molecular and therapeutic discoveries made using these model organisms and how these animals can be further utilized to better understand sarcopenia pathogenesis. In rodents, flies, and worms, dietary restriction improves muscle performance in old animals. In rodents and worms, exercise and a number of naturally occurring compounds alleviate sarcopenia. Reduction in the insulin/IGF1 receptor pathway, well known to promote longevity, improves sarcopenia in worms and flies. Mitochondrial dysfunction may contribute to the pathogenesis of sarcopenia: In rodents, there is age-dependent reduction in mitochondrial mass and a change in morphology; in nematodes, there is age-dependent fragmentation of mitochondria that precedes sarcomeric disorganization. In Drosophila and rats, components of the 26S proteasome are elevated in aged muscle. An advantage of the worm and fly models is that these organisms lack muscle stem cells, and thus processes that promote the maintenance of already assembled muscle, can be identified without the confounding influence of muscle regeneration. Zebrafish are an up and coming model of sarcopenia for future consideration. A better understanding of the molecular changes behind sarcopenia will help researchers develop better therapies to improve the muscle health of elderly individuals.
Collapse
Affiliation(s)
| | - Guy M. Benian
- Department of Pathology Emory University Atlanta Georgia USA
| |
Collapse
|
22
|
Kramerova I, Marinov M, Owens J, Lee SJ, Becerra D, Spencer MJ. Myostatin inhibition promotes fast fibre hypertrophy but causes loss of AMP-activated protein kinase signalling and poor exercise tolerance in a model of limb-girdle muscular dystrophy R1/2A. J Physiol 2020; 598:3927-3939. [PMID: 33460149 DOI: 10.1113/jp279943] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/08/2020] [Indexed: 05/31/2024] Open
Abstract
KEY POINTS Limb-girdle muscular dystrophy R1 (LGMD R1) is caused by mutations in the CAPN3 gene and is characterized by progressive muscle loss, impaired mitochondrial function and reductions in the slow oxidative gene expression programme. Myostatin is a negative regulator of muscle growth, and its inhibition improves the phenotype in several muscle wasting disorders. The effect of genetic and pharmacological inhibition of myostatin signalling on the disease phenotype in a mouse model of LGMD R1 (CAPN3 knockout mouse-C3KO) was studied. Inhibition of myostatin signalling in C3KO muscles resulted in significant muscle hypertrophy; however, there were no improvements in muscle strength and exacerbation of exercise intolerance concomitant with further reduction of muscle oxidative capacity was observed. Inhibition of myostatin signalling is unlikely to be a valid therapeutic strategy for LGMD R1. ABSTRACT Limb-girdle muscular dystrophy R1 (LGMD R1) is caused by mutations in the CAPN3 gene and is characterized by progressive muscle loss, impaired mitochondrial function and reductions in the slow oxidative gene expression programme. There are currently no therapies available to patients. We sought to determine if induction of muscle growth, through myostatin inhibition, represents a viable therapeutic strategy for this disease. Myostatin is a negative regulator of muscle growth, and its inhibition improves the phenotype in several muscle wasting disorders. However, the effect of myostatin depends on the genetic and pathophysiological context and may not be efficacious in all contexts. We found that genetic inhibition of myostatin through overexpression of follistatin (an endogenous inhibitor of myostatin) in our LGMD R1 model (C3KO) resulted in 1.5- to 2-fold increase of muscle mass for the majority of limb muscles. However, muscle strength was not improved and exercise intolerance was exacerbated. Pharmacological inhibition of myostatin, using an anti-myostatin antibody, resulted in statistically significant increases in muscle mass; however, functional testing did not reveal changes in muscle strength nor endurance in treated C3KO mice. Histochemical and biochemical evaluation of follistatin overexpressing mice revealed a reduction in the percentage of oxidative fibres and decreased activation of AMP-activated protein kinase signalling in transgenics compared to C3KO muscles. Our data suggest that muscle hypertrophy, induced by myostatin inhibition, leads to loss of oxidative capacity, which further compromises metabolically impaired C3KO muscles and thus is unlikely to be a valid strategy for treatment of LGMD R1.
Collapse
Affiliation(s)
- Irina Kramerova
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Masha Marinov
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - Se-Jin Lee
- The Jackson Laboratory and University of Connecticut School of Medicine, Farmington, CT, USA
| | - Diana Becerra
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Melissa J Spencer
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
23
|
Duan K, Gao X, Zhu D. The clinical relevance and mechanism of skeletal muscle wasting. Clin Nutr 2020; 40:27-37. [PMID: 32788088 DOI: 10.1016/j.clnu.2020.07.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/15/2020] [Accepted: 07/21/2020] [Indexed: 02/08/2023]
Abstract
Skeletal muscle wasting occurs in both chronic and acute diseases. Increasing evidence has shown this debilitating process is associated with short- and long-term outcomes in critical, cancer and surgical patients. Both muscle quantity and quality, as reflected by the area and density of a given range of attenuation in CT scan, impact the patient prognosis. In addition, ultrasound and bioelectrical impedance analysis (BIA) are also widely used in the assessment of body composition due to their bedside viability and no radioactivity. Mechanism researches have revealed complicated pathways are involved in muscle wasting, which include altered IGF1-Akt-FoxO signaling, elevated levels of myostatin and activin A, activation of NF-κB pathway and glucocorticoid effects. Particularly, central nervous system (CNS) has been proven to participate in regulating muscle wasting in various conditions, such as infection and tumor. Several promising therapeutic agents have been under developing in the treatment of muscle atrophy, such as myostatin antagonist, ghrelin analog, non-steroidal selective androgen receptor modulators (SARMs). Notably, nutritional therapy is still the fundamental support in combating muscle wasting. However, the optimizing and tailored nutrition regimen relies on accurate metabolism measurement and large clinical trials in the future. Here, we will discuss the current understanding of muscle wasting and potential treatment in clinical practice.
Collapse
Affiliation(s)
- Kaipeng Duan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| | - Xin Gao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| | - Dongming Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China.
| |
Collapse
|
24
|
Magnolol inhibits myotube atrophy induced by cancer cachexia through myostatin signaling pathway in vitro. J Nat Med 2020; 74:741-749. [PMID: 32601830 DOI: 10.1007/s11418-020-01428-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/22/2020] [Indexed: 11/27/2022]
Abstract
Cancer cachexia is a complex and multifactorial syndrome that influences about 50-80% of cancer patients and may lead to 20% of cancer deaths and muscle atrophy is the key characteristic of the syndrome. Recent researches have shown that myostatin is a negative regulator in the growth and differentiation of skeletal muscle. Herein, C2C12 cancer cachexia model was established with C26 conditioned culture medium (CCM), then treated with magnolol to evaluate the pharmacological activity of magnolol in myotube atrophy. Our results demonstrated that magnolol inhibited the activity of myostatin promotor and the myostatin signaling pathway. In C2C12 cancer cachexia model, magnolol decreased myostatin expression, inhibited the phosphorylation of SMAD2/3 activated by C26 conditioned culture medium (CCM), and elevated the phosphorylation of FOXO3a lowered by CCM. Myosin heavy chain (MyHC), myogenin (MyoG), and myogenic differentiation (MyoD), as three common myotube markers in C2C12 myotube, were decreased by CCM, which could be effectively reversed by magnolol via activation of AKT/mTOR-regulated protein synthesis and inhibition of ubiquitin-mediated proteolysis. This study reveals that magnolol inhibits myotube atrophy induced by CCM by increasing protein synthesis and decreasing ubiquitin-mediated proteolysis, so that magnolol is a promising leading compound in treating muscle atrophy induced by cancer cachexia.
Collapse
|
25
|
Vohra R, Campbell MD, Park J, Whang S, Gravelle K, Wang YN, Hwang JH, Marcinek DJ, Lee D. Increased tumour burden alters skeletal muscle properties in the KPC mouse model of pancreatic cancer. JCSM RAPID COMMUNICATIONS 2020; 3:44-55. [PMID: 33073264 PMCID: PMC7566781 DOI: 10.1002/rco2.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
BACKGROUND Cancer cachexia is a multifactorial wasting syndrome that is characterized by the loss of skeletal muscle mass and weakness, which compromises physical function, reduces quality of life, and ultimately can lead to mortality. Experimental models of cancer cachexia have recapitulated this skeletal muscle atrophy and consequent decline in muscle force generating capacity. We address these issues in a novel transgenic mouse model Kras, Trp53 and Pdx-1-Cre (KPC) of pancreatic ductal adenocarcinoma (PDA) using multi-parametric magnetic resonance (mp-MR) measures. METHODS KPC mice (n = 10) were divided equally into two groups (n = 5/group) depending on the size of the tumor i.e. tumor size <250 mm3 and >250 mm3. Using mp-MR measures, we demonstrated the changes in the gastrocnemius muscle at the microstructural level. In addition, we evaluated skeletal muscle contractile function in KPC mice using an in vivo approach. RESULTS Increase in tumor size resulted in decrease in gastrocnemius maximum cross sectional area, decrease in T2 relaxation time, increase in magnetization transfer ratio, decrease in mean diffusivity, and decrease in radial diffusivity of water across the muscle fibers. Finally, we detected significant decrease in absolute and specific force production of gastrocnemius muscle with increase in tumor size. CONCLUSIONS Our findings indicate that increase in tumor size may cause alterations in structural and functional parameters of skeletal muscles and that MR parameters may be used as sensitive biomarkers to noninvasively detect structural changes in cachectic muscles.
Collapse
Affiliation(s)
- Ravneet Vohra
- Department of Radiology, University of Washington, Seattle,
USA
| | | | - Joshua Park
- Department of Radiology, University of Washington, Seattle,
USA
| | - Stella Whang
- Department of Medicine, University of Washington, Seattle,
USA
| | - Kayla Gravelle
- Department of Medicine, University of Washington, Seattle,
USA
| | - Yak-Nam Wang
- Applied Physics Laboratory, University of Washington,
Seattle, USA
| | - Joo-Ha Hwang
- Division of Gastroenterology and Hepatology, Stanford
University, Stanford, USA
| | | | - Donghoon Lee
- Department of Radiology, University of Washington, Seattle,
USA
| |
Collapse
|
26
|
Ojima C, Noguchi Y, Miyamoto T, Saito Y, Orihashi H, Yoshimatsu Y, Watabe T, Takayama K, Hayashi Y, Itoh F. Peptide-2 from mouse myostatin precursor protein alleviates muscle wasting in cancer-associated cachexia. Cancer Sci 2020; 111:2954-2964. [PMID: 32519375 PMCID: PMC7419029 DOI: 10.1111/cas.14520] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/01/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cachexia, characterized by continuous muscle wasting, is a key determinant of cancer‐related death; however, there are few medical treatments to combat it. Myostatin (MSTN)/growth differentiation factor 8 (GDF‐8), which is a member of the transforming growth factor‐β family, is secreted in an inactivated form noncovalently bound to the prodomain, negatively regulating the skeletal muscle mass. Therefore, inhibition of MSTN signaling is expected to serve as a therapeutic target for intractable muscle wasting diseases. Here, we evaluated the inhibitory effect of peptide‐2, an inhibitory core of mouse MSTN prodomain, on MSTN signaling. Peptide‐2 selectively suppressed the MSTN signal, although it had no effect on the activin signal. In contrast, peptide‐2 slightly inhibited the GDF‐11 signaling pathway, which is strongly related to the MSTN signaling pathway. Furthermore, we found that the i.m. injection of peptide‐2 to tumor‐implanted C57BL/6 mice alleviated muscle wasting in cancer cachexia. Although peptide‐2 was unable to improve the loss of heart weight and fat mass when cancer cachexia model mice were injected with it, peptide‐2 increased the gastrocnemius muscle weight and muscle cross‐sectional area resulted in the enhanced grip strength in cancer cachexia mice. Consequently, the model mice treated with peptide‐2 could survive longer than those that did not undergo this treatment. Our results suggest that peptide‐2 might be a novel therapeutic candidate to suppress muscle wasting in cancer cachexia.
Collapse
Affiliation(s)
- Chiharu Ojima
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yuri Noguchi
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Tatsuki Miyamoto
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yuki Saito
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Hiroki Orihashi
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yasuhiro Yoshimatsu
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tetsuro Watabe
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kentaro Takayama
- Department of Medicinal Chemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yoshio Hayashi
- Department of Medicinal Chemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Fumiko Itoh
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| |
Collapse
|
27
|
Zhang D, Cao L, Wang Z, Feng H, Cai X, Xu M, Li M, Yu N, Yin Y, Wang W, Kang J. Salidroside mitigates skeletal muscle atrophy in rats with cigarette smoke-induced COPD by up-regulating myogenin and down-regulating myostatin expression. Biosci Rep 2019; 39:BSR20190440. [PMID: 31702007 PMCID: PMC6879355 DOI: 10.1042/bsr20190440] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 10/17/2019] [Accepted: 11/06/2019] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVES The present study aimed at investigating the therapeutic effect of Salidroside on skeletal muscle atrophy in a rat model of cigarette smoking-induced chronic obstructive pulmonary disease (COPD) and its potential mechanisms. METHODS Male Wistar rats were randomized, and treated intraperitoneally (IP) with vehicle (injectable water) or a low, medium or high dose of Salidroside, followed by exposure to cigarette smoking daily for 16 weeks. A healthy control received vehicle injection and air exposure. Their lung function, body weights and gastrocnemius (GN) weights, grip strength and cross-section area (CSA) of individual muscular fibers in the GN were measured. The levels of TNF-α, IL-6, malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH) in serum and GN tissues as well as myostatin and myogenin expression in GN tissues were measured. RESULTS In comparison with that in the healthy control, long-term cigarette smoking induced emphysema, significantly impaired lung function, reduced body and GN weights and CSA values in rats, accompanied by significantly increased levels of TNF-α, IL-6 and MDA, but decreased levels of SOD and GSH in serum and GN tissues. Furthermore, cigarette smoking significantly up-regulated myostatin expression, but down-regulated myogenin expression in GN tissues. Salidroside treatment decreased emphysema, significantly ameliorated lung function, increased antioxidant, but reduced MDA, IL-6 and TNF-α levels in serum and GN tissues of rats, accompanied by decreased myostain, but increased myogenin expression in GN tissues. CONCLUSION Salidroside mitigates the long-term cigarette smoking-induced emphysema and skeletal muscle atrophy in rats by inhibiting oxidative stress and inflammatory responses and regulating muscle-specific transcription factor expression.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
- Department of Respiratory Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian 116000, Liaoning, China
| | - Lihua Cao
- Department of Respiratory Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian 116000, Liaoning, China
| | - Zhenshan Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian 116000, Liaoning, China
| | - Haoshen Feng
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Xu Cai
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Mingtao Xu
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Menglu Li
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Na Yu
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Yan Yin
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Wei Wang
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Jian Kang
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| |
Collapse
|
28
|
Swiderski K, Caldow MK, Naim T, Trieu J, Chee A, Koopman R, Lynch GS. Deletion of suppressor of cytokine signaling 3 (SOCS3) in muscle stem cells does not alter muscle regeneration in mice after injury. PLoS One 2019; 14:e0212880. [PMID: 30811469 PMCID: PMC6392323 DOI: 10.1371/journal.pone.0212880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 02/11/2019] [Indexed: 11/19/2022] Open
Abstract
Muscles of older animals are more susceptible to injury and regenerate poorly, in part due to a persistent inflammatory response. The janus kinase (Jak)/signal transducer and activator of transcription (Stat) pathway mediates inflammatory signaling and is tightly regulated by the suppressor of cytokine signaling (SOCS) proteins, especially SOCS3. SOCS3 expression is altered in the muscle of aged animals and may contribute to the persistent inflammation and impaired regeneration. To test this hypothesis, we performed myotoxic injuries on mice with a tamoxifen-inducible deletion of SOCS3 specifically within the muscle stem cell compartment. Muscle stem cell-specific SOCS3 deletion reduced muscle mass at 14 days post-injury (-14%, P < 0.01), altered the myogenic transcriptional program, and reduced myogenic fusion based on the number of centrally-located nuclei per muscle fiber. Despite the delay in myogenesis, muscles with a muscle stem cell-specific deletion of SOCS3 were still able to regenerate after a single bout or multiple bouts of myotoxic injury. A reduction in SOCS3 expression in muscle stem cells is unlikely to be responsible for the incomplete muscle repair in aged animals.
Collapse
Affiliation(s)
- Kristy Swiderski
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Marissa K. Caldow
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Timur Naim
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Jennifer Trieu
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Annabel Chee
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - René Koopman
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Gordon S. Lynch
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
29
|
Lynch GS, Koopman R. Overcoming nature’s paradox in skeletal muscle to optimise animal production. ANIMAL PRODUCTION SCIENCE 2019. [DOI: 10.1071/an19361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nature’s paradox in skeletal muscle describes the seemingly mutually exclusive relationship between muscle fibre size and oxidative capacity. In mammals, there is a constraint on the size at which mitochondria-rich, high O2-dependent oxidative fibres can attain before they become anoxic or adapt to a glycolytic phenotype, being less reliant on O2. This implies that a muscle fibre can hypertrophy at the expense of its endurance capacity. Adaptations to activity (exercise) generally obey this relationship, with optimal muscle endurance generally being linked to an enhanced proportion of small, slow oxidative fibres and muscle strength (force and/or power) being linked to an enhanced proportion of large, fast glycolytic fibres. This relationship generally constrains not only the physiological limits of performance (e.g. speed and endurance), but also the capacity to manipulate muscle attributes such as fibre size and composition, with important relevance to the livestock and aquaculture industries for producing specific muscle traits such as (flesh) quality, texture and taste. Highly glycolytic (white) muscles have different traits than do highly oxidative (red) muscles and so the ability to manipulate muscle attributes to produce flesh with specific traits has important implications for optimising meat production and quality. Understanding the biological regulation of muscle size, and phenotype and the capacity to manipulate signalling pathways to produce specific attributes, has important implications for promoting ethically sustainable and profitable commercial livestock and aquaculture practices and for developing alternative food sources, including ‘laboratory meat’ or ‘clean meat’. This review describes the exciting potential of manipulating muscle attributes relevant to animal production, through traditional nutritional and pharmacological approaches and through viral-mediated strategies that could theoretically push the limits of muscle fibre growth, adaptation and plasticity.
Collapse
|
30
|
Emerging role of myostatin and its inhibition in the setting of chronic kidney disease. Kidney Int 2018; 95:506-517. [PMID: 30598193 DOI: 10.1016/j.kint.2018.10.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/13/2018] [Accepted: 10/02/2018] [Indexed: 12/25/2022]
Abstract
The past two decades have witnessed tremendous progress in our understanding of the mechanisms underlying wasting and cachexia in chronic kidney disease (CKD) and in other chronic illnesses, such as cancer and heart failure. In all these conditions wasting is an effect of the activation of protein degradation in muscle, a response that increases the risk of morbidity and mortality. Major recent advances in our knowledge on how CKD and inflammation affect cellular signaling include the identification of the myostatin (MSTN)/activin system, and its related transcriptional program that promotes protein degradation. In addition, the identification of the role of MSTN/activin in the vascular wall shows premise that its inhibition can better control or prevent some effects of CKD on vessels, such as accelerated atherosclerosis and vascular calcifications. In this review, we summarize the expanding role of MSTN activation in promoting muscle atrophy and the recent clinical studies that investigated the efficacy of MSTN/activin pathway antagonism in sarcopenic patients. Moreover, we also review the utility of MSTN inhibition in the experimental models of CKD and its potential advantages in CKD patients. Lessons learned from clinical studies on MSTN antagonism in sarcopenic patients tell us that the anabolic intervention is likely better if we use a block of the two ActRII receptors. At the same time, however, it is becoming clear that MSTN-targeted therapies should not be seen as a substitute for physical activity and nutritional supplementation which are mandatory to successfully manage patients with wasting.
Collapse
|
31
|
Murphy KT, Hossain MI, Swiderski K, Chee A, Naim T, Trieu J, Haynes V, Read SJ, Stapleton DI, Judge SM, Trevino JG, Judge AR, Lynch GS. Mas Receptor Activation Slows Tumor Growth and Attenuates Muscle Wasting in Cancer. Cancer Res 2018; 79:706-719. [PMID: 30420474 DOI: 10.1158/0008-5472.can-18-1207] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 09/27/2018] [Accepted: 11/07/2018] [Indexed: 01/06/2023]
Abstract
Cancer cachexia is a multifactorial syndrome characterized by a progressive loss of skeletal muscle mass associated with significant functional impairment. Cachexia robs patients of their strength and capacity to perform daily tasks and live independently. Effective treatments are needed urgently. Here, we investigated the therapeutic potential of activating the "alternative" axis of the renin-angiotensin system, involving ACE2, angiotensin-(1-7), and the mitochondrial assembly receptor (MasR), for treating cancer cachexia. Plasmid overexpression of the MasR or pharmacologic angiotensin-(1-7)/MasR activation did not affect healthy muscle fiber size in vitro or in vivo but attenuated atrophy induced by coculture with cancer cells in vitro. In mice with cancer cachexia, the MasR agonist AVE 0991 slowed tumor development, reduced weight loss, improved locomotor activity, and attenuated muscle wasting, with the majority of these effects dependent on the orexigenic and not antitumor properties of AVE 0991. Proteomic profiling and IHC revealed that mechanisms underlying AVE 0991 effects on skeletal muscle involved miR-23a-regulated preservation of the fast, glycolytic fibers. MasR activation is a novel regulator of muscle phenotype, and AVE 0991 has orexigenic, anticachectic, and antitumorigenic effects, identifying it as a promising adjunct therapy for cancer and other serious muscle wasting conditions. SIGNIFICANCE: These findings demonstrate that MasR activation has multiple benefits of being orexigenic, anticachectic, and antitumorigenic, revealing it as a potential adjunct therapy for cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/4/706/F1.large.jpg.See related commentary by Rupert et al., p. 699.
Collapse
Affiliation(s)
- Kate T Murphy
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia.
| | - Mohammed I Hossain
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Kristy Swiderski
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Annabel Chee
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Timur Naim
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Jennifer Trieu
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Vanessa Haynes
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Suzannah J Read
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - David I Stapleton
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Sarah M Judge
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, Florida
| | - Jose G Trevino
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, Florida
| | - Andrew R Judge
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, Florida
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Golan T, Geva R, Richards D, Madhusudan S, Lin BK, Wang HT, Walgren RA, Stemmer SM. LY2495655, an antimyostatin antibody, in pancreatic cancer: a randomized, phase 2 trial. J Cachexia Sarcopenia Muscle 2018; 9:871-879. [PMID: 30051975 PMCID: PMC6204586 DOI: 10.1002/jcsm.12331] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 06/10/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Cachexia is a formidable clinical challenge in pancreatic cancer. We assessed LY2495655 (antimyostatin antibody) plus standard-of-care chemotherapy in pancreatic cancer using cachexia status as a stratifier. METHODS In this randomized, phase 2 trial, patients with stage II-IV pancreatic cancer were randomized to 300 mg LY2495655, 100 mg LY2495655, or placebo, plus physician-choice chemotherapy from a prespecified list of standard-of-care regimens for first and later lines of care. Investigational treatment was continued during second-line treatment. The primary endpoint was overall survival. RESULTS Overall, 125 patients were randomized. In August 2014, 300 mg LY2495655 was terminated due to imbalance in death rates between the treatment arms; in January 2015, 100 mg LY2495655 treatment was terminated due to futility. LY2495655 did not improve overall survival: the hazard ratio was 1.70 (90% confidence interval, 1.1-2.7) for 300 mg vs. placebo and 1.3 (0.82-2.1) for 100 mg vs. placebo (recommended doses). Progression-free survival results were consistent with the overall survival results. A numerically higher hazard ratio was observed in patients with weight loss (WL) of ≥5% (cachexia) than with <5% WL within 6 months before randomization. Subgroup analyses for patients stratified by WL in the 6 months preceding enrollment suggested that functional responses to LY2495655 (either dose) may have been superior in patients with <5% WL vs. patients with ≥5% WL. Among possibly drug-related adverse events, fatigue, diarrhoea, and anorexia were more common in LY2495655-treated than in placebo-treated patients. CONCLUSIONS In the intention-to-treat analysis, LY2495655 did not confer clinical benefit in pancreatic cancer. Our data highlight the importance of assessing survival when investigating therapeutic management of cachexia and support the use of WL as a stratifier (independent of performance status).
Collapse
Affiliation(s)
| | - Ravit Geva
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | - Srinivasan Madhusudan
- Academic Oncology, University of Nottingham, School of Medicine, Nottingham University Hospitals, City Hospital Campus, Nottingham, UK
| | | | | | | | - Salomon M Stemmer
- Rabin Medical Center, Petach Tikva, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
33
|
Satkunskiene D, Ratkevicius A, Kamandulis S, Venckunas T. Effects of myostatin on the mechanical properties of muscles during repeated active lengthening in the mouse. Appl Physiol Nutr Metab 2018; 44:381-388. [PMID: 30222937 DOI: 10.1139/apnm-2018-0369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of the present study was to investigate how myostatin dysfunction affects fast and slow muscle stiffness and viscosity during severe repeated loading. Isolated extensor digitorum longus (EDL) and soleus muscles of young adult female mice of the BEH (dysfunctional myostatin) and BEH+/+ (functional myostatin) strains were subjected to 100 contraction-stretching loading cycles during which contractile and mechanical properties were assessed. BEH mice exhibited greater exercise-induced muscle damage, although the effect was muscle- and age-dependent and limited to the early phases of simulated exercise. The relative reduction of the EDL muscle isometric force recorded during the initial 10-30 loading cycles was greater in BEH mice than in BEH+/+ mice and exceeded that of the soleus muscle of either strain. The induced damage was associated with lower muscle stiffness. The effects of myostatin on the mechanical properties of muscles depend on muscle type and maturity.
Collapse
Affiliation(s)
- Danguole Satkunskiene
- Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania.,Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania
| | - Aivaras Ratkevicius
- Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania.,Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania
| | - Sigitas Kamandulis
- Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania.,Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania
| | - Tomas Venckunas
- Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania.,Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania
| |
Collapse
|
34
|
Han YQ, Ming SL, Wu HT, Zeng L, Ba G, Li J, Lu WF, Han J, Du QJ, Sun MM, Yang GY, Wang J, Chu BB. Myostatin knockout induces apoptosis in human cervical cancer cells via elevated reactive oxygen species generation. Redox Biol 2018; 19:412-428. [PMID: 30241032 PMCID: PMC6146590 DOI: 10.1016/j.redox.2018.09.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 08/20/2018] [Accepted: 09/12/2018] [Indexed: 12/31/2022] Open
Abstract
Myostatin (Mstn) is postulated to be a key determinant of muscle loss and cachexia in cancer. However, no experimental evidence supports a role for Mstn in cancer, particularly in regulating the survival and growth of cancer cells. In this study, we showed that the expression of Mstn was significantly increased in different tumor tissues and human cancer cells. Mstn knockdown inhibited the proliferation of cancer cells. A knockout (KO) of Mstn created by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (Cas) 9 (CRISPR/Cas9) induced mitochondria-dependent apoptosis in HeLa cells. Furthermore, KO of Mstn reduced the lipid content. Molecular analyses demonstrated that the expression levels of fatty acid oxidation-related genes were upregulated and then increased rate of fatty acid oxidation. Mstn deficiency-induced apoptosis took place along with generation of reactive oxygen species (ROS) and elevated fatty acid oxidation, which may play a role in triggering mitochondrial membrane depolarization, the release of cytochrome c (Cyt-c), and caspase activation. Importantly, apoptosis induced by Mstn KO was partially rescued by antioxidants and etomoxir, thereby suggesting that the increased level of ROS was functionally involved in mediating apoptosis. Overall, our findings demonstrate a novel function of Mstn in regulating mitochondrial metabolism and apoptosis within cancer cells. Hence, inhibiting the production and function of Mstn may be an effective therapeutic intervention during cancer progression and muscle loss in cachexia. Mstn is expressed in different tumor tissues and human cancer cells. Mstn knockdown inhibits the proliferation of cancer cells. Mstn KO induces mitochondria-dependent apoptosis in HeLa cells. Mstn KO increases the rate of fatty acid oxidation. ROS generation induces apoptosis in HeLa/Mstn KO cells.
Collapse
Affiliation(s)
- Ying-Qian Han
- College of Animal Sciences and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, PR China
| | - Sheng-Li Ming
- College of Animal Sciences and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, PR China
| | - Hong-Tao Wu
- College of Animal Sciences and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, PR China
| | - Lei Zeng
- College of Animal Sciences and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, PR China
| | - Gen Ba
- College of Animal Sciences and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, PR China
| | - Jian Li
- College of Animal Sciences and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, PR China
| | - Wei-Fei Lu
- College of Animal Sciences and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, PR China; Department of Radiology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Jie Han
- Department of Endocrinology, the First Hospital of Lanzhou University, Lanzhou, Gansu Province, PR China
| | - Qia-Jun Du
- Clinical Laboratory, the Second Hospital of Lanzhou University, Lanzhou, Gansu Province, PR China
| | - Miao-Miao Sun
- The Pathology Department of the Affiliated Cancer Hospital, Zhengzhou University, Zhengzhou, Henan Province, PR China
| | - Guo-Yu Yang
- College of Animal Sciences and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, PR China.
| | - Jiang Wang
- College of Animal Sciences and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, PR China.
| | - Bei-Bei Chu
- College of Animal Sciences and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, PR China.
| |
Collapse
|
35
|
Kutz LC, Mukherji ST, Wang X, Bryant A, Larre I, Heiny JA, Lingrel JB, Pierre SV, Xie Z. Isoform-specific role of Na/K-ATPase α1 in skeletal muscle. Am J Physiol Endocrinol Metab 2018; 314:E620-E629. [PMID: 29438630 PMCID: PMC6032065 DOI: 10.1152/ajpendo.00275.2017] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The distribution of Na/K-ATPase α-isoforms in skeletal muscle is unique, with α1 as the minor (15%) isoform and α2 comprising the bulk of the Na/K-ATPase pool. The acute and isoform-specific role of α2 in muscle performance and resistance to fatigue is well known, but the isoform-specific role of α1 has not been as thoroughly investigated. In vitro, we reported that α1 has a role in promoting cell growth that is not supported by α2. To assess whether α1 serves this isoform-specific trophic role in the skeletal muscle, we used Na/K-ATPase α1-haploinsufficient (α1+/-) mice. A 30% decrease of Na/K-ATPase α1 protein expression without change in α2 induced a modest yet significant decrease of 10% weight in the oxidative soleus muscle. In contrast, the mixed plantaris and glycolytic extensor digitorum longus weights were not significantly affected, likely because of their very low expression level of α1 compared with the soleus. The soleus mass reduction occurred without change in total Na/K-ATPase activity or glycogen metabolism. Serum analytes including K+, fat tissue mass, and exercise capacity were not altered in α1+/- mice. The impact of α1 content on soleus muscle mass is consistent with a Na/K-ATPase α1-specific role in skeletal muscle growth that cannot be fulfilled by α2. The preserved running capacity in α1+/- is in sharp contrast with previously reported consequences of genetic manipulation of α2. Taken together, these results lend further support to the concept of distinct isoform-specific functions of Na/K-ATPase α1 and α2 in skeletal muscle.
Collapse
Affiliation(s)
- Laura C Kutz
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Shreya T Mukherji
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Xiaoliang Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Amber Bryant
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Isabel Larre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Judith A Heiny
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine , Cincinnati, Ohio
| | - Jerry B Lingrel
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine , Cincinnati, Ohio
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| |
Collapse
|
36
|
Muscle Atrophy: Present and Future. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:605-624. [DOI: 10.1007/978-981-13-1435-3_29] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
37
|
Winbanks CE, Murphy KT, Bernardo BC, Qian H, Liu Y, Sepulveda PV, Beyer C, Hagg A, Thomson RE, Chen JL, Walton KL, Loveland KL, McMullen JR, Rodgers BD, Harrison CA, Lynch GS, Gregorevic P. Smad7 gene delivery prevents muscle wasting associated with cancer cachexia in mice. Sci Transl Med 2017; 8:348ra98. [PMID: 27440729 DOI: 10.1126/scitranslmed.aac4976] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 06/22/2016] [Indexed: 12/12/2022]
Abstract
Patients with advanced cancer often succumb to complications arising from striated muscle wasting associated with cachexia. Excessive activation of the type IIB activin receptor (ActRIIB) is considered an important mechanism underlying this wasting, where circulating procachectic factors bind ActRIIB and ultimately lead to the phosphorylation of SMAD2/3. Therapeutics that antagonize the binding of ActRIIB ligands are in clinical development, but concerns exist about achieving efficacy without off-target effects. To protect striated muscle from harmful ActRIIB signaling, and to reduce the risk of off-target effects, we developed an intervention using recombinant adeno-associated viral vectors (rAAV vectors) that increase expression of Smad7 in skeletal and cardiac muscles. SMAD7 acts as an intracellular negative regulator that prevents SMAD2/3 activation and promotes degradation of ActRIIB complexes. In mouse models of cachexia, rAAV:Smad7 prevented wasting of skeletal muscles and the heart independent of tumor burden and serum levels of procachectic ligands. Mechanistically, rAAV:Smad7 administration abolished SMAD2/3 signaling downstream of ActRIIB and inhibited expression of the atrophy-related ubiquitin ligases MuRF1 and MAFbx. These findings identify muscle-directed Smad7 gene delivery as a potential approach for preventing muscle wasting under conditions where excessive ActRIIB signaling occurs, such as cancer cachexia.
Collapse
Affiliation(s)
| | - Kate T Murphy
- Department of Physiology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Bianca C Bernardo
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Hongwei Qian
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Yingying Liu
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | | | - Claudia Beyer
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Adam Hagg
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Rachel E Thomson
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Justin L Chen
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia. Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Kelly L Walton
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Kate L Loveland
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Julie R McMullen
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia. Department of Medicine, Monash University, Clayton, Victoria 3800, Australia. Department of Physiology, Monash University, Clayton, Victoria 3800, Australia
| | - Buel D Rodgers
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| | - Craig A Harrison
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia. Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia. Department of Physiology, Monash University, Clayton, Victoria 3800, Australia
| | - Gordon S Lynch
- Department of Physiology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Paul Gregorevic
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia. Department of Physiology, The University of Melbourne, Melbourne, Victoria 3010, Australia. Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia. Department of Neurology, The University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
38
|
Dasarathy S. Myostatin and beyond in cirrhosis: all roads lead to sarcopenia. J Cachexia Sarcopenia Muscle 2017; 8:864-869. [PMID: 29168629 PMCID: PMC5700432 DOI: 10.1002/jcsm.12262] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 12/22/2022] Open
Affiliation(s)
- Srinivasan Dasarathy
- Professor of Medicine, Cleveland Clinic Lerner College of Medicine; Director, Liver Metabolism Research; Staff, Departments of Gastroenterology, Hepatology and Pathobiology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
39
|
Mashinchian O, Pisconti A, Le Moal E, Bentzinger CF. The Muscle Stem Cell Niche in Health and Disease. Curr Top Dev Biol 2017; 126:23-65. [PMID: 29305000 DOI: 10.1016/bs.ctdb.2017.08.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The regulation of stem cells that maintain and regenerate postnatal tissues depends on extrinsic signals originating from their microenvironment, commonly referred to as the stem cell niche. Complex higher-order regulatory interrelationships with the tissue and factors in the systemic circulation are integrated and propagated to the stem cells through the niche. The stem cell niche in skeletal muscle tissue is both a paradigm for a structurally and functionally relatively static niche that maintains stem cell quiescence during tissue homeostasis, and a highly dynamic regenerative niche that is subject to extensive structural remodeling and a flux of different support cell populations. Conditions ranging from aging to chronically degenerative skeletal muscle diseases affect the composition of the niche and thereby impair the regenerative potential of muscle stem cells. A holistic and integrative understanding of the extrinsic mechanisms regulating muscle stem cells in health and disease in a broad systemic context will be imperative for the identification of regulatory hubs in the niche interactome that can be targeted to maintain, restore, or enhance the regenerative capacity of muscle tissue. Here, we review the microenvironmental regulation of muscle stem cells, summarize how niche dysfunction can contribute to disease, and discuss emerging therapeutic implications.
Collapse
Affiliation(s)
- Omid Mashinchian
- Nestlé Institute of Health Sciences, Lausanne, Switzerland; École Polytechnique Fédérale de Lausanne, Doctoral Program in Biotechnology and Bioengineering, Lausanne, Switzerland
| | - Addolorata Pisconti
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Emmeran Le Moal
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - C Florian Bentzinger
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
40
|
Wurtzel CNW, Gumucio JP, Grekin JA, Khouri RK, Davis CS, Russell AJ, Bedi A, Mendias CL. Pharmacological inhibition of myostatin protects against skeletal muscle atrophy and weakness after anterior cruciate ligament tear. J Orthop Res 2017; 35:2499-2505. [PMID: 28176368 PMCID: PMC5548641 DOI: 10.1002/jor.23537] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 02/02/2017] [Indexed: 02/04/2023]
Abstract
Anterior cruciate ligament (ACL) tears are among the most frequent knee injuries in sports medicine, with tear rates in the US up to 250,000 per year. Many patients who suffer from ACL tears have persistent atrophy and weakness even after considerable rehabilitation. Myostatin is a cytokine that directly induces muscle atrophy, and previous studies rodent models and patients have demonstrated an upregulation of myostatin after ACL tear. Using a preclinical rat model, our objective was to determine if the use of a bioneutralizing antibody against myostatin could prevent muscle atrophy and weakness after ACL tear. Rats underwent a surgically induced ACL tear and were treated with either a bioneutralizing antibody against myostatin (10B3, GlaxoSmithKline) or a sham antibody (E1-82.15, GlaxoSmithKline). Muscles were harvested at either 7 or 21 days after induction of a tear to measure changes in contractile function, fiber size, and genes involved in muscle atrophy and hypertrophy. These time points were selected to evaluate early and later changes in muscle structure and function. Compared to the sham antibody group, 7 days after ACL tear, myostatin inhibition reduced the expression of proteolytic genes and induced the expression of hypertrophy genes. These early changes in gene expression lead to a 22% increase in muscle fiber cross-sectional area and a 10% improvement in maximum isometric force production that were observed 21 days after ACL tear. Overall, myostatin inhibition lead to several favorable, although modest, changes in molecular biomarkers of muscle regeneration and reduced muscle atrophy and weakness following ACL tear. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2499-2505, 2017.
Collapse
Affiliation(s)
- Caroline N W Wurtzel
- Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Jonathan P Gumucio
- Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109,Department of Molecular & Integrative Physiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Jeremy A Grekin
- Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Roger K Khouri
- Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Carol S Davis
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Alan J Russell
- Muscle Metabolism DPU, GlaxoSmithKline Pharmaceuticals, 2301 Renaissance Blvd, King of Prussia, PA, 19406
| | - Asheesh Bedi
- Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Christopher L Mendias
- Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109,Department of Molecular & Integrative Physiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109,To whom correspondence should be addressed: Christopher L Mendias, PhD, ATC, Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 2017, Ann Arbor, MI 48109-2200, 734-764-3250,
| |
Collapse
|
41
|
Sakuma K, Yamaguchi A. Recent advances in pharmacological, hormonal, and nutritional intervention for sarcopenia. Pflugers Arch 2017; 470:449-460. [DOI: 10.1007/s00424-017-2077-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 12/25/2022]
|
42
|
Powers SK, Lynch GS, Murphy KT, Reid MB, Zijdewind I. Disease-Induced Skeletal Muscle Atrophy and Fatigue. Med Sci Sports Exerc 2017; 48:2307-2319. [PMID: 27128663 DOI: 10.1249/mss.0000000000000975] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Numerous health problems, including acute critical illness, cancer, diseases associated with chronic inflammation, and neurological disorders, often result in skeletal muscle weakness and fatigue. Disease-related muscle atrophy and fatigue is an important clinical problem because acquired skeletal muscle weakness can increase the duration of hospitalization, result in exercise limitation, and contribute to a poor quality of life. Importantly, skeletal muscle atrophy is also associated with increased morbidity and mortality of patients. Therefore, improving our understanding of the mechanism(s) responsible for skeletal muscle weakness and fatigue in patients is a required first step to develop clinical protocols to prevent these skeletal muscle problems. This review will highlight the consequences and potential mechanisms responsible for skeletal muscle atrophy and fatigue in patients experiencing acute critical illness, cancer, chronic inflammatory diseases, and neurological disorders.
Collapse
Affiliation(s)
- Scott K Powers
- 1Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL; 2Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, Victoria, AUSTRALIA; and 3Medical Physiology, Department of Neuroscience, University Medical Center Groningen, Groningen, THE NETHERLANDS
| | | | | | | | | |
Collapse
|
43
|
Disrupted Skeletal Muscle Mitochondrial Dynamics, Mitophagy, and Biogenesis during Cancer Cachexia: A Role for Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3292087. [PMID: 28785374 PMCID: PMC5530417 DOI: 10.1155/2017/3292087] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/06/2017] [Accepted: 06/19/2017] [Indexed: 12/22/2022]
Abstract
Chronic inflammation is a hallmark of cancer cachexia in both patients and preclinical models. Cachexia is prevalent in roughly 80% of cancer patients and accounts for up to 20% of all cancer-related deaths. Proinflammatory cytokines IL-6, TNF-α, and TGF-β have been widely examined for their regulation of cancer cachexia. An established characteristic of cachectic skeletal muscle is a disrupted capacity for oxidative metabolism, which is thought to contribute to cancer patient fatigue, diminished metabolic function, and muscle mass loss. This review's primary objective is to highlight emerging evidence linking cancer-induced inflammation to the dysfunctional regulation of mitochondrial dynamics, mitophagy, and biogenesis in cachectic muscle. The potential for either muscle inactivity or exercise to alter mitochondrial dysfunction during cancer cachexia will also be discussed.
Collapse
|
44
|
Ishida J, Konishi M, Saitoh M, Anker M, Anker SD, Springer J. Myostatin signaling is up-regulated in female patients with advanced heart failure. Int J Cardiol 2017; 238:37-42. [DOI: 10.1016/j.ijcard.2017.03.153] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 03/31/2017] [Accepted: 03/31/2017] [Indexed: 12/20/2022]
|
45
|
Ruivo EAB, Mello JRC, Cavenaghi OM, Werneck AL, Ferreira LL. Respiratory muscle strength of patients with esophagus and stomach neoplasms. FISIOTERAPIA EM MOVIMENTO 2017. [DOI: 10.1590/1980-5918.030.s01.ao13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Abstract Introduction: In cancer patients, the reduced food intake causes weight loss and promotes protein-calorie malnutrition. This results in loss of lean body mass, which affects both skeletal muscles and respiratory muscles. Objective: Evaluate and compare the respiratory muscle strength of patients with esophageal and stomach neoplasia during the preoperative period. Methods: This is a cross-sectional study carried out with 24 patients of both genders hospitalized in a teaching hospital. They underwent a physical therapy evaluation composed of anthropometric data and measurement of respiratory muscle strength through manovacuometry. Paired and unpaired t-tests were used to compare the values obtained with the predicted equations. Results: Regarding the disease prevalence, 66.66%(16) of the individuals had stomach neoplasm and 33.33%(8) esophageal neoplasm. Of the patients with esophageal neoplasm, 100% were men with a mean age of 63 ± 9.16 years. Of those with stomach neoplasm, 68.75% were men with a mean age of 69.36 ± 10.92 years. Female patients with stomach neoplasm had significantly higher BMI (p = 0.01) than male patients, and they were classified as overweight. Both neoplasms had significantly lower real values (p ≤ 0.05) than predicted values at the maximal expiratory pressure. Conclusion: Patients with esophageal and stomach neoplasms in the preoperative period present reduction in the expiratory muscle strength. There were no statistically significant differences, when we compared the maximum respiratory pressures between the two types of neoplasms investigated.
Collapse
|
46
|
Shyh-Chang N. Metabolic Changes During Cancer Cachexia Pathogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:233-249. [PMID: 29282687 DOI: 10.1007/978-981-10-6020-5_11] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Wasting of adipose tissue and skeletal muscle is a hallmark of metastatic cancer and a major cause of death. Like patients with cachexia caused by other chronic infections or inflammatory diseases, the cancer subject manifests both malnutrition and metabolic stress. Both carbohydrate utilization and amino acid incorporation are decreased in the muscles of cancer cachexia patients. Cancer cells affect host metabolism in two ways: (a) their own metabolism of nutrients into other metabolites and (b) circulating factors they secrete or induce the host to secrete. Accelerated glycolysis and lactate production, i.e., the Warburg effect and the resultant increase in Cori cycle activity, are the most widely discussed metabolic effects. Meanwhile, although a large number of pro-cachexia circulating factors have been found, such as TNFa, IL-6, myostatin, and PTHrp, none have been shown to be a dominant factor that can be targeted singly to treat cancer cachexia in humans. It is possible that given the complex multifactorial nature of the cachexia secretome, and the personalized differences between cancer patients, targeting any single circulating factor would always be insufficient to treat cachexia for all patients. Here we review the metabolic changes that occur in response to tumor growth and tumor-secreted factors during cachexia.
Collapse
Affiliation(s)
- Ng Shyh-Chang
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore.
| |
Collapse
|
47
|
Swiderski K, Thakur SS, Naim T, Trieu J, Chee A, Stapleton DI, Koopman R, Lynch GS. Muscle-specific deletion of SOCS3 increases the early inflammatory response but does not affect regeneration after myotoxic injury. Skelet Muscle 2016; 6:36. [PMID: 27800152 PMCID: PMC5078888 DOI: 10.1186/s13395-016-0108-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/05/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Muscles of old animals are injured more easily and regenerate poorly, attributed in part to increased levels of circulating pro-inflammatory cytokines. The Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling cascade is a key mediator of inflammatory cytokine action, and signaling via this pathway is increased in muscles with aging. As a negative regulator of JAK/STAT signaling, a key mediator of myogenic proliferation and differentiation, altered expression of suppressor of cytokine signaling (SOCS3) is likely to have important consequences for muscle regeneration. To model this scenario, we investigated the effect of SOCS3 deletion within mature muscle fibers on injury and repair. We tested the hypothesis that reduced SOCS3 function would alter the inflammatory response and impair muscle regeneration after myotoxic injury. METHODS Mice with a specific deletion of SOCS3 within mature skeletal muscle fibers were used to assess the effect of SOCS3 deletion on muscle injury and repair. Twelve-week-old or 24-month-old SOCS3 muscle-specific knockout (SOCS3 MKO) mice and littermate controls were either left uninjured or injured with a single injection of notexin (10 μg/ml) into the right tibialis anterior (TA) muscle. At 1, 2, 3, 5, 7, or 14 days post-injury, the right TA muscle was excised and subjected to histological, western immunoblotting, and gene expression analyses. Force production and fatigue were assessed in uninjured muscles and at 7 days post-notexin injury. RESULTS In uninjured muscles, SOCS3 deletion decreased force production during fatigue but had no effect on the gross or histological appearance of the TA muscles. After notexin injury, deletion of SOCS3 increased STAT3 phosphorylation at day 1 and increased the mRNA expression of the inflammatory cytokine TNF-α, and the inflammatory cell markers F4/80 and CD68 at day 2. Gene expression analysis of the regeneration markers Pax7, MyoD, and Myogenin indicated SOCS3 deletion had no effect on the progression of muscle repair after notexin injury. Inflammation and regeneration were also unchanged in the muscles of 24-month-old SOCS3 MKO mice compared with control. CONCLUSIONS Loss of SOCS3 expression in mature muscle fibers increased the inflammatory response to myotoxic injury but did not impair muscle regeneration in either adult or old mice. Therefore, reduced SOCS3 expression in muscle fibers is unlikely to underlie impaired muscle regeneration. Further investigation into the role of SOCS3 in other cell types involved in muscle repair is warranted.
Collapse
Affiliation(s)
- Kristy Swiderski
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, 3010 Australia
| | - Savant S Thakur
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, 3010 Australia
| | - Timur Naim
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, 3010 Australia
| | - Jennifer Trieu
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, 3010 Australia
| | - Annabel Chee
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, 3010 Australia
| | - David I Stapleton
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, 3010 Australia
| | - René Koopman
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, 3010 Australia
| | - Gordon S Lynch
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, 3010 Australia
| |
Collapse
|
48
|
Kennedy TL, Swiderski K, Murphy KT, Gehrig SM, Curl CL, Chandramouli C, Febbraio MA, Delbridge LMD, Koopman R, Lynch GS. BGP-15 Improves Aspects of the Dystrophic Pathology in mdx and dko Mice with Differing Efficacies in Heart and Skeletal Muscle. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:3246-3260. [PMID: 27750047 DOI: 10.1016/j.ajpath.2016.08.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 08/14/2016] [Accepted: 08/17/2016] [Indexed: 11/19/2022]
Abstract
Duchenne muscular dystrophy is a severe and progressive striated muscle wasting disorder that leads to premature death from respiratory and/or cardiac failure. We have previously shown that treatment of young dystrophic mdx and dystrophin/utrophin null (dko) mice with BGP-15, a coinducer of heat shock protein 72, ameliorated the dystrophic pathology. We therefore tested the hypothesis that later-stage BGP-15 treatment would similarly benefit older mdx and dko mice when the dystrophic pathology was already well established. Later stage treatment of mdx or dko mice with BGP-15 did not improve maximal force of tibialis anterior (TA) muscles (in situ) or diaphragm muscle strips (in vitro). However, collagen deposition (fibrosis) was reduced in TA muscles of BGP-15-treated dko mice but unchanged in TA muscles of treated mdx mice and diaphragm of treated mdx and dko mice. We also examined whether BGP-15 treatment could ameliorate aspects of the cardiac pathology, and in young dko mice it reduced collagen deposition and improved both membrane integrity and systolic function. These results confirm BGP-15's ability to improve aspects of the dystrophic pathology but with differing efficacies in heart and skeletal muscles at different stages of the disease progression. These findings support a role for BGP-15 among a suite of pharmacological therapies for Duchenne muscular dystrophy and related disorders.
Collapse
Affiliation(s)
- Tahnee L Kennedy
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, Victoria
| | - Kristy Swiderski
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, Victoria
| | - Kate T Murphy
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, Victoria
| | - Stefan M Gehrig
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, Victoria
| | - Claire L Curl
- Cardiac Phenomics Laboratory, Department of Physiology, The University of Melbourne, Melbourne, Victoria
| | - Chanchal Chandramouli
- Cardiac Phenomics Laboratory, Department of Physiology, The University of Melbourne, Melbourne, Victoria
| | - Mark A Febbraio
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Lea M D Delbridge
- Cardiac Phenomics Laboratory, Department of Physiology, The University of Melbourne, Melbourne, Victoria
| | - René Koopman
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, Victoria
| | - Gordon S Lynch
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, Victoria.
| |
Collapse
|
49
|
Swiderski K, Martins KJB, Chee A, Trieu J, Naim T, Gehrig SM, Baum DM, Brenmoehl J, Chau L, Koopman R, Gregorevic P, Metzger F, Hoeflich A, Lynch GS. Skeletal muscle-specific overexpression of IGFBP-2 promotes a slower muscle phenotype in healthy but not dystrophic mdx mice and does not affect the dystrophic pathology. Growth Horm IGF Res 2016; 30-31:1-10. [PMID: 27544574 DOI: 10.1016/j.ghir.2016.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/25/2016] [Accepted: 07/27/2016] [Indexed: 01/14/2023]
Abstract
OBJECTIVE The insulin-like growth factor binding proteins (IGFBPs) are thought to modulate cell size and homeostasis via IGF-I-dependent and -independent pathways. There is a considerable dearth of information regarding the function of IGFBPs in skeletal muscle, particularly their role in the pathophysiology of Duchenne muscular dystrophy (DMD). In this study we tested the hypothesis that intramuscular IGFBP-2 overexpression would ameliorate the pathology in mdx dystrophic mice. DESIGN 4week old male C57Bl/10 and mdx mice received a single intramuscular injection of AAV6-empty or AAV6-IGFBP-2 vector into the tibialis anterior muscle. At 8weeks post-injection the effect of IGFBP-2 overexpression on the structure and function of the injected muscle was assessed. RESULTS AAV6-mediated IGFBP-2 overexpression in the tibialis anterior (TA) muscles of 4-week-old C57BL/10 and mdx mice reduced the mass of injected muscle after 8weeks, inducing a slower muscle phenotype in C57BL/10 but not mdx mice. Analysis of inflammatory and fibrotic gene expression revealed no changes between control and IGFBP-2 injected muscles in dystrophic (mdx) mice. CONCLUSIONS Together these results indicate that the IGFBP-2-induced promotion of a slower muscle phenotype is impaired in muscles of dystrophin-deficient mdx mice, which contributes to the inability of IGFBP-2 to ameliorate the dystrophic pathology. The findings implicate the dystrophin-glycoprotein complex (DGC) in the signaling required for this adaptation.
Collapse
Affiliation(s)
- Kristy Swiderski
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Karen Janet Bernice Martins
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Annabel Chee
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Jennifer Trieu
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Timur Naim
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Stefan Martin Gehrig
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Dale Michael Baum
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Julia Brenmoehl
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Luong Chau
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - René Koopman
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Paul Gregorevic
- Muscle Biology and Therapeutics Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Friedrich Metzger
- F. Hoffmann-La Roche Ltd., pRED, Pharma Research & Early Development, DTA Neuroscience, 4070, Basel, Switzerland
| | - Andreas Hoeflich
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Gordon Stuart Lynch
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia.
| |
Collapse
|
50
|
Emerging therapies for the treatment of skeletal muscle wasting in chronic obstructive pulmonary disease. Pharmacol Ther 2016; 166:56-70. [DOI: 10.1016/j.pharmthera.2016.06.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2016] [Indexed: 12/18/2022]
|