101
|
Rohm M, Zeigerer A, Machado J, Herzig S. Energy metabolism in cachexia. EMBO Rep 2019; 20:embr.201847258. [PMID: 30890538 DOI: 10.15252/embr.201847258] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/11/2019] [Accepted: 02/05/2019] [Indexed: 12/26/2022] Open
Abstract
Cachexia is a wasting disorder that accompanies many chronic diseases including cancer and results from an imbalance of energy requirements and energy uptake. In cancer cachexia, tumor-secreted factors and/or tumor-host interactions cause this imbalance, leading to loss of adipose tissue and skeletal and cardiac muscle, which weakens the body. In this review, we discuss how energy enters the body and is utilized by the different organs, including the gut, liver, adipose tissue, and muscle, and how these organs contribute to the energy wasting observed in cachexia. We also discuss futile cycles both between the organs and within the cells, which are often used to fine-tune energy supply under physiologic conditions. Ultimately, understanding the complex interplay of pathologic energy-wasting circuits in cachexia can bring us closer to identifying effective treatment strategies for this devastating wasting disease.
Collapse
Affiliation(s)
- Maria Rohm
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Anja Zeigerer
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Juliano Machado
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany .,Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany.,Chair Molecular Metabolic Control, Technical University Munich, Munich, Germany
| |
Collapse
|
102
|
The role of omega 3 fatty acids in suppressing muscle protein catabolism: A possible therapeutic strategy to reverse cancer cachexia? J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.12.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
103
|
Moderate Exercise Improves Experimental Cancer Cachexia by Modulating the Redox Homeostasis. Cancers (Basel) 2019; 11:cancers11030285. [PMID: 30823492 PMCID: PMC6468783 DOI: 10.3390/cancers11030285] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/20/2019] [Accepted: 02/23/2019] [Indexed: 12/19/2022] Open
Abstract
Cachexia is a debilitating syndrome that complicates the management of cancer patients. Muscle wasting, one of the main features of cachexia, is associated with hyper-activation of protein degradative pathways and altered mitochondrial function that could both result from impaired redox homeostasis. This study aimed to investigate the contribution of oxidative stress to cancer-induced cachexia in the presence or in the absence of moderate exercise training. Mice bearing the colon C26 carcinoma, either sedentary or exercised, were used. The former showed muscle wasting and redox imbalance, with the activation of an antioxidant response and with upregulation of markers of proteasome-dependent protein degradation and autophagy. Moderate exercise was able to relieve muscle wasting and prevented the loss of muscle strength; such a pattern was associated with reduced levels of Reactive Oxygen Species (ROS), carbonylated proteins and markers of autophagy and with improved antioxidant capacity. The muscle of sedentary tumor hosts also showed increased levels of molecular markers of mitophagy and reduced mitochondrial mass. Conversely, exercise in the C26 hosts led to increased mitochondrial mass. In conclusion, moderate exercise could be an effective non-pharmacological approach to prevent muscle wasting in cancer patients, decreasing muscle protein catabolism and oxidative stress and preserving mitochondria.
Collapse
|
104
|
Fernando R, Drescher C, Nowotny K, Grune T, Castro JP. Impaired proteostasis during skeletal muscle aging. Free Radic Biol Med 2019; 132:58-66. [PMID: 30194981 DOI: 10.1016/j.freeradbiomed.2018.08.037] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 08/03/2018] [Accepted: 08/30/2018] [Indexed: 01/03/2023]
Abstract
Aging is a complex phenomenon that has detrimental effects on tissue homeostasis. The skeletal muscle is one of the earliest tissues to be affected and to manifest age-related changes such as functional impairment and the loss of mass. Common to these alterations and to most of tissues during aging is the disruption of the proteostasis network by detrimental changes in the ubiquitin-proteasomal system (UPS) and the autophagy-lysosomal system (ALS). In fact, during aging the accumulation of protein aggregates, a process mainly driven by increased levels of oxidative stress, has been observed, clearly demonstrating UPS and ALS dysregulation. Since the UPS and ALS are the two most important pathways for the removal of misfolded and aggregated proteins and also of damaged organelles, we provide here an overview on the current knowledge regarding the connection between the loss of proteostasis and skeletal muscle functional impairment and also how redox regulation can play a role during aging. Therefore, this review serves for a better understanding of skeletal muscle aging in regard to the loss of proteostasis and how redox regulation can impact its function and maintenance.
Collapse
Affiliation(s)
- Raquel Fernando
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
| | - Cathleen Drescher
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
| | - Kerstin Nowotny
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany; German Center for Cardiovascular Research (DZHK), 10117 Berlin, Germany; University of Potsdam, Institute of Nutritional Science, 14558 Nuthetal, Germany
| | - José Pedro Castro
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany; Faculty of Medicine, Department for Biomedicine, University of Porto, 4200-319, Portugal; Institute for Innovation and Health Research (I3S), Aging and Stress Group, R. Alfredo Allen, 4200-135 Porto, Portugal.
| |
Collapse
|
105
|
Lee DE, Bareja A, Bartlett DB, White JP. Autophagy as a Therapeutic Target to Enhance Aged Muscle Regeneration. Cells 2019; 8:cells8020183. [PMID: 30791569 PMCID: PMC6406986 DOI: 10.3390/cells8020183] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/30/2019] [Accepted: 02/14/2019] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle has remarkable regenerative capacity, relying on precise coordination between resident muscle stem cells (satellite cells) and the immune system. The age-related decline in skeletal muscle regenerative capacity contributes to the onset of sarcopenia, prolonged hospitalization, and loss of autonomy. Although several age-sensitive pathways have been identified, further investigation is needed to define targets of cellular dysfunction. Autophagy, a process of cellular catabolism, is emerging as a key regulator of muscle regeneration affecting stem cell, immune cell, and myofiber function. Muscle stem cell senescence is associated with a suppression of autophagy during key phases of the regenerative program. Macrophages, a key immune cell involved in muscle repair, also rely on autophagy to aid in tissue repair. This review will focus on the role of autophagy in various aspects of the regenerative program, including adult skeletal muscle stem cells, monocytes/macrophages, and corresponding age-associated dysfunction. Furthermore, we will highlight rejuvenation strategies that alter autophagy to improve muscle regenerative function.
Collapse
Affiliation(s)
- David E Lee
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA.
| | - Akshay Bareja
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA.
| | - David B Bartlett
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA.
- Division of Medical Oncology, Department of Medicine, Duke University School of Medicine, Durham, NC 27701, USA.
- Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC 27701, USA.
| | - James P White
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA.
- Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC 27701, USA.
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC 27701, USA.
| |
Collapse
|
106
|
Penna F, Ballarò R, Beltrà M, De Lucia S, García Castillo L, Costelli P. The Skeletal Muscle as an Active Player Against Cancer Cachexia. Front Physiol 2019; 10:41. [PMID: 30833900 PMCID: PMC6387914 DOI: 10.3389/fphys.2019.00041] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/14/2019] [Indexed: 12/20/2022] Open
Abstract
The management of cancer patients is frequently complicated by the occurrence of cachexia. This is a complex syndrome that markedly impacts on quality of life as well as on tolerance and response to anticancer treatments. Loss of body weight, wasting of both adipose tissue and skeletal muscle and reduced survival rates are among the main features of cachexia. Skeletal muscle wasting has been shown to depend, mainly at least, on the induction of protein degradation rates above physiological levels. Such hypercatabolic pattern is driven by overactivation of different intracellular proteolytic systems, among which those dependent on ubiquitin-proteasome and autophagy. Selective rather than bulk degradation of altered proteins and organelles was also proposed to occur. Within the picture described above, the muscle is frequently considered a sort of by-stander tissue where external stimuli, directly or indirectly, can poise protein metabolism toward a catabolic setting. By contrast, several observations suggest that the muscle reacts to the wasting drive imposed by cancer growth by activating different compensatory strategies that include anabolic capacity, the activation of autophagy and myogenesis. Even if muscle response is eventually ill-fated, its occurrence supports the idea that in the presence of appropriate treatments the development of cancer-induced wasting might not be an ineluctable event in tumor hosts.
Collapse
Affiliation(s)
| | | | | | | | | | - Paola Costelli
- Department of Clinical and Biological Sciences, Interuniversity Institute of Myology, University of Turin, Turin, Italy
| |
Collapse
|
107
|
Ramesh J, Ronsard L, Gao A, Venugopal B. Autophagy Intertwines with Different Diseases-Recent Strategies for Therapeutic Approaches. Diseases 2019; 7:diseases7010015. [PMID: 30717078 PMCID: PMC6473623 DOI: 10.3390/diseases7010015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a regular and substantial “clear-out process” that occurs within the cell and that gets rid of debris that accumulates in membrane-enclosed vacuoles by using enzyme-rich lysosomes, which are filled with acids that degrade the contents of the vacuoles. This machinery is well-connected with many prevalent diseases, including cancer, HIV, and Parkinson’s disease. Considering that autophagy is well-known for its significant connections with a number of well-known fatal diseases, a thorough knowledge of the current findings in the field is essential in developing therapies to control the progression rate of diseases. Thus, this review summarizes the critical events comprising autophagy in the cellular system and the significance of its key molecules in manifesting this pathway in various diseases for down- or upregulation. We collectively reviewed the role of autophagy in various diseases, mainly neurodegenerative diseases, cancer, inflammatory diseases, and renal disorders. Here, some collective reports on autophagy showed that this process might serve as a dual performer: either protector or contributor to certain diseases. The aim of this review is to help researchers to understand the role of autophagy-regulating genes encoding functional open reading frames (ORFs) and its connection with diseases, which will eventually drive better understanding of both the progression and suppression of different diseases at various stages. This review also focuses on certain novel therapeutic strategies which have been published in the recent years based on targeting autophagy key proteins and its interconnecting signaling cascades.
Collapse
Affiliation(s)
- Janani Ramesh
- Department of Medical Biochemistry, Dr. A.L.M. Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai 600113, India.
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Larance Ronsard
- The Ragon Institute of Massachusetts General Hospital, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02140, USA.
| | - Anthony Gao
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Bhuvarahamurthy Venugopal
- Department of Medical Biochemistry, Dr. A.L.M. Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai 600113, India.
| |
Collapse
|
108
|
Hentilä J, Nissinen TA, Korkmaz A, Lensu S, Silvennoinen M, Pasternack A, Ritvos O, Atalay M, Hulmi JJ. Activin Receptor Ligand Blocking and Cancer Have Distinct Effects on Protein and Redox Homeostasis in Skeletal Muscle and Liver. Front Physiol 2019; 9:1917. [PMID: 30713500 PMCID: PMC6345696 DOI: 10.3389/fphys.2018.01917] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/20/2018] [Indexed: 12/25/2022] Open
Abstract
Muscle wasting in cancer cachexia can be alleviated by blocking activin receptor type 2 (ACVR2) ligands through changes in protein synthesis/degradation. These changes in cellular and protein metabolism may alter protein homeostasis. First, we elucidated the acute (1–2 days) and 2-week effects of blocking ACVR2 ligands by soluble activin receptor 2B (sACVR2B-Fc) on unfolded protein response (UPR), heat shock proteins (HSPs) and redox balance in a healthy mouse skeletal muscle. Second, we examined UPR, autophagy and redox balance with or without sACVR2B-Fc administration in muscle and liver of C26 tumor-bearing mice. The indicators of UPR and HSPs were not altered 1–2 days after a single sACVR2B-Fc administration in healthy muscles, but protein carbonyls increased (p < 0.05). Two weeks of sACVR2B-Fc administration increased muscle size, which was accompanied by increased UPR markers: GRP78 (p < 0.05), phosphorylated eIF2α (p < 0.01) and HSP47 (p < 0.01). Additionally, protein carbonyls and reduced form of glutathione increased (GSH) (p < 0.05). On the other hand, C26 cancer cachexia manifested decreased UPR markers (p-eIF2α, HSP47, p-JNK; p < 0.05) and antioxidant GSH (p < 0.001) in muscle, whereas the ratio of oxidized to reduced glutathione increased (GSSG/GSH; p < 0.001). Administration of sACVR2B-Fc prevented the decline in GSH and increased some of the UPR indicators in tumor-bearing mice. Additionally, autophagy markers LC3II/I (p < 0.05), Beclin-1 (p < 0.01), and P62 (p < 0.05) increased in the skeletal muscle of tumor-bearing mice. Finally, indicators of UPR, PERK, p-eIF2α and GRP78, increased (p < 0.05), whereas ATF4 was strongly decreased (p < 0.01) in the liver of tumor-bearing mice while sACVR2B-Fc had no effect. Muscle GSH and many of the altered UPR indicators correlated with tumor mass, fat mass and body mass loss. In conclusion, experimental cancer cachexia is accompanied by distinct and tissue-specific changes in proteostasis. Muscle hypertrophy induced by blocking ACVR2B ligands may be accompanied by the induction of UPR and increased protein carbonyls but blocking ACVR2B ligands may upregulate antioxidant protection.
Collapse
Affiliation(s)
- Jaakko Hentilä
- Faculty of Sport and Health Sciences, Neuromuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Tuuli A Nissinen
- Faculty of Sport and Health Sciences, Neuromuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Ayhan Korkmaz
- Institute of Biomedicine, Physiology, University of Eastern Finland, Kuopio, Finland
| | - Sanna Lensu
- Faculty of Sport and Health Sciences, Neuromuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Mika Silvennoinen
- Faculty of Sport and Health Sciences, Neuromuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Arja Pasternack
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mustafa Atalay
- Institute of Biomedicine, Physiology, University of Eastern Finland, Kuopio, Finland
| | - Juha J Hulmi
- Faculty of Sport and Health Sciences, Neuromuscular Research Center, University of Jyväskylä, Jyväskylä, Finland.,Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
109
|
Ballarò R, Beltrà M, De Lucia S, Pin F, Ranjbar K, Hulmi JJ, Costelli P, Penna F. Moderate exercise in mice improves cancer plus chemotherapy-induced muscle wasting and mitochondrial alterations. FASEB J 2019; 33:5482-5494. [PMID: 30653354 DOI: 10.1096/fj.201801862r] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cancer cachexia is a multifactorial syndrome characterized by anorexia, body wasting, and muscle and adipose tissue loss, impairing patient's tolerance to anticancer treatments and survival. The aim of the present study was to compare the effects induced in mice by tumor growth alone (C26) or in combination with chemotherapy [C26 oxaliplatin and 5-fluorouracil (oxfu)] and to evaluate the potential of moderate exercise. Oxfu administration to C26 mice exacerbated muscle wasting and triggered autophagy or mitophagy, decreased protein synthesis, and induced mitochondrial alterations. Exercise in C26 oxfu mice counteracted the loss of muscle mass and strength, partially rescuing autophagy and mitochondrial function. Nevertheless, exercise worsened survival in C26 oxfu mice in late stages of cachexia. In summary, chemotherapy further impinges on cancer-induced alterations, worsening muscle wasting. An ideal multifactorial and early intervention to prevent cancer cachexia could take advantage of exercise, improving patient's energy metabolism, mobility, and quality of life.-Ballarò, R., Beltrà, M., De Lucia, S., Pin, F., Ranjbar, K., Hulmi, J. J., Costelli, P., Penna, F. Moderate exercise in mice improves cancer plus chemotherapy-induced muscle wasting and mitochondrial alterations.
Collapse
Affiliation(s)
- Riccardo Ballarò
- Department of Clinical and Biological Sciences, Experimental Medicine and Clinical Pathology Unit, University of Torino, Torino, Italy.,Interuniversity Institute of Myology, Assisi, Italy
| | - Marc Beltrà
- Department of Clinical and Biological Sciences, Experimental Medicine and Clinical Pathology Unit, University of Torino, Torino, Italy.,Interuniversity Institute of Myology, Assisi, Italy
| | - Serena De Lucia
- Department of Clinical and Biological Sciences, Experimental Medicine and Clinical Pathology Unit, University of Torino, Torino, Italy.,Interuniversity Institute of Myology, Assisi, Italy
| | - Fabrizio Pin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kia Ranjbar
- Department of Exercise Physiology, Faculty of Physical Education and Sport Science, University of Tarbiat Modares, Tehran, Iran
| | - Juha J Hulmi
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Paola Costelli
- Department of Clinical and Biological Sciences, Experimental Medicine and Clinical Pathology Unit, University of Torino, Torino, Italy.,Interuniversity Institute of Myology, Assisi, Italy
| | - Fabio Penna
- Department of Clinical and Biological Sciences, Experimental Medicine and Clinical Pathology Unit, University of Torino, Torino, Italy.,Interuniversity Institute of Myology, Assisi, Italy
| |
Collapse
|
110
|
Kretschmar C, Peña-Oyarzun D, Hernando C, Hernández-Moya N, Molina-Berríos A, Hernández-Cáceres MP, Lavandero S, Budini M, Morselli E, Parra V, Troncoso R, Criollo A. Polycystin-2 Is Required for Starvation- and Rapamycin-Induced Atrophy in Myotubes. Front Endocrinol (Lausanne) 2019; 10:280. [PMID: 31133985 PMCID: PMC6517509 DOI: 10.3389/fendo.2019.00280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 04/16/2019] [Indexed: 12/13/2022] Open
Abstract
Muscle atrophy involves a massive catabolism of intracellular components leading to a significant reduction in cellular and tissue volume. In this regard, autophagy, an intracellular mechanism that degrades proteins and organelles, has been implicated with muscle breakdown. Recently, it has shown that polycystin-2 (PC2), a membrane protein that belongs to the transient receptor potential (TRP) family, is required for the maintenance of cellular proteostasis, by regulating autophagy in several cell types. The role of PC2 in the control of atrophy and autophagy in skeletal muscle remains unknown. Here, we show that PC2 is required for the induction of atrophy in C2C12 myotubes caused by nutrient deprivation or rapamycin exposure. Consistently, overexpression of PC2 induces atrophy in C2C12 myotubes as indicated by decreasing of the myogenic proteins myogenin and caveolin-3. In addition, we show that inhibition of mTORC1, by starvation or rapamycin is inhibited in cells when PC2 is silenced. Importantly, even if PC2 regulates mTORC1, our results show that the regulation of atrophy by PC2 is independent of autophagy. This study provides novel evidence regarding the role of PC2 in skeletal muscle cell atrophy.
Collapse
Affiliation(s)
- Catalina Kretschmar
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas, Universidad de Chile, Santiago, Chile
- Facultad Ciencias Químicas y Farmacéuticas and Facultad Medicina, Advanced Center for Chronic Diseases, Universidad de Chile, Santiago, Chile
| | - Daniel Peña-Oyarzun
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas, Universidad de Chile, Santiago, Chile
- Facultad Ciencias Químicas y Farmacéuticas and Facultad Medicina, Advanced Center for Chronic Diseases, Universidad de Chile, Santiago, Chile
| | - Cecilia Hernando
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas, Universidad de Chile, Santiago, Chile
- Facultad Ciencias Químicas y Farmacéuticas and Facultad Medicina, Advanced Center for Chronic Diseases, Universidad de Chile, Santiago, Chile
| | - Nadia Hernández-Moya
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas, Universidad de Chile, Santiago, Chile
- Facultad Ciencias Químicas y Farmacéuticas and Facultad Medicina, Advanced Center for Chronic Diseases, Universidad de Chile, Santiago, Chile
| | - Alfredo Molina-Berríos
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas, Universidad de Chile, Santiago, Chile
| | - María Paz Hernández-Cáceres
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sergio Lavandero
- Facultad Ciencias Químicas y Farmacéuticas and Facultad Medicina, Advanced Center for Chronic Diseases, Universidad de Chile, Santiago, Chile
- Facultad de Medicina, Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Universidad de Chile, Santiago, Chile
- Cardiology Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Mauricio Budini
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas, Universidad de Chile, Santiago, Chile
- Autophagy Research Center, Santiago, Chile
| | - Eugenia Morselli
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Autophagy Research Center, Santiago, Chile
| | - Valentina Parra
- Facultad Ciencias Químicas y Farmacéuticas and Facultad Medicina, Advanced Center for Chronic Diseases, Universidad de Chile, Santiago, Chile
- Facultad de Medicina, Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Universidad de Chile, Santiago, Chile
- Autophagy Research Center, Santiago, Chile
| | - Rodrigo Troncoso
- Facultad Ciencias Químicas y Farmacéuticas and Facultad Medicina, Advanced Center for Chronic Diseases, Universidad de Chile, Santiago, Chile
- Autophagy Research Center, Santiago, Chile
- Laboratorio de Investigación en Nutrición y Actividad Física, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
- *Correspondence: Rodrigo Troncoso
| | - Alfredo Criollo
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas, Universidad de Chile, Santiago, Chile
- Facultad Ciencias Químicas y Farmacéuticas and Facultad Medicina, Advanced Center for Chronic Diseases, Universidad de Chile, Santiago, Chile
- Autophagy Research Center, Santiago, Chile
- Alfredo Criollo
| |
Collapse
|
111
|
Saavedra P, Perrimon N. Drosophila as a Model for Tumor-Induced Organ Wasting. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1167:191-205. [PMID: 31520356 DOI: 10.1007/978-3-030-23629-8_11] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In humans, cancer-associated cachexia is a complex syndrome that reduces the overall quality of life and survival of cancer patients, particularly for those undergoing chemotherapy. The most easily observable sign of cachexia is organ wasting, the dramatic loss of skeletal muscle and adipose tissue mass. Estimates suggest that 80% of patients in advanced stages of cancer show signs of the syndrome and about 20% of cancer patients die directly of cachexia. Because there is no treatment or drug available to ameliorate organ wasting induced by cancer, cachexia is a relevant clinical problem. However, it is unclear how cachexia is mediated, what factors drive interactions between tumors and host tissues, and which markers of cachexia might be used to allow early detection before the observable signs of organ wasting. In this chapter, we review the current mammalian models of cachexia and the need to use new models of study. We also explain recent developments in Drosophila as a model for studying organ wasting induced by tumors and how fly studies can help unravel important mechanisms that drive cachexia. In particular, we discuss what lessons have been learned from tumor models recently reported to induce systemic organ wasting in Drosophila.
Collapse
Affiliation(s)
- Pedro Saavedra
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA. .,Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
112
|
Afroze D, Kumar A. ER stress in skeletal muscle remodeling and myopathies. FEBS J 2019; 286:379-398. [PMID: 29239106 PMCID: PMC6002870 DOI: 10.1111/febs.14358] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 11/24/2017] [Accepted: 12/07/2017] [Indexed: 12/18/2022]
Abstract
Skeletal muscle is a highly plastic tissue in the human body that undergoes extensive adaptation in response to environmental cues, such as physical activity, metabolic perturbation, and disease conditions. The endoplasmic reticulum (ER) plays a pivotal role in protein folding and calcium homeostasis in many mammalian cell types, including skeletal muscle. However, overload of misfolded or unfolded proteins in the ER lumen cause stress, which results in the activation of a signaling network called the unfolded protein response (UPR). The UPR is initiated by three ER transmembrane sensors: protein kinase R-like endoplasmic reticulum kinase, inositol-requiring protein 1α, and activating transcription factor 6. The UPR restores ER homeostasis through modulating the rate of protein synthesis and augmenting the gene expression of many ER chaperones and regulatory proteins. However, chronic heightened ER stress can also lead to many pathological consequences including cell death. Accumulating evidence suggests that ER stress-induced UPR pathways play pivotal roles in the regulation of skeletal muscle mass and metabolic function in multiple conditions. They have also been found to be activated in skeletal muscle under catabolic states, degenerative muscle disorders, and various types of myopathies. In this article, we have discussed the recent advancements toward understanding the role and mechanisms through which ER stress and individual arms of the UPR regulate skeletal muscle physiology and pathology.
Collapse
Affiliation(s)
- Dil Afroze
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Immunology and Molecular Medicine, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar, Kashmir, INDIA
| | - Ashok Kumar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
113
|
Bouchè M, Lozanoska-Ochser B, Proietti D, Madaro L. Do neurogenic and cancer-induced muscle atrophy follow common or divergent paths? Eur J Transl Myol 2018; 28:7931. [PMID: 30662704 PMCID: PMC6317130 DOI: 10.4081/ejtm.2018.7931] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 12/05/2018] [Indexed: 11/22/2022] Open
Abstract
Skeletal muscle is a dynamic tissue capable of responding to a large variety of physiological stimuli by adjusting muscle fiber size, metabolism and function. However, in pathological conditions such as cancer and neural disorders, this finely regulated homeostasis is impaired leading to severe muscle wasting, reduced muscle fiber size (atrophy), and impaired function. These disease features develop due to enhanced protein breakdown, which relies on two major degradation systems: the ubiquitin-proteasome and the autophagy-lysosome. These systems are independently regulated by different signalling pathways, which in physiological conditions, determine protein and organelle turnover. However, alterations in one or both systems, as it happens in several disorders, leads to enhanced protein breakdown and muscle atrophy. Although this is a common feature in the different types of muscle atrophy, the relative contribution of each of these systems is still under debate. Here, we will briefly describe the regulation and the activity of the ubiquitin-proteasome and the autophagy-lysosome systems during muscle wasting. We will then discuss what we know regarding how these pathways are involved in cancer induced and in neurogenic muscle atrophy, highlighting common and divergent paths. It is now clear that there is no one unifying common mechanism that can be applied to all models of muscle loss. Detailed understanding of the pathways and proteolysis mechanisms involved in each model will hopefully help the development of drugs to counteract muscle wasting in specific conditions.
Collapse
Affiliation(s)
- Marina Bouchè
- DAHFMO, Unit of Histology, Sapienza University of Rome, 00161 Rome, Italy.,Interuniversity Institute of Myology, Italy
| | | | - Daisy Proietti
- DAHFMO, Unit of Histology, Sapienza University of Rome, 00161 Rome, Italy.,IRCCS, Fondazione Santa Lucia, Rome, Italy
| | - Luca Madaro
- IRCCS, Fondazione Santa Lucia, Rome, Italy.,Interuniversity Institute of Myology, Italy
| |
Collapse
|
114
|
Liu Z, Sin KWT, Ding H, Doan HA, Gao S, Miao H, Wei Y, Wang Y, Zhang G, Li YP. p38β MAPK mediates ULK1-dependent induction of autophagy in skeletal muscle of tumor-bearing mice. Cell Stress 2018; 2:311-324. [PMID: 31225455 PMCID: PMC6551802 DOI: 10.15698/cst2018.11.163] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Muscle wasting is the key manifestation of cancer-associated cachexia, a lethal metabolic disorder seen in over 50% of cancer patients. Autophagy is activated in cachectic muscle of cancer hosts along with the ubiquitin-proteasome pathway (UPP), contributing to accelerated protein degradation and muscle wasting. However, established signaling mechanism that activates autophagy in response to fasting or denervation does not seem to mediate cancer-provoked autophagy in skeletal myocytes. Here, we show that p38β MAPK mediates autophagy activation in cachectic muscle of tumor-bearing mice via novel mechanisms. Complementary genetic and pharmacological manipulations reveal that activation of p38β MAPK, but not p38α MAPK, is necessary and sufficient for Lewis lung carcinoma (LLC)-induced autophagy activation in skeletal muscle cells. Particularly, muscle-specific knockout of p38β MAPK abrogates LLC tumor-induced activation of autophagy and UPP, sparing tumor-bearing mice from muscle wasting. Mechanistically, p38β MAPK-mediated activation of transcription factor C/EBPβ is required for LLC-induced autophagy activation, and upregulation of autophagy-related genes LC3b and Gabarapl1. Surprisingly, ULK1 activation (phosphorylation at S555) by cancer requires p38β MAPK, rather than AMPK. Activated ULK1 forms a complex with p38β MAPK in myocytes, which is markedly increased by a tumor burden. Overexpression of a constitutively active p38Tbeta; MAPK in HEK293 cells increases phosphorylation at S555 and other amino acid residues of ULK1, but not several of AMPK-mediated sites. Finally, ULK1 activation is abrogated in tumor-bearing mice with muscle-specific knockout of p38β MAPK. Thus, p38β MAPK appears a key mediator of cancer-provoked autophagy activation, and a therapeutic target of cancer-induced muscle wasting.
Collapse
Affiliation(s)
- Zhelong Liu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, USA.,Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ka Wai Thomas Sin
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Hui Ding
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, USA.,Department of Respiratory Medicine, Yixing Hospital affiliated to Jiangsu University, Yixing, China
| | - HoangAnh Amy Doan
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Song Gao
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Hongyu Miao
- School of Public Health, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Yahui Wei
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Yiman Wang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Guohua Zhang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Yi-Ping Li
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, USA
| |
Collapse
|
115
|
Pigna E, Sanna K, Coletti D, Li Z, Parlakian A, Adamo S, Moresi V. Increasing autophagy does not affect neurogenic muscle atrophy. Eur J Transl Myol 2018; 28:7687. [PMID: 30344980 PMCID: PMC6176397 DOI: 10.4081/ejtm.2018.7687] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/11/2018] [Accepted: 08/10/2018] [Indexed: 11/23/2022] Open
Abstract
Physiological autophagy plays a crucial role in the regulation of muscle mass and metabolism, while the excessive induction or the inhibition of the autophagic flux contributes to the progression of several diseases. Autophagy can be activated by different stimuli, including cancer, exercise, caloric restriction and denervation. The latter leads to muscle atrophy through the activation of catabolic pathways, i.e. the ubiquitin-proteasome system and autophagy. However, the kinetics of autophagy activation and the upstream molecular pathways in denervated skeletal muscle have not been reported yet. In this study, we characterized the kinetics of autophagic induction, quickly triggered by denervation, and report the Akt/mTOR axis activation. Besides, with the aim to assess the relative contribution of autophagy in neurogenic muscle atrophy, we triggered autophagy with different stimuli along with denervation, and observed that four week-long autophagic induction, by either intermitted fasting or rapamycin treatment, did not significantly affect muscle mass loss. We conclude that: i) autophagy does not play a major role in inducing muscle loss following denervation; ii) nonetheless, autophagy may have a regulatory role in denervation induced muscle atrophy, since it is significantly upregulated as early as eight hours after denervation; iii) Akt/mTOR axis, AMPK and FoxO3a are activated consistently with the progression of muscle atrophy, further highlighting the complexity of the signaling response to the atrophying stimulus deriving from denervation.
Collapse
Affiliation(s)
- Eva Pigna
- Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome, Italy
| | - Krizia Sanna
- Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome, Italy
| | - Dario Coletti
- Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome, Italy.,Interuniversity Institute of Myology, Italy.,Sorbonne Université, CNRS UMR 8256-INSERM ERL U1164, IBPS B2A Department of Biological Adaptation and Aging, Paris, France
| | - Zhenlin Li
- Sorbonne Université, CNRS UMR 8256-INSERM ERL U1164, IBPS B2A Department of Biological Adaptation and Aging, Paris, France
| | - Ara Parlakian
- Sorbonne Université, CNRS UMR 8256-INSERM ERL U1164, IBPS B2A Department of Biological Adaptation and Aging, Paris, France
| | | | - Viviana Moresi
- Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome, Italy.,Interuniversity Institute of Myology, Italy
| |
Collapse
|
116
|
Contribution of STAT3 to Inflammatory and Fibrotic Diseases and Prospects for its Targeting for Treatment. Int J Mol Sci 2018; 19:ijms19082299. [PMID: 30081609 PMCID: PMC6121470 DOI: 10.3390/ijms19082299] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/13/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023] Open
Abstract
Signal transducer and activator of transcription (STAT) 3 plays a central role in the host response to injury. It is activated rapidly within cells by many cytokines, most notably those in the IL-6 family, leading to pro-proliferative and pro-survival programs that assist the host in regaining homeostasis. With persistent activation, however, chronic inflammation and fibrosis ensue, leading to a number of debilitating diseases. This review summarizes advances in our understanding of the role of STAT3 and its targeting in diseases marked by chronic inflammation and/or fibrosis with a focus on those with the largest unmet medical need.
Collapse
|
117
|
Scicchitano BM, Dobrowolny G, Sica G, Musarò A. Molecular Insights into Muscle Homeostasis, Atrophy and Wasting. Curr Genomics 2018; 19:356-369. [PMID: 30065611 PMCID: PMC6030854 DOI: 10.2174/1389202919666180101153911] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Muscle homeostasis is guaranteed by a delicate balance between synthesis and degradation of cell proteins and its alteration leads to muscle wasting and diseases. In this review, we describe the major anabolic pathways that are involved in muscle growth and homeostasis and the proteolytic systems that are over-activated in muscle pathologies. Modulation of these pathways comprises an attractive target for drug intervention.
Collapse
Affiliation(s)
- Bianca Maria Scicchitano
- Istituto di Istologia e Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1-00168, Roma, Italy
| | - Gabriella Dobrowolny
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Gigliola Sica
- Istituto di Istologia e Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1-00168, Roma, Italy
| | - Antonio Musarò
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
118
|
mTOR and Tumor Cachexia. Int J Mol Sci 2018; 19:ijms19082225. [PMID: 30061533 PMCID: PMC6121479 DOI: 10.3390/ijms19082225] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 07/23/2018] [Accepted: 07/25/2018] [Indexed: 12/11/2022] Open
Abstract
Cancer cachexia affects most patients with advanced forms of cancers. It is mainly characterized by weight loss, due to muscle and adipose mass depletion. As cachexia is associated with increased morbidity and mortality in cancer patients, identifying the underlying mechanisms leading to cachexia is essential in order to design novel therapeutic strategies. The mechanistic target of rapamycin (mTOR) is a major intracellular signalling intermediary that participates in cell growth by upregulating anabolic processes such as protein and lipid synthesis. Accordingly, emerging evidence suggests that mTOR and mTOR inhibitors influence cancer cachexia. Here, we review the role of mTOR in cellular processes involved in cancer cachexia and highlight the studies supporting the contribution of mTOR in cancer cachexia.
Collapse
|
119
|
van der Ende M, Grefte S, Plas R, Meijerink J, Witkamp RF, Keijer J, van Norren K. Mitochondrial dynamics in cancer-induced cachexia. Biochim Biophys Acta Rev Cancer 2018; 1870:137-150. [PMID: 30059724 DOI: 10.1016/j.bbcan.2018.07.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/25/2018] [Accepted: 07/26/2018] [Indexed: 12/25/2022]
Abstract
Cancer-induced cachexia has a negative impact on quality of life and adversely affects therapeutic outcomes and survival rates. It is characterized by, often severe, loss of muscle, with or without loss of fat mass. Insight in the pathophysiology of this complex metabolic syndrome and direct treatment options are still limited, which creates a research demand. Results from recent studies point towards a significant involvement of muscle mitochondrial networks. However, data are scattered and a comprehensive overview is lacking. This paper aims to fill existing knowledge gaps by integrating published data sets on muscle protein or gene expression from cancer-induced cachexia animal models. To this end, a database was compiled from 94 research papers, comprising 11 different rodent models. This was combined with four genome-wide transcriptome datasets of cancer-induced cachexia rodent models. Analysis showed that the expression of genes involved in mitochondrial fusion, fission, ATP production and mitochondrial density is decreased, while that of genes involved ROS detoxification and mitophagy is increased. Our results underline the relevance of including post-translational modifications of key proteins involved in mitochondrial functioning in future studies on cancer-induced cachexia.
Collapse
Affiliation(s)
- Miranda van der Ende
- Division of Human Nutrition, Wageningen University and Research, Wageningen, Netherlands; Human and Animal Physiology, Wageningen University and Research, Wageningen, Netherlands
| | - Sander Grefte
- Human and Animal Physiology, Wageningen University and Research, Wageningen, Netherlands
| | - Rogier Plas
- Division of Human Nutrition, Wageningen University and Research, Wageningen, Netherlands
| | - Jocelijn Meijerink
- Division of Human Nutrition, Wageningen University and Research, Wageningen, Netherlands
| | - Renger F Witkamp
- Division of Human Nutrition, Wageningen University and Research, Wageningen, Netherlands
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University and Research, Wageningen, Netherlands
| | - Klaske van Norren
- Division of Human Nutrition, Wageningen University and Research, Wageningen, Netherlands.
| |
Collapse
|
120
|
Gouzi F, Blaquière M, Catteau M, Bughin F, Maury J, Passerieux E, Ayoub B, Mercier J, Hayot M, Pomiès P. Oxidative stress regulates autophagy in cultured muscle cells of patients with chronic obstructive pulmonary disease. J Cell Physiol 2018; 233:9629-9639. [PMID: 29943813 DOI: 10.1002/jcp.26868] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 05/23/2018] [Indexed: 12/21/2022]
Abstract
The proteolytic autophagy pathway is enhanced in the lower limb muscles of patients with chronic obstructive pulmonary disease (COPD). Reactive oxygen species (ROS) have been shown to regulate autophagy in the skeletal muscles, but the role of oxidative stress in the muscle autophagy of patients with COPD is unknown. We used cultured myoblasts and myotubes from the quadriceps of eight healthy subjects and twelve patients with COPD (FEV1% predicted: 102.0% and 32.0%, respectively; p < 0.0001). We compared the autophagosome formation, the expression of autophagy markers, and the autophagic flux in healthy subjects and the patients with COPD, and we evaluated the effects of the 3-methyladenine (3-MA) autophagy inhibitor on the atrophy of COPD myotubes. Autophagy was also assessed in COPD myotubes treated with an antioxidant molecule, ascorbic acid. Autophagosome formation was increased in COPD myoblasts and myotubes (p = 0.011; p < 0.001), and the LC3 2/LC3 1 ratio (p = 0.002), SQSTM1 mRNA and protein expression (p = 0.023; p = 0.007), BNIP3 expression (p = 0.031), and autophagic flux (p = 0.002) were higher in COPD myoblasts. Inhibition of autophagy with 3-MA increased the COPD myotube diameter (p < 0.001) to a level similar to the diameter of healthy subject myotubes. Treatment of COPD myotubes with ascorbic acid decreased ROS concentration (p < 0.001), ROS-induced protein carbonylation (p = 0.019), the LC3 2/LC3 1 ratio (p = 0.037), the expression of SQSTM1 (p < 0.001) and BNIP3 (p < 0.001), and increased the COPD myotube diameter (p < 0.001). Thus, autophagy signaling is enhanced in cultured COPD muscle cells. Furthermore, the oxidative stress level contributes to the regulation of autophagy, which is involved in the atrophy of COPD myotubes in vitro.
Collapse
Affiliation(s)
- Fares Gouzi
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Department of Clinical Physiology, CHRU of Montpellier, Montpellier, France
| | - Marine Blaquière
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Department of Clinical Physiology, CHRU of Montpellier, Montpellier, France
| | - Matthias Catteau
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - François Bughin
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Department of Clinical Physiology, CHRU of Montpellier, Montpellier, France
| | - Jonathan Maury
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Clinique du Souffle "La Solane," Fontalvie/5-Santé Group, Osséja, France
| | - Emilie Passerieux
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Bronia Ayoub
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Department of Clinical Physiology, CHRU of Montpellier, Montpellier, France
| | - Jacques Mercier
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Department of Clinical Physiology, CHRU of Montpellier, Montpellier, France
| | - Maurice Hayot
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.,Department of Clinical Physiology, CHRU of Montpellier, Montpellier, France
| | - Pascal Pomiès
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| |
Collapse
|
121
|
Wang D, Sun H, Song G, Yang Y, Zou X, Han P, Li S. Resveratrol Improves Muscle Atrophy by Modulating Mitochondrial Quality Control in STZ-Induced Diabetic Mice. Mol Nutr Food Res 2018; 62:e1700941. [PMID: 29578301 PMCID: PMC6001753 DOI: 10.1002/mnfr.201700941] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/10/2018] [Indexed: 12/14/2022]
Abstract
SCOPE In this study, we aim to determine the effects of resveratrol (RSV) on muscle atrophy in streptozocin-induced diabetic mice and to explore mitochondrial quality control (MQC) as a possible mechanism. METHODS AND RESULTS The experimental mice were fed either a control diet or an identical diet containing 0.04% RSV for 8 weeks. Examinations were subsequently carried out, including the effects of RSV on muscle atrophy and muscle function, as well as on the signaling pathways related to protein degradation and MQC processes. The results show that RSV supplementation improves muscle atrophy and muscle function, attenuates the increase in ubiquitin and muscle RING-finger protein-1 (MuRF-1), and simultaneously attenuates LC3-II and cleaved caspase-3 in the skeletal muscle of diabetic mice. Moreover, RSV treatment of diabetic mice results in an increase in mitochondrial biogenesis and inhibition of the activation of mitophagy in skeletal muscle. RSV also protects skeletal muscle against excess mitochondrial fusion and fission in the diabetic mice. CONCLUSION The results suggest that RSV ameliorates diabetes-induced skeletal muscle atrophy by modulating MQC.
Collapse
MESH Headings
- Animals
- Antioxidants/therapeutic use
- Apoptosis
- Autophagy
- Biomarkers/metabolism
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/physiopathology
- Dietary Supplements
- Gene Expression Regulation
- Male
- Mice, Inbred C57BL
- Microscopy, Electron, Transmission
- Mitochondria, Muscle/metabolism
- Mitochondria, Muscle/pathology
- Mitochondria, Muscle/ultrastructure
- Mitochondrial Dynamics
- Muscle Proteins/antagonists & inhibitors
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscle Strength
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiopathology
- Muscle, Skeletal/ultrastructure
- Muscular Atrophy/complications
- Muscular Atrophy/metabolism
- Muscular Atrophy/pathology
- Muscular Atrophy/prevention & control
- Muscular Disorders, Atrophic/complications
- Muscular Disorders, Atrophic/metabolism
- Muscular Disorders, Atrophic/pathology
- Muscular Disorders, Atrophic/prevention & control
- Resveratrol/therapeutic use
- Signal Transduction
- Streptozocin
- Tripartite Motif Proteins/antagonists & inhibitors
- Tripartite Motif Proteins/genetics
- Tripartite Motif Proteins/metabolism
- Ubiquitin/antagonists & inhibitors
- Ubiquitin/genetics
- Ubiquitin/metabolism
- Ubiquitin-Protein Ligases/antagonists & inhibitors
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
Collapse
Affiliation(s)
- Dongtao Wang
- Department of Traditional Chinese MedicineShenzhen HospitalSouthern Medical UniversityShenzhenGuangdong518000China
- Department of NephrologyShenzhen Traditional Chinese Medicine HospitalGuangzhou University of Chinese MedicineShenzhenGuangdong518033China
- Department of NephrologyRuikang Affiliated HospitalGuangxi University of Chinese MedicineNanning530011China
| | - Huili Sun
- Department of NephrologyShenzhen Traditional Chinese Medicine HospitalGuangzhou University of Chinese MedicineShenzhenGuangdong518033China
| | - Gaofeng Song
- Department of NephrologyShenzhen Traditional Chinese Medicine HospitalGuangzhou University of Chinese MedicineShenzhenGuangdong518033China
| | - Yajun Yang
- Department of PharmacologyGuangdong Key Laboratory for R&D of Natural DrugGuangdong Medical CollegeZhanjiang524023China
| | - Xiaohu Zou
- Department of Traditional Chinese MedicineShenzhen HospitalSouthern Medical UniversityShenzhenGuangdong518000China
| | - Pengxun Han
- Department of NephrologyShenzhen Traditional Chinese Medicine HospitalGuangzhou University of Chinese MedicineShenzhenGuangdong518033China
| | - Shunmin Li
- Department of NephrologyShenzhen Traditional Chinese Medicine HospitalGuangzhou University of Chinese MedicineShenzhenGuangdong518033China
| |
Collapse
|
122
|
Castets P, Frank S, Sinnreich M, Rüegg MA. "Get the Balance Right": Pathological Significance of Autophagy Perturbation in Neuromuscular Disorders. J Neuromuscul Dis 2018; 3:127-155. [PMID: 27854220 PMCID: PMC5271579 DOI: 10.3233/jnd-160153] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent research has revealed that autophagy, a major catabolic process in cells, is dysregulated in several neuromuscular diseases and contributes to the muscle wasting caused by non-muscle disorders (e.g. cancer cachexia) or during aging (i.e. sarcopenia). From there, the idea arose to interfere with autophagy or manipulate its regulatory signalling to help restore muscle homeostasis and attenuate disease progression. The major difficulty for the development of therapeutic strategies is to restore a balanced autophagic flux, due to the dynamic nature of autophagy. Thus, it is essential to better understand the mechanisms and identify the signalling pathways at play in the control of autophagy in skeletal muscle. A comprehensive analysis of the autophagic flux and of the causes of its dysregulation is required to assess the pathogenic role of autophagy in diseased muscle. Furthermore, it is essential that experiments distinguish between primary dysregulation of autophagy (prior to disease onset) and impairments as a consequence of the pathology. Of note, in most muscle disorders, autophagy perturbation is not caused by genetic modification of an autophagy-related protein, but rather through indirect alteration of regulatory signalling or lysosomal function. In this review, we will present the mechanisms involved in autophagy, and those ensuring its tight regulation in skeletal muscle. We will then discuss as to how autophagy dysregulation contributes to the pathogenesis of neuromuscular disorders and possible ways to interfere with this process to limit disease progression.
Collapse
Affiliation(s)
| | - Stephan Frank
- Institute of Pathology, Division of Neuropathology Basel University Hospital, Basel, Switzerland
| | - Michael Sinnreich
- Neuromuscular Research Center, Departments of Neurology and Biomedicine, Pharmazentrum, Basel, Switzerland
| | | |
Collapse
|
123
|
Parvy JP, Hodgson JA, Cordero JB. Drosophila as a Model System to Study Nonautonomous Mechanisms Affecting Tumour Growth and Cell Death. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7152962. [PMID: 29725601 PMCID: PMC5872677 DOI: 10.1155/2018/7152962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 02/04/2018] [Indexed: 12/26/2022]
Abstract
The study of cancer has represented a central focus in medical research for over a century. The great complexity and constant evolution of the pathology require the use of multiple research model systems and interdisciplinary approaches. This is necessary in order to achieve a comprehensive understanding into the mechanisms driving disease initiation and progression, to aid the development of appropriate therapies. In recent decades, the fruit fly Drosophila melanogaster and its associated powerful genetic tools have become a very attractive model system to study tumour-intrinsic and non-tumour-derived processes that mediate tumour development in vivo. In this review, we will summarize recent work on Drosophila as a model system to study cancer biology. We will focus on the interactions between tumours and their microenvironment, including extrinsic mechanisms affecting tumour growth and how tumours impact systemic host physiology.
Collapse
Affiliation(s)
- Jean-Philippe Parvy
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Joseph A. Hodgson
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Julia B. Cordero
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| |
Collapse
|
124
|
Kawanishi N, Funakoshi T, Machida S. Time-course study of macrophage infiltration and inflammation in cast immobilization-induced atrophied muscle of mice. Muscle Nerve 2018; 57:1006-1013. [DOI: 10.1002/mus.26061] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 01/04/2018] [Accepted: 01/06/2018] [Indexed: 01/07/2023]
Affiliation(s)
- Noriaki Kawanishi
- Graduate School of Health and Sports Science; Juntendo University; 1-1 Hirakagakuendai, Inzai 270-1695 Japan
- Japan Society for the Promotion of Sciences; Tokyo Japan
- Institute of Health & Sports Science and Medicine; Juntendo University; Inzai Japan
- Faculty of Advanced Engineering, Chiba Institute of Technology; Narashino Japan
| | - Tomoko Funakoshi
- Graduate School of Health and Sports Science; Juntendo University; 1-1 Hirakagakuendai, Inzai 270-1695 Japan
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology; Tokyo Japan
| | - Shuichi Machida
- Graduate School of Health and Sports Science; Juntendo University; 1-1 Hirakagakuendai, Inzai 270-1695 Japan
- Institute of Health & Sports Science and Medicine; Juntendo University; Inzai Japan
| |
Collapse
|
125
|
Modulating Metabolism to Improve Cancer-Induced Muscle Wasting. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7153610. [PMID: 29785246 PMCID: PMC5896402 DOI: 10.1155/2018/7153610] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/25/2017] [Indexed: 12/13/2022]
Abstract
Muscle wasting is one of the main features of cancer cachexia, a multifactorial syndrome frequently occurring in oncologic patients. The onset of cachexia is associated with reduced tolerance and response to antineoplastic treatments, eventually leading to clinical conditions that are not compatible with survival. Among the mechanisms underlying cachexia, protein and energy dysmetabolism play a major role. In this regard, several potential treatments have been proposed, mainly on the basis of promising results obtained in preclinical models. However, at present, no treatment yet reached validation to be used in the clinical practice, although several drugs are currently tested in clinical trials for their ability to improve muscle metabolism in cancer patients. Along this line, the results obtained in both experimental and clinical studies clearly show that cachexia can be effectively approached by a multidirectional strategy targeting nutrition, inflammation, catabolism, and inactivity at the same time. In the present study, approaches aimed to modulate muscle metabolism in cachexia will be reviewed.
Collapse
|
126
|
Barreto R, Waning DL, Gao H, Liu Y, Zimmers TA, Bonetto A. Chemotherapy-related cachexia is associated with mitochondrial depletion and the activation of ERK1/2 and p38 MAPKs. Oncotarget 2017; 7:43442-43460. [PMID: 27259276 PMCID: PMC5190036 DOI: 10.18632/oncotarget.9779] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 05/16/2016] [Indexed: 12/21/2022] Open
Abstract
Cachexia affects the majority of cancer patients, with currently no effective treatments. Cachexia is defined by increased fatigue and loss of muscle function resulting from muscle and fat depletion. Previous studies suggest that chemotherapy may contribute to cachexia, although the causes responsible for this association are not clear. The purpose of this study was to investigate the mechanism(s) associated with chemotherapy-related effects on body composition and muscle function. Normal mice were administered chemotherapy regimens used for the treatment of colorectal cancer, such as Folfox (5-FU, leucovorin, oxaliplatin) or Folfiri (5-FU, leucovorin, irinotecan) for 5 weeks. The animals that received chemotherapy exhibited concurrent loss of muscle mass and muscle weakness. Consistently with previous findings, muscle wasting was associated with up-regulation of ERK1/2 and p38 MAPKs. No changes in ubiquitin-dependent proteolysis or in the expression of TGFβ-family members were detected. Further, marked decreases in mitochondrial content, associated with abnormalities at the sarcomeric level and with increase in the number of glycolytic fibers were observed in the muscle of mice receiving chemotherapy. Finally, ACVR2B/Fc or PD98059 prevented Folfiri-associated ERK1/2 activation and myofiber atrophy in C2C12 cultures. Our findings demonstrate that chemotherapy promotes MAPK-dependent muscle atrophy as well as mitochondrial depletion and alterations of the sarcomeric units. Therefore, these findings suggest that chemotherapy potentially plays a causative role in the occurrence of muscle loss and weakness. Moreover, the present observations provide a strong rationale for testing ACVR2B/Fc or MEK1 inhibitors in combination with anticancer drugs as novel strategies aimed at preventing chemotherapy-associated muscle atrophy.
Collapse
Affiliation(s)
- Rafael Barreto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - David L Waning
- Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,IUPUI Center for Cachexia Research, Innovation and Therapy, Indianapolis, IN 46202, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Teresa A Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,IUPUI Center for Cachexia Research, Innovation and Therapy, Indianapolis, IN 46202, USA
| | - Andrea Bonetto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,IUPUI Center for Cachexia Research, Innovation and Therapy, Indianapolis, IN 46202, USA
| |
Collapse
|
127
|
Molinari F, Pin F, Gorini S, Chiandotto S, Pontecorvo L, Penna F, Rizzuto E, Pisu S, Musarò A, Costelli P, Rosano G, Ferraro E. The mitochondrial metabolic reprogramming agent trimetazidine as an 'exercise mimetic' in cachectic C26-bearing mice. J Cachexia Sarcopenia Muscle 2017; 8:954-973. [PMID: 29130633 PMCID: PMC5700442 DOI: 10.1002/jcsm.12226] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/07/2017] [Accepted: 07/05/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cancer cachexia is characterized by muscle depletion and exercise intolerance caused by an imbalance between protein synthesis and degradation and by impaired myogenesis. Myofibre metabolic efficiency is crucial so as to assure optimal muscle function. Some drugs are able to reprogram cell metabolism and, in some cases, to enhance metabolic efficiency. Based on these premises, we chose to investigate the ability of the metabolic modulator trimetazidine (TMZ) to counteract skeletal muscle dysfunctions and wasting occurring in cancer cachexia. METHODS For this purpose, we used mice bearing the C26 colon carcinoma as a model of cancer cachexia. Mice received 5 mg/kg TMZ (i.p.) once a day for 12 consecutive days. A forelimb grip strength test was performed and tibialis anterior, and gastrocnemius muscles were excised for analysis. Ex vivo measurement of skeletal muscle contractile properties was also performed. RESULTS Our data showed that TMZ induces some effects typically achieved through exercise, among which is grip strength increase, an enhanced fast-to slow myofibre phenotype shift, reduced glycaemia, PGC1α up-regulation, oxidative metabolism, and mitochondrial biogenesis. TMZ also partially restores the myofibre cross-sectional area in C26-bearing mice, while modulation of autophagy and apoptosis were excluded as mediators of TMZ effects. CONCLUSIONS In conclusion, our data show that TMZ acts like an 'exercise mimetic' and is able to enhance some mechanisms of adaptation to stress in cancer cachexia. This makes the modulation of the metabolism, and in particular TMZ, a suitable candidate for a therapeutic rehabilitative protocol design, particularly considering that TMZ has already been approved for clinical use.
Collapse
Affiliation(s)
- Francesca Molinari
- Laboratory of Pathophysiology of Cachexia and Metabolism of Skeletal Muscle, IRCCS San Raffaele Pisana, Via di Val Cannuta, 00166, Rome, Italy
| | - Fabrizio Pin
- Department of Clinical and Biological Sciences, IIM, University of Turin, Corso Raffaello, 10125, Turin, Italy
| | - Stefania Gorini
- Laboratory of Pathophysiology of Cachexia and Metabolism of Skeletal Muscle, IRCCS San Raffaele Pisana, Via di Val Cannuta, 00166, Rome, Italy
| | - Sergio Chiandotto
- DMCM Department of Molecular and Clinical Medicine, c/o Department of Surgery 'Pietro Valdoni', Sapienza University of Rome, Via Scarpa, 00161, Rome, Italy
| | - Laura Pontecorvo
- Laboratory of Pathophysiology of Cachexia and Metabolism of Skeletal Muscle, IRCCS San Raffaele Pisana, Via di Val Cannuta, 00166, Rome, Italy
| | - Fabio Penna
- Department of Clinical and Biological Sciences, IIM, University of Turin, Corso Raffaello, 10125, Turin, Italy
| | - Emanuele Rizzuto
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Via Eudossiana, 00184, Rome, Italy
| | - Simona Pisu
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM, Sapienza University of Rome, Via Scarpa, 00161, Rome, Italy
| | - Antonio Musarò
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM, Sapienza University of Rome, Via Scarpa, 00161, Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena, 00161, Rome, Italy
| | - Paola Costelli
- Department of Clinical and Biological Sciences, IIM, University of Turin, Corso Raffaello, 10125, Turin, Italy
| | - Giuseppe Rosano
- Laboratory of Pathophysiology of Cachexia and Metabolism of Skeletal Muscle, IRCCS San Raffaele Pisana, Via di Val Cannuta, 00166, Rome, Italy.,Cardiovascular and Cell Sciences Institute, St George's University of London, Cranmer Terrace London, SW17, UK
| | - Elisabetta Ferraro
- Laboratory of Pathophysiology of Cachexia and Metabolism of Skeletal Muscle, IRCCS San Raffaele Pisana, Via di Val Cannuta, 00166, Rome, Italy
| |
Collapse
|
128
|
Segatto M, Fittipaldi R, Pin F, Sartori R, Dae Ko K, Zare H, Fenizia C, Zanchettin G, Pierobon ES, Hatakeyama S, Sperti C, Merigliano S, Sandri M, Filippakopoulos P, Costelli P, Sartorelli V, Caretti G. Epigenetic targeting of bromodomain protein BRD4 counteracts cancer cachexia and prolongs survival. Nat Commun 2017; 8:1707. [PMID: 29167426 PMCID: PMC5700099 DOI: 10.1038/s41467-017-01645-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 10/05/2017] [Indexed: 02/08/2023] Open
Abstract
Cancer cachexia is a devastating metabolic syndrome characterized by systemic inflammation and massive muscle and adipose tissue wasting. Although it is responsible for approximately one-third of cancer deaths, no effective therapies are available and the underlying mechanisms have not been fully elucidated. We previously identified the bromodomain and extra-terminal domain (BET) protein BRD4 as an epigenetic regulator of muscle mass. Here we show that the pan-BET inhibitor (+)-JQ1 protects tumor-bearing mice from body weight loss and muscle and adipose tissue wasting. Remarkably, in C26-tumor-bearing mice (+)-JQ1 administration dramatically prolongs survival, without directly affecting tumor growth. By ChIP-seq and ChIP analyses, we unveil that BET proteins directly promote the muscle atrophy program during cachexia. In addition, BET proteins are required to coordinate an IL6-dependent AMPK nuclear signaling pathway converging on FoxO3 transcription factor. Overall, these findings indicate that BET proteins may represent a promising therapeutic target in the management of cancer cachexia.
Collapse
Affiliation(s)
- Marco Segatto
- Department of Biosciences, Universita' degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Raffaella Fittipaldi
- Department of Biosciences, Universita' degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Fabrizio Pin
- Department of Clinical and Biological Sciences, Unit of General and Clinical Pathology, University of Turin, 10124, Torino, Italy
| | - Roberta Sartori
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
- Venetian Institute of Molecular Medicine, 35131, Padova, Italy
| | - Kyung Dae Ko
- Laboratory of Muscle Stem Cells and Gene Regulation, NIH/NIAMS, 50 South Drive, Bethesda, MD, USA
| | - Hossein Zare
- Laboratory of Muscle Stem Cells and Gene Regulation, NIH/NIAMS, 50 South Drive, Bethesda, MD, USA
| | - Claudio Fenizia
- Department of Biosciences, Universita' degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Gianpietro Zanchettin
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padua, 35122, Padova, Italy
| | - Elisa Sefora Pierobon
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padua, 35122, Padova, Italy
| | - Shinji Hatakeyama
- Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research Basel, Novartis Pharma AG, 4056, Basel, Switzerland
| | - Cosimo Sperti
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padua, 35122, Padova, Italy
| | - Stefano Merigliano
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padua, 35122, Padova, Italy
| | - Marco Sandri
- Venetian Institute of Molecular Medicine, 35131, Padova, Italy
- Laboratory of Muscle Stem Cells and Gene Regulation, NIH/NIAMS, 50 South Drive, Bethesda, MD, USA
| | - Panagis Filippakopoulos
- Structural Genomics Consortium, Old Road Campus Research Building, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ, UK
- Ludwig Institute for Cancer Research, Old Road Campus Research Building, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ, UK
| | - Paola Costelli
- Department of Clinical and Biological Sciences, Unit of General and Clinical Pathology, University of Turin, 10124, Torino, Italy
| | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, NIH/NIAMS, 50 South Drive, Bethesda, MD, USA
| | - Giuseppina Caretti
- Department of Biosciences, Universita' degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
129
|
MRI-defined paraspinal muscle morphology in Japanese population: The Wakayama Spine Study. PLoS One 2017; 12:e0187765. [PMID: 29117256 PMCID: PMC5678698 DOI: 10.1371/journal.pone.0187765] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 10/25/2017] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE This study aimed to establish sex- and age-dependent distributions of the cross sectional area and fatty infiltration ratio of paraspinal muscles, and to examine the correlation between paraspinal muscle degeneration and low back pain in the Japanese population. METHODS In this cross-sectional study, data from 796 participants (241 men, 555 women; mean age, 63.5 years) were analyzed. The measurement of the cross sectional area and fatty infiltration ratio of the erector spinae and multifidus from the level of T12/L1 to L4/5 and psoas major at the level of T12/L1 was performed using axial T2-weighted magnetic resonance imaging. Multivariate logistic regression analysis was used to estimate the association between fatty infiltration of the paraspinal muscles and the prevalence of low back pain. RESULTS The cross sectional area was larger in men than women, and tended to decrease with age, with the exception of the erector spinae at T12/L1 and L1/2 in women. The fatty infiltration ratio was lower in men than women, except for multifidus at T12/L1 in 70-79 year-olds and psoas major in those less than 50 years-old, and tended to increase with age. Logistic regression analysis adjusted for age, sex, and body mass index showed that the fatty infiltration ratio of the erector spinae at L1/2 and L2/3 was significantly associated with low back pain (L1/2 level: odds ratio, 1.05; 95% confidence interval, 1.005-1.104; L2/3 level: odds ratio, 1.05; 95% confidence interval, 1.001-1.113). CONCLUSION This study measured the cross sectional area and fatty infiltration ratio of paraspinal muscles in the Japanese population using magnetic resonance imaging, and demonstrated that the fatty infiltration ratio of the erector spinae in the upper lumbar spine was significantly associated with the presence of low back pain. The measurements could be used as reference values, which are important for future comparative studies.
Collapse
|
130
|
Fujiwara M, Iwata M, Inoue T, Aizawa Y, Yoshito N, Hayashi K, Suzuki S. Decreased grip strength, muscle pain, and atrophy occur in rats following long-term exposure to excessive repetitive motion. FEBS Open Bio 2017; 7:1737-1749. [PMID: 29123982 PMCID: PMC5666401 DOI: 10.1002/2211-5463.12315] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/31/2017] [Accepted: 09/05/2017] [Indexed: 12/22/2022] Open
Abstract
Work‐related musculoskeletal disorders (WMSD) are caused by the overuse of muscles in the workplace. Performing repetitive tasks is a primary risk factor for the development of WMSD. Many workers in highly repetitive jobs exhibit muscle pain and decline in handgrip strength, yet the mechanisms underlying these dysfunctions are poorly understood. In our study, rats performed voluntary repetitive reaching and grasping tasks (Task group), while Control group rats did not perform these activities. In the Task group, grip strength and forearm flexor withdrawal threshold declined significantly from week 2 to week 6, compared with these values at week 0 (P < 0.05). Relative muscle weight and muscle fiber cross‐sectional area of flexor digitorum superficialis (FDS) muscles decreased significantly in the Task group, compared with the Control group, at 6 weeks (P < 0.05 and P < 0.01, respectively). Nerve growth factor, glial cell line‐derived neurotrophic factor, and tumor necrosis factor α‐expression in FDS muscles were not significantly different in Control and Task groups at 3 and 6 weeks. At 6 weeks, the Task group had elevated MuRF1 protein levels (P = 0.065) and significant overexpression of the autophagy‐related (Atg) proteins, Beclin1 and Atg5–Atg12, compared with in the Control group (both P < 0.05). These data suggested that long‐term exposure to excessive repetitive motion causes loss of grip strength, muscle pain, and skeletal muscle atrophy. Furthermore, this exposure may enhance protein degradation through both the ubiquitin‐proteasome and autophagy‐lysosome systems, thereby decreasing skeletal muscle mass.
Collapse
Affiliation(s)
- Mitsuhiro Fujiwara
- Program in Physical and Occupational Therapy Nagoya University Graduate School of Medicine Japan.,Department of Rehabilitation Kamiiida Rehabilitation Hospital Nagoya Japan
| | - Masahiro Iwata
- Program in Physical and Occupational Therapy Nagoya University Graduate School of Medicine Japan.,Department of Rehabilitation Faculty of Health Sciences Nihon Fukushi University Handa Aichi Japan
| | - Takayuki Inoue
- Department of Rehabilitation Nagoya University Hospital Japan
| | - Yosuke Aizawa
- Department of Rehabilitation Japanese Red Cross Nagoya Daiichi Hospital Nagoya Japan
| | - Natsumi Yoshito
- Department of Rehabilitation Nagoya City University Hospital Japan
| | - Kazuhiro Hayashi
- Multidisciplinary Pain Center Aichi Medical University Nagakute Aichi Japan
| | - Shigeyuki Suzuki
- Program in Physical and Occupational Therapy Nagoya University Graduate School of Medicine Japan
| |
Collapse
|
131
|
Zeng X, Chen S, Yang Y, Ke Z. Acylated and unacylated ghrelin inhibit atrophy in myotubes co-cultured with colon carcinoma cells. Oncotarget 2017; 8:72872-72885. [PMID: 29069832 PMCID: PMC5641175 DOI: 10.18632/oncotarget.20531] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/30/2017] [Indexed: 01/06/2023] Open
Abstract
Cancer cachexia is a result of increased protein degradation and decreased protein synthesis. The multifunctional circulating hormone ghrelin promotes synthesis and inhibits degradation of muscle protein, but its mechanism of action is not fully understood. Here, we investigated whether co-culturing C2C12 myotubes with CT26 colon carcinoma cells induces myotube atrophy, and whether acylated ghrelin (AG) and unacylated ghrelin (UnAG) had anti-atrophic effects. We found that co-culture induced myotube atrophy and increased tumor necrosis factor-alpha (TNF-α) and myostatin concentrations in the culture medium. Moreover, co-culture down-regulated myogenin and MyoD expression, inhibited the Akt signaling, up-regulated ubiquitin E3 ligase expression, and activated the calpain system and autophagy in myotubes. Both AG and UnAG inhibited these changes. Our study describes a novel in vitro model that can be employed to investigate cancer cachexia, and our findings suggest a possible use for AG and UnAG in treating cancer cachexia.
Collapse
Affiliation(s)
- Xianliang Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Sizeng Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Yang Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Zhao Ke
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| |
Collapse
|
132
|
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: 125] [Impact Index Per Article: 15.6] [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
|
133
|
Toll-like receptor 4 mediates Lewis lung carcinoma-induced muscle wasting via coordinate activation of protein degradation pathways. Sci Rep 2017; 7:2273. [PMID: 28536426 PMCID: PMC5442131 DOI: 10.1038/s41598-017-02347-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/10/2017] [Indexed: 12/22/2022] Open
Abstract
Cancer-induced cachexia, characterized by muscle wasting, is a lethal metabolic syndrome with undefined etiology. Current consensus is that multiple factors contribute to cancer-induced muscle wasting, and therefore therapy requires combinational strategies. Here, we show that Toll-like receptor 4 (TLR4) mediates cancer-induced muscle wasting by directly activating muscle catabolism as well as stimulating an innate immune response in mice bearing Lewis lung carcinoma (LLC), and targeting TLR4 alone effectively abrogate muscle wasting. Utilizing specific siRNAs we observed that LLC cell-conditioned medium (LCM)-treated C2C12 myotubes underwent a rapid catabolic response in a TLR4-dependent manner, including activation of the p38 MAPK−C/EBPβ signaling pathway as well as the ubiquitin-proteasome and autophagy-lysosome pathways, resulting in myotube atrophy. Utilizing a reporter cell-line it was confirmed that LCM activated TLR4. These results suggest that LLC-released cachexins directly activate muscle catabolism via activating TLR4 on muscle cells independent of immune responses. Critically, LLC tumor-bearing TLR4−/− mice were spared from muscle wasting due to a blockade in muscle catabolic pathways. Further, tumor-induced elevation of circulating TNFα and interleukin-6 (IL-6) was abolished in TLR4−/− mice. These data suggest that TLR4 is a central mediator and therapeutic target of cancer-induced muscle wasting.
Collapse
|
134
|
Pettersen K, Andersen S, Degen S, Tadini V, Grosjean J, Hatakeyama S, Tesfahun AN, Moestue S, Kim J, Nonstad U, Romundstad PR, Skorpen F, Sørhaug S, Amundsen T, Grønberg BH, Strasser F, Stephens N, Hoem D, Molven A, Kaasa S, Fearon K, Jacobi C, Bjørkøy G. Cancer cachexia associates with a systemic autophagy-inducing activity mimicked by cancer cell-derived IL-6 trans-signaling. Sci Rep 2017; 7:2046. [PMID: 28515477 PMCID: PMC5435723 DOI: 10.1038/s41598-017-02088-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 04/05/2017] [Indexed: 12/20/2022] Open
Abstract
The majority of cancer patients with advanced disease experience weight loss, including loss of lean body mass. Severe weight loss is characteristic for cancer cachexia, a condition that significantly impairs functional status and survival. The underlying causes of cachexia are incompletely understood, and currently no therapeutic approach can completely reverse the condition. Autophagy coordinates lysosomal destruction of cytosolic constituents and is systemically induced by starvation. We hypothesized that starvation-mimicking signaling compounds secreted from tumor cells may cause a systemic acceleration of autophagy during cachexia. We found that IL-6 secreted by tumor cells accelerates autophagy in myotubes when complexed with soluble IL-6 receptor (trans-signaling). In lung cancer patients, were cachexia is prevalent, there was a significant correlation between elevated IL-6 expression in the tumor and poor prognosis of the patients. We found evidence for an autophagy-inducing bioactivity in serum from cancer patients and that this is clearly associated with weight loss. Importantly, the autophagy-inducing bioactivity was reduced by interference with IL-6 trans-signaling. Together, our findings suggest that IL-6 trans-signaling may be targeted in cancer cachexia.
Collapse
Affiliation(s)
- Kristine Pettersen
- Department of Medical Laboratory Technology, Faculty of Natural Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway.,Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway
| | - Sonja Andersen
- Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway
| | - Simone Degen
- Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research Basel, Novartis Pharma AG, 4056, Basel, Switzerland
| | - Valentina Tadini
- Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research Basel, Novartis Pharma AG, 4056, Basel, Switzerland
| | - Joël Grosjean
- Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research Basel, Novartis Pharma AG, 4056, Basel, Switzerland
| | - Shinji Hatakeyama
- Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research Basel, Novartis Pharma AG, 4056, Basel, Switzerland
| | - Almaz N Tesfahun
- Department of Medical Laboratory Technology, Faculty of Natural Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway.,Department of Laboratory Medicine, Children's and Women's Health, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Siver Moestue
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Jana Kim
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Unni Nonstad
- Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway
| | - Pål R Romundstad
- Department of Public Health and General Practice, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Frank Skorpen
- Department of Laboratory Medicine, Children's and Women's Health, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway.,European Palliative Care Research Centre, Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway
| | - Sveinung Sørhaug
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway.,Department of Thoracic Medicine, St.Olavs Hospital - Trondheim University Hospital, 7006, Trondheim, Norway
| | - Tore Amundsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway.,Department of Thoracic Medicine, St.Olavs Hospital - Trondheim University Hospital, 7006, Trondheim, Norway
| | - Bjørn H Grønberg
- The Cancer Clinic, St.Olavs Hospital - Trondheim University Hospital, 7030, Trondheim, Norway.,Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Florian Strasser
- Oncological Palliative Medicine, Division ofClinic Oncology/Hematology, Department of Internal Medicine and Palliative Care Center, Cantonal Hospital, St. Gallen, Switzerland
| | - Nathan Stephens
- Clinical and Surgical Sciences, School of Clinical Sciences and Community Health, The University of Edinburgh, Royal Infirmary, N-5021, Edinburgh, UK
| | - Dag Hoem
- Department of Gastrointestinal Surgery, Haukeland University Hospital, N-5020, Bergen, Norway
| | - Anders Molven
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5021, Bergen, Norway
| | - Stein Kaasa
- European Palliative Care Research Centre, Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway.,The Cancer Clinic, St.Olavs Hospital - Trondheim University Hospital, 7030, Trondheim, Norway
| | - Kenneth Fearon
- European Palliative Care Research Centre, Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway.,Clinical and Surgical Sciences, School of Clinical Sciences and Community Health, The University of Edinburgh, Royal Infirmary, N-5021, Edinburgh, UK
| | - Carsten Jacobi
- Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research Basel, Novartis Pharma AG, 4056, Basel, Switzerland.
| | - Geir Bjørkøy
- Department of Medical Laboratory Technology, Faculty of Natural Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway. .,Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway.
| |
Collapse
|
135
|
Bohnert KR, McMillan JD, Kumar A. Emerging roles of ER stress and unfolded protein response pathways in skeletal muscle health and disease. J Cell Physiol 2017; 233:67-78. [PMID: 28177127 DOI: 10.1002/jcp.25852] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 02/07/2017] [Indexed: 12/17/2022]
Abstract
Skeletal muscle is the most abundant tissue in the human body and can adapt its mass as a consequence of physical activity, metabolism, growth factors, and disease conditions. Skeletal muscle contains an extensive network of endoplasmic reticulum (ER), called sarcoplasmic reticulum, which plays an important role in the regulation of proteostasis and calcium homeostasis. In many cell types, environmental and genetic factors that disrupt ER function cause an accumulation of misfolded and unfolded proteins in the ER lumen that ultimately leads to ER stress. To alleviate the stress and restore homeostasis, the ER activates a signaling network called the unfolded protein response (UPR). The UPR has three arms, which regulate protein synthesis and expression of many ER chaperone and regulatory proteins. However, the role of individual UPR pathways in skeletal muscle has just begun to be investigated. Recent studies suggest that UPR pathways play pivotal roles in muscle stem cell homeostasis, myogenic differentiation, and regeneration of injured skeletal muscle. Moreover, markers of ER stress and the UPR are activated in skeletal muscle in diverse conditions such as exercise, denervation, starvation, high fat diet, cancer cachexia, and aging. Accumulating evidence also suggests that ER stress may have important roles in the pathogenesis of inflammatory myopathies and genetic muscle disorders. The purpose of this review article is to discuss the role and potential mechanisms by which ER stress and the individual arms of the UPR regulate skeletal muscle formation, plasticity, and function in various physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Kyle R Bohnert
- Department of Anatomical Sciences Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Joseph D McMillan
- Department of Anatomical Sciences Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Ashok Kumar
- Department of Anatomical Sciences Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
136
|
Leitner LM, Wilson RJ, Yan Z, Gödecke A. Reactive Oxygen Species/Nitric Oxide Mediated Inter-Organ Communication in Skeletal Muscle Wasting Diseases. Antioxid Redox Signal 2017; 26:700-717. [PMID: 27835923 PMCID: PMC5421600 DOI: 10.1089/ars.2016.6942] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Cachexia is defined as a complex metabolic syndrome that is associated with underlying illness and a loss of muscle with or without loss of fat mass. This disease is associated with a high incidence with chronic diseases such as heart failure, cancer, chronic obstructive pulmonary disease (COPD), and acquired immunodeficiency syndrome (AIDS), among others. Since there is currently no effective treatment available, cachectic patients have a poor prognosis. Elucidation of the underlying mechanisms is, therefore, an important medical task. Recent Advances: There is accumulating evidence that the diseased organs such as heart, lung, kidney, or cancer tissue secrete soluble factors, including Angiotensin II, myostatin (growth differentiation factor 8 [GDF8]), GDF11, tumor growth factor beta (TGFβ), which act on skeletal muscle. There, they induce a set of genes called atrogenes, which, among others, induce the ubiquitin-proteasome system, leading to protein degradation. Moreover, elevated reactive oxygen species (ROS) levels due to modulation of NADPH oxidases (Nox) and mitochondrial function contribute to disease progression, which is characterized by loss of muscle mass, exercise resistance, and frailty. CRITICAL ISSUES Although substantial progress was achieved to elucidate the pathophysiology of cachexia, effectice therapeutic strategies are urgently needed. FUTURE DIRECTIONS With the identification of key components of the aberrant inter-organ communication leading to cachexia, studies in mice and men to inhibit ROS formation, induction of anti-oxidative superoxide dismutases, and upregulation of muscular nitric oxide (NO) formation either by pharmacological tools or by exercise are promising approaches to reduce the extent of skeletal muscle wasting. Antioxid. Redox Signal. 26, 700-717.
Collapse
Affiliation(s)
- Lucia M Leitner
- 1 Institut für Herz- und Kreislaufphysiologie, Heinrich-Heine-Universität Düsseldorf, Universitätsklinikum , Düsseldorf, Germany
| | - Rebecca J Wilson
- 2 Department of Medicine-Cardiovascular Medicine, University of Virginia , Charlottesville, Virginia
| | - Zhen Yan
- 2 Department of Medicine-Cardiovascular Medicine, University of Virginia , Charlottesville, Virginia.,3 Center for Skeletal Muscle Research at Robert Berne Cardiovascular Research Center, University of Virginia , Charlottesville, Virginia
| | - Axel Gödecke
- 1 Institut für Herz- und Kreislaufphysiologie, Heinrich-Heine-Universität Düsseldorf, Universitätsklinikum , Düsseldorf, Germany
| |
Collapse
|
137
|
Pin F, Minero VG, Penna F, Muscaritoli M, De Tullio R, Baccino FM, Costelli P. Interference with Ca 2+-Dependent Proteolysis Does Not Alter the Course of Muscle Wasting in Experimental Cancer Cachexia. Front Physiol 2017; 8:213. [PMID: 28469577 PMCID: PMC5395607 DOI: 10.3389/fphys.2017.00213] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 03/23/2017] [Indexed: 01/24/2023] Open
Abstract
Protein hypercatabolism significantly contributes to the onset and progression of muscle wasting in cancer cachexia. In this regard, a major role is played by the ATP-ubiquitin-proteasome-dependent pathway and by autophagy. However, little is known about the relevance of the Ca2+-dependent proteolytic system. Since previous results suggested that this pathway is activated in the skeletal muscle of tumor hosts, the present study was aimed to investigate whether inhibition of Ca2+-dependent proteases (calpains) may improve cancer-induced muscle wasting. Two experimental models of cancer cachexia were used, namely the AH-130 Yoshida hepatoma and the C26 colon carcinoma. The Ca2+-dependent proteolytic system was inhibited by treating the animals with dantrolene or by overexpressing in the muscle calpastatin, the physiologic inhibitor of Ca2+-dependent proteases. The results confirm that calpain-1 is overexpressed and calpastatin is reduced in the muscle of rats implanted with the AH-130 hepatoma, and show for the first time that the Ca2+-dependent proteolytic system is overactivated also in the C26-bearing mice. Yet, administration of dantrolene, an inhibitor of the Ca2+-dependent proteases, did not modify tumor-induced body weight loss and muscle wasting in the AH-130 hosts. Dantrolene was also unable to reduce the enhancement of protein degradation rates occurring in rats bearing the AH-130 hepatoma. Similarly, overexpression of calpastatin in the tibialis muscle of the C26 hosts did not improve muscle wasting at all. These observations suggest that inhibiting a single proteolytic system is not a good strategy to contrast cancer-induced muscle wasting. In this regard, a more general and integrated approach aimed at targeting the catabolic stimuli rather than the proteolytic activity of a single pathway would likely be the most appropriate therapeutic intervention.
Collapse
Affiliation(s)
- Fabrizio Pin
- Department of Clinical and Biological Sciences, University of TurinTurin, Italy
| | - Valerio G Minero
- Department of Clinical and Biological Sciences, University of TurinTurin, Italy
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of TurinTurin, Italy
| | | | - Roberta De Tullio
- Department of Experimental Medicine, University of GenovaGenova, Italy
| | - Francesco M Baccino
- Department of Clinical and Biological Sciences, University of TurinTurin, Italy
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of TurinTurin, Italy
| |
Collapse
|
138
|
Su Z, Klein JD, Du J, Franch HA, Zhang L, Hassounah F, Hudson MB, Wang XH. Chronic kidney disease induces autophagy leading to dysfunction of mitochondria in skeletal muscle. Am J Physiol Renal Physiol 2017; 312:F1128-F1140. [PMID: 28381463 PMCID: PMC5495886 DOI: 10.1152/ajprenal.00600.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/04/2017] [Accepted: 04/04/2017] [Indexed: 01/07/2023] Open
Abstract
Chronic kidney disease (CKD) causes loss of lean body mass by multiple mechanisms. This study examines whether autophagy-mediated proteolysis contributes to CKD-induced muscle wasting. We tested autophagy in the muscle of CKD mice with plantaris muscle overloading to mimic resistance exercise or with acupuncture plus low-frequency electrical stimulation (Acu/LFES) treatment. In CKD muscle, Bnip3, Beclin-1, and LC3II mRNAs and proteins were increased compared with those in control muscle, indicating autophagosome-lysosome formation induction. Acu/LFES suppressed the CKD-induced upregulation of autophagy. However, overloading increased autophagy-related proteins in normal and CKD muscle. Serum from uremic mice induces autophagy formation but did not increase the myosin degradation or actin break down in cultured muscle satellite cells. We examined mitochondrial biogenesis, copy number, and ATP production in cultured myotubes, and found all three aspects to be decreased by uremic serum. Inhibition of autophagy partially reversed this decline in cultured myotubes. In CKD mice, the mitochondrial copy number, biogenesis marker peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), mitochondrial transcription factor A (TFAM), and mitochondrial fusion marker Mitofusin-2 (Mfn2) are decreased. Both muscle overloading and Acu/LFES increased mitochondrial copy number, and reversed the CKD-induced decreases in PGC-1α, TFAM, and Mfn2. We conclude that the autophagy is activated in the muscle of CKD mice. However, myofibrillar protein is not directly broken down through autophagy. Instead, CKD-induced upregulation of autophagy leads to dysfunction of mitochondria and decrease of ATP production.
Collapse
Affiliation(s)
- Zhen Su
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Janet D Klein
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Jie Du
- The Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing Anzhen Hospital Affiliated with Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Harold A Franch
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Liping Zhang
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas; and
| | - Faten Hassounah
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Matthew B Hudson
- Department of Kinesiology, Temple University, Philadelphia, Pennsylvania.,Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Xiaonan H Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
139
|
Ding H, Zhang G, Sin KWT, Liu Z, Lin RK, Li M, Li YP. Activin A induces skeletal muscle catabolism via p38β mitogen-activated protein kinase. J Cachexia Sarcopenia Muscle 2017; 8:202-212. [PMID: 27897407 PMCID: PMC5377410 DOI: 10.1002/jcsm.12145] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 07/08/2016] [Accepted: 07/25/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Activation of type IIB activin receptor (ActRIIB) in skeletal muscle leads to muscle atrophy because of increased muscle protein degradation. However, the intracellular signalling mechanism that mediates ActRIIB-activated muscle catabolism is poorly defined. METHODS We investigated the role of p38β mitogen-activated protein kinases (MAPK) in mediating ActRIIB ligand activin A-activated muscle catabolic pathways in C2C12 myotubes and in mice with perturbation of this kinase pharmacologically and genetically. RESULTS Treatment of C2C12 myotubes with activin A or myostatin rapidly activated p38 MAPK and its effector C/EBPβ within 1 h. Paradoxically, Akt was activated at the same time through a p38 MAPK-independent mechanism. These events were followed by up-regulation of ubiquitin ligases atrogin1 (MAFbx) and UBR2 (E3α-II), as well as increase in LC3-II, a marker of autophagosome formation, leading to myofibrillar protein loss and myotube atrophy. The catabolic effects of activin A were abolished by p38α/β MAPK inhibitor SB202190. Using small interfering RNA-mediated gene knockdown, we found that the catabolic activity of activin A was dependent on p38β MAPK specifically. Importantly, systemic administration of activin A to mice similarly activated the catabolic pathways in vivo, and this effect was blocked by SB202190. Further, activin A failed to activate the catabolic pathways in mice with muscle-specific knockout of p38β MAPK. Interestingly, activin A up-regulated MuRF1 in a p38 MAPK-independent manner, and MuRF1 did not appear responsible for activin A-induced myosin heavy chain loss and muscle atrophy. CONCLUSIONS ActRIIB-mediated activation of muscle catabolism is dependent on p38β MAPK-activated signalling.
Collapse
Affiliation(s)
- Hui Ding
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, 77030, USA.,Department of Respiratory Medicine, Yixing Hospital affiliated to Jiangsu University, Yixing, China
| | - Guohua Zhang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Ka Wai Thomas Sin
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Zhelong Liu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, 77030, USA.,Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ren-Kuo Lin
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Min Li
- Department of Medicine and Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yi-Ping Li
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, 77030, USA
| |
Collapse
|
140
|
Lipina C, Hundal HS. Lipid modulation of skeletal muscle mass and function. J Cachexia Sarcopenia Muscle 2017; 8:190-201. [PMID: 27897400 PMCID: PMC5377414 DOI: 10.1002/jcsm.12144] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 07/15/2016] [Accepted: 07/25/2016] [Indexed: 12/22/2022] Open
Abstract
Loss of skeletal muscle mass is a characteristic feature of various pathologies including cancer, diabetes, and obesity, as well as being a general feature of ageing. However, the processes underlying its pathogenesis are not fully understood and may involve multiple factors. Importantly, there is growing evidence which supports a role for fatty acids and their derived lipid intermediates in the regulation of skeletal muscle mass and function. In this review, we discuss evidence pertaining to those pathways which are involved in the reduction, increase and/or preservation of skeletal muscle mass by such lipids under various pathological conditions, and highlight studies investigating how these processes may be influenced by dietary supplementation as well as genetic and/or pharmacological intervention.
Collapse
Affiliation(s)
- Christopher Lipina
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Harinder S Hundal
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| |
Collapse
|
141
|
Sakuma K, Aoi W, Yamaguchi A. Molecular mechanism of sarcopenia and cachexia: recent research advances. Pflugers Arch 2017; 469:573-591. [PMID: 28101649 DOI: 10.1007/s00424-016-1933-3] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 12/28/2016] [Accepted: 12/29/2016] [Indexed: 12/17/2022]
Abstract
Skeletal muscle provides a fundamental basis for human function, enabling locomotion and respiration. Muscle loss occurs as a consequence of several chronic diseases (cachexia) and normal aging (sarcopenia). Although many negative regulators (atrogin-1, muscle ring finger-1, nuclear factor-kappaB (NF-κB), myostatin, etc.) have been proposed to enhance protein degradation during both sarcopenia and cachexia, the adaptation of these mediators markedly differs within both conditions. Sarcopenia and cachectic muscles have been demonstrated to be abundant in myostatin-linked molecules. The ubiquitin-proteasome system (UPS) is activated during rapid atrophy model (cancer cachexia), but few mediators of the UPS change during sarcopenia. NF-κB signaling is activated in cachectic, but not in sarcopenic, muscle. Recent studies have indicated the age-related defect of autophagy signaling in skeletal muscle, whereas autophagic activation occurs in cachectic muscle. This review provides recent research advances dealing with molecular mediators modulating muscle mass in both sarcopenia and cachexia.
Collapse
Affiliation(s)
- Kunihiro Sakuma
- Institute for Liberal Arts, Environment and Society, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro-ku, Tokyo, 152-8550, Japan.
| | - Wataru Aoi
- Laboratory of Health Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-cho Shimogamo, Sakyo-ku, Kyoto, 606-8522, Japan
| | - Akihiko Yamaguchi
- Department of Physical Therapy, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| |
Collapse
|
142
|
Altered mitochondrial quality control signaling in muscle of old gastric cancer patients with cachexia. Exp Gerontol 2017; 87:92-99. [DOI: 10.1016/j.exger.2016.10.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/19/2016] [Accepted: 10/12/2016] [Indexed: 01/09/2023]
|
143
|
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: 31] [Impact Index Per Article: 3.9] [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
|
144
|
Musolino V, Palus S, Tschirner A, Drescher C, Gliozzi M, Carresi C, Vitale C, Muscoli C, Doehner W, von Haehling S, Anker SD, Mollace V, Springer J. Megestrol acetate improves cardiac function in a model of cancer cachexia-induced cardiomyopathy by autophagic modulation. J Cachexia Sarcopenia Muscle 2016; 7:555-566. [PMID: 27239419 PMCID: PMC4864048 DOI: 10.1002/jcsm.12116] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 02/24/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Cachexia is a complex metabolic syndrome associated with cancer. One of the features of cachexia is the loss of muscle mass, characterized by an imbalance between protein synthesis and protein degradation. Muscle atrophy is caused by the hyperactivation of some of the main cellular catabolic pathways, including autophagy. Cachexia also affects the cardiac muscle. As a consequence of the atrophy of the heart, cardiac function is impaired and mortality is increased. Anti-cachectic therapy in patients with cancer cachexia is so far limited to nutritional support and anabolic steroids. The use of the appetite stimulant megestrol acetate (MA) has been discussed as a treatment for cachexia. METHODS In this study the effects of MA were tested in cachectic tumour-bearing rats (Yoshida AH-130 ascites hepatoma). Rats were treated daily with 100 mg/kg of MA or placebo starting one day after tumour inoculation, and for a period of 16 days. Body weight and body composition were assessed at baseline and at the end of the study. Cardiac function was analysed by echocardiography at baseline and at day 11. Locomotor activity and food intake were assessed before tumour inoculation and at day 11. Autophagic markers were assessed in gastrocnemius muscle and heart by western blot analysis. RESULTS Treatment with 100 mg/kg/day MA significantly attenuated the loss of body weight (-9 ± 12%, P < 0.05) and the wasting of lean and fat mass (-7.0 ± 6% and -22.4 ± 3 %, P < 0.001 and P < 0.05, respectively). Administration of 100 mg/kg/day MA significantly protected the heart from general atrophy (633.8 ± 30 mg vs. placebo 474 ± 13 mg, P < 0.001). Tumour-bearing rats displayed cardiac dysfunction, as indicated by the significant impairment of the left ventricular ejection fraction, the left ventricular fractional shortening, the stroke volume, the end dyastolic volume, and the end systolic volume. In contrast, MA significantly improved left ventricular ejection fraction, left ventricular fractional shortening, and left ventricular end systolic volume. Western blotting analysis showed an upregulation of the autophagic pathway in the gastrocnemius and hearts of the placebo-treated tumour-bearing rats. Treatment with MA, however, was able to modulate the autophagic markers (e.g. Beclin-1, p62, TRAF6, and LC3) in the gastrocnemius and in the hearts of tumour-bearing rats. Most importantly, 100 mg/kg/day MA reduced mortality [hazard ratio (HR): 0.44; 95%CI: 0.20-1.00; P = 0.0486]. CONCLUSIONS Megestrol acetate improved survival and reduced wasting through a marked downregulation of autophagy, occurring in both skeletal and heart muscle, the latter effect leading to a significant improvement of cardiac function. Our data suggest that MA might represent a valuable strategy to counteract the development of cancer cachexia-induced cardiomyopathy.
Collapse
Affiliation(s)
- Vincenzo Musolino
- Institute of Research for Food Safety and Health (IRC-FSH) University of Catanzaro 'Magna Graecia' Catanzaro Italy
| | - Sandra Palus
- Division of Innovative Clinical Trials, Department of Cardiology University Medical Center Göttingen (UMG) Göttingen Germany
| | - Anika Tschirner
- Applied Cachexia Research, Department of Cardiology Charité Medical School Berlin Germany
| | - Cathleen Drescher
- Division of Innovative Clinical Trials, Department of Cardiology University Medical Center Göttingen (UMG) Göttingen Germany
| | - Micaela Gliozzi
- Institute of Research for Food Safety and Health (IRC-FSH) University of Catanzaro 'Magna Graecia' Catanzaro Italy
| | - Cristina Carresi
- Institute of Research for Food Safety and Health (IRC-FSH) University of Catanzaro 'Magna Graecia' Catanzaro Italy
| | - Cristiana Vitale
- Centre for Clinical and Basic Research IRCCS San Raffaele Pisana Rome Italy
| | - Carolina Muscoli
- Institute of Research for Food Safety and Health (IRC-FSH) University of Catanzaro 'Magna Graecia' Catanzaro Italy; Centre for Clinical and Basic Research IRCCS San Raffaele Pisana Rome Italy
| | - Wolfram Doehner
- Center for Stroke Research Charité Medical School, Campus Virchow- Klinikum Berlin Germany
| | - Stephan von Haehling
- Division of Innovative Clinical Trials, Department of Cardiology University Medical Center Göttingen (UMG) Göttingen Germany
| | - Stefan D Anker
- Division of Innovative Clinical Trials, Department of Cardiology University Medical Center Göttingen (UMG) Göttingen Germany
| | - Vincenzo Mollace
- Institute of Research for Food Safety and Health (IRC-FSH) University of Catanzaro 'Magna Graecia' Catanzaro Italy; Centre for Clinical and Basic Research IRCCS San Raffaele Pisana Rome Italy
| | - Jochen Springer
- Division of Innovative Clinical Trials, Department of Cardiology University Medical Center Göttingen (UMG) Göttingen Germany
| |
Collapse
|
145
|
Loss of strength capacity is associated with mortality, but resistance exercise training promotes only modest effects during cachexia progression. Life Sci 2016; 163:11-22. [DOI: 10.1016/j.lfs.2016.08.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 08/14/2016] [Accepted: 08/25/2016] [Indexed: 12/31/2022]
|
146
|
Wang C, Guo D, Wang Q, You S, Qiao Z, Liu Y, Dai H, Tang H. Aliskiren targets multiple systems to alleviate cancer cachexia. Oncol Rep 2016; 36:3014-3022. [PMID: 27667116 DOI: 10.3892/or.2016.5118] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/26/2016] [Indexed: 11/05/2022] Open
Abstract
To examine the effects of aliskiren, a small-molecule renin inhibitor, on cancer cachexia and to explore the underlying mechanisms. A cancer cachexia model was established by subcutaneously injecting C26 mouse colon carcinoma cells into isogenic BALB/c mice. Aliskiren was administered intragastrically [10 mg/kg body weight (BW)] on day 5 (as a preventive strategy, AP group) or on day 12 (as a therapeutic strategy, AT group) after C26 injection. Mice that received no C26 injection (healthy controls, HC group) or only C26 injection but not aliskiren (cancer, CA group) were used as controls. BW, tumor growth, whole body functions, and survival were monitored daily in half of the mice in each group, whereas serum, tumors, and gastrocnemius muscles were harvested from the other mice after sacrifice on day 20 for further analysis. Aliskiren significantly alleviated multiple cachexia‑associated symptoms, including BW loss, tumor burden, muscle wasting, muscular dysfunction, and shortened survival. On the molecular level, aliskiren antagonized cachexia‑induced activation of the renin‑angiotensin system (RAS), systematic and muscular inflammation, oxidative stress, and autophagy‑lysosome as well as ubiquitin‑proteasome stimulation. In addition, early administration of aliskiren before cachexia development (AP group) resulted in more robust effects in alleviating cachexia or targeting underlying mechanisms than administration after cachexia development (AT group). Aliskiren exhibited potent anti‑cachexia activities. These activities were achieved through the targeting of at least four mechanisms underlying cachexia development: RAS activation, increase in systematic inflammation, upregulation of oxidative stress, and stimulation of autophagy-lysosome pathway (ALP) and ubiquitin-proteasome pathway (UPP).
Collapse
Affiliation(s)
- Chaoyi Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Dunwei Guo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qiang Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Song You
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zhongpeng Qiao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yong Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hang Dai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hua Tang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
147
|
Bohnert KR, Gallot YS, Sato S, Xiong G, Hindi SM, Kumar A. Inhibition of ER stress and unfolding protein response pathways causes skeletal muscle wasting during cancer cachexia. FASEB J 2016; 30:3053-68. [PMID: 27206451 PMCID: PMC5001510 DOI: 10.1096/fj.201600250rr] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/12/2016] [Indexed: 12/25/2022]
Abstract
Cachexia is a devastating syndrome that causes morbidity and mortality in a large number of patients with cancer. However, the mechanisms of cancer cachexia remain poorly understood. Accumulation of misfolded proteins in the endoplasmic reticulum (ER) causes stress. The ER responds to this stress through activating certain pathways commonly known as the unfolding protein response (UPR). The main function of UPR is to restore homeostasis, but excessive or prolonged activation of UPR can lead to pathologic conditions. In this study, we examined the role of ER stress and UPR in regulation of skeletal muscle mass in naïve conditions and during cancer cachexia. Our results demonstrate that multiple markers of ER stress are highly activated in skeletal muscle of Lewis lung carcinoma (LLC) and Apc(Min/+) mouse models of cancer cachexia. Treatment of mice with 4-phenylbutyrate (4-PBA), a chemical chaperon and a potent inhibitor of ER stress, significantly reduced skeletal muscle strength and mass in both control and LLC-bearing mice. Blocking the UPR also increased the proportion of fast-type fibers in soleus muscle of both control and LLC-bearing mice. Inhibition of UPR reduced the activity of Akt/mTOR pathway and increased the expression of the components of the ubiquitin-proteasome system and autophagy in LLC-bearing mice. Moreover, we found that the inhibition of UPR causes severe atrophy in cultured myotubes. Our study provides initial evidence that ER stress and UPR pathways are essential for maintaining skeletal muscle mass and strength and for protection against cancer cachexia.-Bohnert, K. R., Gallot, Y. S., Sato, S., Xiong, G., Hindi, S. M., Kumar, A. Inhibition of ER stress and unfolding protein response pathways causes skeletal muscle wasting during cancer cachexia.
Collapse
Affiliation(s)
- Kyle R Bohnert
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Yann S Gallot
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Shuichi Sato
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Guangyan Xiong
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Sajedah M Hindi
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Ashok Kumar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
148
|
Costamagna D, Mommaerts H, Sampaolesi M, Tylzanowski P. Noggin inactivation affects the number and differentiation potential of muscle progenitor cells in vivo. Sci Rep 2016; 6:31949. [PMID: 27573479 PMCID: PMC5004166 DOI: 10.1038/srep31949] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 07/28/2016] [Indexed: 10/25/2022] Open
Abstract
Inactivation of Noggin, a secreted antagonist of Bone Morphogenetic Proteins (BMPs), in mice leads, among others, to severe malformations of the appendicular skeleton and defective skeletal muscle fibers. To determine the molecular basis of the phenotype, we carried out a histomorphological and molecular analysis of developing muscles Noggin(-/-) mice. We show that in 18.5 dpc embryos there is a marked reduction in muscle fiber size and a failure of nuclei migration towards the cell membrane. Molecularly, the absence of Noggin results in an increased BMP signaling in muscle tissue as shown by the increase in SMAD1/5/8 phosphorylation, concomitant with the induction of BMP target genes such as Id1, 2, 3 as well as Msx1. Finally, upon removal of Noggin, the number of mesenchymal Pax7(+) muscle precursor cells is reduced and they are more prone to differentiate into adipocytes in vitro. Thus, our results highlight the importance of Noggin/BMP balance for myogenic commitment of early fetal progenitor cells.
Collapse
Affiliation(s)
- Domiziana Costamagna
- Translational Cardiomyology Lab, Stem Cell Biology and Embryology, Dept. Development and Regeneration, KU Leuven, Belgium.,Laboratory of Experimental Medicine and Clinical Pathology, Dept. Clinical and Biological Sciences, University of Turin, Italy
| | - Hendrik Mommaerts
- Department of Development and Regeneration, Laboratory for Developmental and Stem Cell Biology, Skeletal Biology and Engineering Research Centre, KU Leuven, Belgium
| | - Maurilio Sampaolesi
- Translational Cardiomyology Lab, Stem Cell Biology and Embryology, Dept. Development and Regeneration, KU Leuven, Belgium.,Division of Human Anatomy, Dept. of Public Health, Experimental and Forensic Medicine, University of Pavia, Italy
| | - Przemko Tylzanowski
- Department of Development and Regeneration, Laboratory for Developmental and Stem Cell Biology, Skeletal Biology and Engineering Research Centre, KU Leuven, Belgium.,Department of Biochemistry and Molecular Biology, Medical University, Lublin, Poland
| |
Collapse
|
149
|
Aversa Z, Pin F, Lucia S, Penna F, Verzaro R, Fazi M, Colasante G, Tirone A, Fanelli FR, Ramaccini C, Costelli P, Muscaritoli M. Autophagy is induced in the skeletal muscle of cachectic cancer patients. Sci Rep 2016; 6:30340. [PMID: 27459917 PMCID: PMC4962093 DOI: 10.1038/srep30340] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 06/30/2016] [Indexed: 12/13/2022] Open
Abstract
Basal rates of autophagy can be markedly accelerated by environmental stresses. Recently, autophagy has been involved in cancer-induced muscle wasting. Aim of this study has been to evaluate if autophagy is induced in the skeletal muscle of cancer patients. The expression (mRNA and protein) of autophagic markers has been evaluated in intraoperative muscle biopsies. Beclin-1 protein levels were increased in cachectic cancer patients, suggesting autophagy induction. LC3B-I protein levels were not significantly modified. LC3B-II protein levels were significantly increased in cachectic cancer patients suggesting either increased autophagosome formation or reduced autophagosome turnover. Conversely, p62 protein levels were increased in cachectic and non-cachectic cancer patients, suggesting impaired autophagosome clearance. As for mitophagy, both Bnip3 and Nix/Bnip3L show a trend to increase in cachectic patients. In the same patients, Parkin levels significantly increased, while PINK1 was unchanged. At gene level, Beclin-1, p-62, BNIP3, NIX/BNIP3L and TFEB mRNAs were not significantly modulated, while LC3B and PINK1 mRNA levels were increased and decreased, respectively, in cachectic cancer patients. Autophagy is induced in the skeletal muscle of cachectic cancer patients, although autophagosome clearance appears to be impaired. Further studies should evaluate whether modulation of autophagy could represent a relevant therapeutic strategy in cancer cachexia.
Collapse
Affiliation(s)
- Zaira Aversa
- Department of Clinical Medicine, Sapienza University of Rome, Rome, Italy
| | - Fabrizio Pin
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
- Interuniversity Institute of Myology, Italy
| | - Simone Lucia
- Department of Clinical Medicine, Sapienza University of Rome, Rome, Italy
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
- Interuniversity Institute of Myology, Italy
| | | | - Maurizio Fazi
- Department of Surgery, M.G. Vannini Hospital, Rome, Italy
| | - Giuseppina Colasante
- UOSA Chirurgia Bariatrica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Andrea Tirone
- UOSA Chirurgia Bariatrica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | | - Cesarina Ramaccini
- Department of Clinical Medicine, Sapienza University of Rome, Rome, Italy
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
- Interuniversity Institute of Myology, Italy
| | | |
Collapse
|
150
|
Dietary supplementation with shiikuwasha extract attenuates dexamethasone-induced skeletal muscle atrophy in aged rats. SPRINGERPLUS 2016; 5:816. [PMID: 27390656 PMCID: PMC4916103 DOI: 10.1186/s40064-016-2427-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 05/26/2016] [Indexed: 12/13/2022]
Abstract
Background Skeletal muscle atrophy is caused by a variety of diseases and conditions. In particular, skeletal muscle atrophy in the elderly contributes to a loss of independence with advanced age and increases the risk of falling. However, the effect of food consumed on a daily basis on skeletal muscle atrophy has been the focus of little research. In this study, the effects of dietary supplementation with shiikuwasha extract or grape extract on dexamethasone-induced skeletal muscle atrophy were evaluated in aged rats. Methods Aged male rats (15-month-old) were fed a diet supplemented with either 1 % shiikuwasha extract or 1 % grape extract for 19 days. During the last 5 days of the feeding period, rats were injected with dexamethasone to induce muscle atrophy. Results Body weight and hind-limb muscle weight were significantly decreased by dexamethasone treatment. The supplementation of shiikuwasha extract showed no effect on body weight loss, but markedly attenuated tibialis anterior muscle weight loss induced by dexamethasone. On the other hand, grape extract did not affect muscle weight loss. Furthermore, shiikuwasha extract significantly reduced dexamethasone-induced expression of atrogin-1 and MuRF1 mRNA, but did not reduce LC3B-II protein levels. Conclusion These results suggest that shiikuwasha extract may partially inhibit the activation of the ubiquitin–proteasome system and may consequently attenuate skeletal muscle atrophy induced by dexamethasone in aged rats.
Collapse
|