1
|
Ouellette JM, Mallender MD, Hian-Cheong DJ, Scurto DL, Nicholas JE, Trumble SJ, Hawke TJ, Krause MP. Altered sphingolipid profile in response to skeletal muscle injury in a mouse model of type 1 diabetes mellitus. Am J Physiol Cell Physiol 2025; 328:C273-C287. [PMID: 39611411 DOI: 10.1152/ajpcell.00158.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Abstract
A complication of type 1 diabetes mellitus (T1DM) is diabetic myopathy that includes reduced regenerative capacity of skeletal muscle. Sphingolipids are a diverse family of lipids with roles in skeletal muscle regeneration. Some studies have found changes in sphingolipid species levels in T1DM, however, the effect of T1DM on a sphingolipid panel in regenerating skeletal muscle has not been examined. Wild-type (WT) and diabetic Ins2Akita+/- (Akita) mice received cardiotoxin-induced muscle injury in their left quadriceps, gastrocnemius-plantaris-soleus, and tibialis anterior muscles with the contralateral muscles serving as uninjured controls. Muscles were collected at 1, 3, 5, or 7 days postinjury. In regenerating muscle from Akita mice, lipid staining with BODIPY 493/503 revealed increased intramyocellular and total lipids and perilipin-1-positive cell numbers as compared with WT. Liquid chromatography-mass spectrometry of quadriceps was used to identify sphingolipid levels in skeletal muscle. The C22:0 and C24:0 ceramides were significantly elevated in uninjured Akita, whereas ceramide C24:1 was decreased in injured Akita compared with WT. Ceramide-1-phosphate was increased in Akita compared with WT regardless of injury, whereas sphingosine-1-phosphate (S1P) was elevated with injury in WT but this response was muted in Akita mice. Western blotting of key enzymes involved in sphingolipid metabolism revealed S1P lyase, the enzyme that degrades S1P irreversibly, was significantly elevated in the injured muscle in Akita mice during regeneration, in accordance with lower S1P levels. This mouse model of T1DM demonstrates sphingolipidomic changes that may contribute to delayed muscle regeneration.NEW & NOTEWORTHY Muscle lipids become elevated, and the sphingolipid profile is altered by T1DM in skeletal muscle regeneration. A loss of S1P is accompanied by greater expression of sphingosine-1-phosphate lyase (SPL) in response to injury in Akita mice, suggesting a role for sphingolipids in the attenuated repair of skeletal muscle in T1DM rodent models. Although ceramide-1-phosphate (C1P) is increased with T1DM, there was no increase in ceramide kinase (CerK) suggesting an alternative route of ceramide phosphorylation in skeletal muscle.
Collapse
Affiliation(s)
- Jacob M Ouellette
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, Windsor, Ontario, Canada
| | - Michael D Mallender
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, Windsor, Ontario, Canada
| | - Dylan J Hian-Cheong
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, Windsor, Ontario, Canada
| | - Daniel L Scurto
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, Windsor, Ontario, Canada
| | - James E Nicholas
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, Windsor, Ontario, Canada
| | | | - Thomas J Hawke
- Department of Pathology & Molecular Medicine, Faculty of Health Science, McMaster University, Hamilton, Ontario, Canada
| | - Matthew P Krause
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, Windsor, Ontario, Canada
| |
Collapse
|
2
|
Seo JH, Koh J, Cho HJ, Kim H, Lee Y, Kim SJ, Yoon PW, Kim W, Bae SJ, Kim H, Yoo HJ, Lee SH. Sphingolipid metabolites as potential circulating biomarkers for sarcopenia in men. J Cachexia Sarcopenia Muscle 2024; 15:2476-2486. [PMID: 39229927 PMCID: PMC11634516 DOI: 10.1002/jcsm.13582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/27/2024] [Accepted: 07/23/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Sarcopenia is an age-related progressive loss of muscle mass and function. Sarcopenia is a multifactorial disorder, including metabolic disturbance; therefore, metabolites may be used as circulating biomarkers for sarcopenia. We aimed to investigate potential biomarkers of sarcopenia using metabolomics. METHODS After non-targeted metabolome profiling of plasma from mice of an aging mouse model of sarcopenia, sphingolipid metabolites and muscle cells from the animal model were evaluated using targeted metabolome profiling. The associations between sphingolipid metabolites identified from mouse and cell studies and sarcopenia status were assessed in men in an age-matched discovery (72 cases and 72 controls) and validation (36 cases and 128 controls) cohort; women with sarcopenia (36 cases and 36 controls) were also included as a discovery cohort. RESULTS Both non-targeted and targeted metabolome profiling in the experimental studies showed an association between sphingolipid metabolites, including ceramides (CERs) and sphingomyelins (SMs), and sarcopenia. Plasma SM (16:0), CER (24:1), and SM (24:1) levels in men with sarcopenia were significantly higher in the discovery cohort than in the controls (all P < 0.05). There were no significant differences in plasma sphingolipid levels for women with or without sarcopenia. In men in the discovery cohort, an area under the receiver-operating characteristic curve (AUROC) of SM (16:0) for low muscle strength and low muscle mass was 0.600 (95% confidence interval [CI]: 0.501-0.699) and 0.647 (95% CI: 0.557-0.737). The AUROC (95% CI) of CER (24:1) and SM (24:1) for low muscle mass in men was 0.669 (95% CI: 0.581-0.757) and 0.670 (95% CI: 0.582-0.759), respectively. Using a regression equation combining CER (24:1) and SM (16:0) levels, a sphingolipid (SphL) score was calculated; an AUROC of the SphL score for sarcopenia was 0.712 (95% CI: 0.626-0.798). The addition of the SphL score to HGS significantly improved the AUC from 0.646 (95% CI: 0.575-0.717; HGS only) to 0.751 (95% CI: 0.671-0.831, P = 0.002; HGS + SphL) in the discovery cohort. The predictive ability of the SphL score for sarcopenia was confirmed in the validation cohort (AUROC = 0.695, 95% CI: 0.591-0.799). CONCLUSIONS SM (16:0), reflecting low muscle strength, and CER (24:1) and SM (16:0), reflecting low muscle mass, are potential circulating biomarkers for sarcopenia in men. Further research on sphingolipid metabolites is required to confirm these results and provide additional insights into the metabolomic changes relevant to the pathogenesis and diagnosis of sarcopenia.
Collapse
Affiliation(s)
- Je Hyun Seo
- Veterans Health Service Medical CenterVeterans Medical Research InstituteSeoulSouth Korea
| | - Jung‐Min Koh
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Han Jin Cho
- Biomedical Research CenterAsan Institute for Life Sciences, Asan Medical CenterSeoulSouth Korea
| | - Hanjun Kim
- Biomedical Research CenterAsan Institute for Life Sciences, Asan Medical CenterSeoulSouth Korea
| | - Young‐Sun Lee
- Biomedical Research CenterAsan Institute for Life Sciences, Asan Medical CenterSeoulSouth Korea
| | - Su Jung Kim
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Pil Whan Yoon
- Department of Orthopedic SurgerySeoul Now HospitalAnyangSouth Korea
| | - Won Kim
- Department of Rehabilitation Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Sung Jin Bae
- Health Screening and Promotion Center, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Hong‐Kyu Kim
- Health Screening and Promotion Center, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Hyun Ju Yoo
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Seung Hun Lee
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| |
Collapse
|
3
|
Dlamini M, Khathi A. Prediabetes-Associated Changes in Skeletal Muscle Function and Their Possible Links with Diabetes: A Literature Review. Int J Mol Sci 2023; 25:469. [PMID: 38203642 PMCID: PMC10778616 DOI: 10.3390/ijms25010469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
The skeletal muscle plays a critical role in regulating systemic blood glucose homeostasis. Impaired skeletal muscle glucose homeostasis associated with type 2 diabetes mellitus (T2DM) has been observed to significantly affect the whole-body glucose homeostasis, thereby resulting in other diabetic complications. T2DM does not only affect skeletal muscle glucose homeostasis, but it also affects skeletal muscle structure and functional capacity. Given that T2DM is a global health burden, there is an urgent need to develop therapeutic medical therapies that will aid in the management of T2DM. Prediabetes (PreDM) is a prominent risk factor of T2DM that usually goes unnoticed in many individuals as it is an asymptomatic condition. Hence, research on PreDM is essential because establishing diabetic biomarkers during the prediabetic state would aid in preventing the development of T2DM, as PreDM is a reversible condition if it is detected in the early stages. The literature predominantly documents the changes in skeletal muscle during T2DM, but the changes in skeletal muscle during prediabetes are not well elucidated. In this review, we seek to review the existing literature on PreDM- and T2DM-associated changes in skeletal muscle function.
Collapse
Affiliation(s)
| | - Andile Khathi
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban X54001, South Africa;
| |
Collapse
|
4
|
Ali-Berrada S, Guitton J, Tan-Chen S, Gyulkhandanyan A, Hajduch E, Le Stunff H. Circulating Sphingolipids and Glucose Homeostasis: An Update. Int J Mol Sci 2023; 24:12720. [PMID: 37628901 PMCID: PMC10454113 DOI: 10.3390/ijms241612720] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Sphingolipids are a family of lipid molecules produced through different pathways in mammals. Sphingolipids are structural components of membranes, but in response to obesity, they are implicated in the regulation of various cellular processes, including inflammation, apoptosis, cell proliferation, autophagy, and insulin resistance which favors dysregulation of glucose metabolism. Of all sphingolipids, two species, ceramides and sphingosine-1-phosphate (S1P), are also found abundantly secreted into the bloodstream and associated with lipoproteins or extracellular vesicles. Plasma concentrations of these sphingolipids can be altered upon metabolic disorders and could serve as predictive biomarkers of these diseases. Recent important advances suggest that circulating sphingolipids not only serve as biomarkers but could also serve as mediators in the dysregulation of glucose homeostasis. In this review, advances of molecular mechanisms involved in the regulation of ceramides and S1P association to lipoproteins or extracellular vesicles and how they could alter glucose metabolism are discussed.
Collapse
Affiliation(s)
- Sarah Ali-Berrada
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, F-75006 Paris, France; (S.A.-B.); (S.T.-C.); (A.G.)
- Institut Hospitalo-Universitaire ICAN, 75013 Paris, France
| | - Jeanne Guitton
- Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, CNRS UMR 9197, 91400 Saclay, France;
| | - Sophie Tan-Chen
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, F-75006 Paris, France; (S.A.-B.); (S.T.-C.); (A.G.)
- Institut Hospitalo-Universitaire ICAN, 75013 Paris, France
| | - Anna Gyulkhandanyan
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, F-75006 Paris, France; (S.A.-B.); (S.T.-C.); (A.G.)
- Institut Hospitalo-Universitaire ICAN, 75013 Paris, France
| | - Eric Hajduch
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, F-75006 Paris, France; (S.A.-B.); (S.T.-C.); (A.G.)
- Institut Hospitalo-Universitaire ICAN, 75013 Paris, France
| | - Hervé Le Stunff
- Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, CNRS UMR 9197, 91400 Saclay, France;
| |
Collapse
|
5
|
Ichii S, Matsuoka I, Okazaki F, Shimada Y. Zebrafish Models for Skeletal Muscle Senescence: Lessons from Cell Cultures and Rodent Models. Molecules 2022; 27:molecules27238625. [PMID: 36500717 PMCID: PMC9739860 DOI: 10.3390/molecules27238625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/08/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Human life expectancy has markedly increased over the past hundred years. Consequently, the percentage of elderly people is increasing. Aging and sarcopenic changes in skeletal muscles not only reduce locomotor activities in elderly people but also increase the chance of trauma, such as bone fractures, and the incidence of other diseases, such as metabolic syndrome, due to reduced physical activity. Exercise therapy is currently the only treatment and prevention approach for skeletal muscle aging. In this review, we aimed to summarize the strategies for modeling skeletal muscle senescence in cell cultures and rodents and provide future perspectives based on zebrafish models. In cell cultures, in addition to myoblast proliferation and myotube differentiation, senescence induction into differentiated myotubes is also promising. In rodents, several models have been reported that reflect the skeletal muscle aging phenotype or parts of it, including the accelerated aging models. Although there are fewer models of skeletal muscle aging in zebrafish than in mice, various models have been reported in recent years with the development of CRISPR/Cas9 technology, and further advancements in the field using zebrafish models are expected in the future.
Collapse
Affiliation(s)
- Shogo Ichii
- Graduate School of Bioresources, Mie University, Tsu, Mie 514-8507, Japan
| | - Izumi Matsuoka
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Mie 514-8507, Japan
| | - Fumiyoshi Okazaki
- Graduate School of Bioresources, Mie University, Tsu, Mie 514-8507, Japan
- Zebrafish Drug Screening Center, Mie University, Tsu, Mie 514-8507, Japan
| | - Yasuhito Shimada
- Zebrafish Drug Screening Center, Mie University, Tsu, Mie 514-8507, Japan
- Department of Bioinformatics, Mie University Advanced Science Research Promotion Center, Tsu, Mie 514-8507, Japan
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
- Correspondence: ; Tel.: +81-592-31-5411
| |
Collapse
|
6
|
Laurila PP, Wohlwend M, Imamura de Lima T, Luan P, Herzig S, Zanou N, Crisol B, Bou-Sleiman M, Porcu E, Gallart-Ayala H, Handzlik MK, Wang Q, Jain S, D'Amico D, Salonen M, Metallo CM, Kutalik Z, Eichmann TO, Place N, Ivanisevic J, Lahti J, Eriksson JG, Auwerx J. Sphingolipids accumulate in aged muscle, and their reduction counteracts sarcopenia. NATURE AGING 2022; 2:1159-1175. [PMID: 37118545 DOI: 10.1038/s43587-022-00309-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/07/2022] [Indexed: 04/30/2023]
Abstract
Age-related muscle dysfunction and sarcopenia are major causes of physical incapacitation in older adults and currently lack viable treatment strategies. Here we find that sphingolipids accumulate in mouse skeletal muscle upon aging and that both genetic and pharmacological inhibition of sphingolipid synthesis prevent age-related decline in muscle mass while enhancing strength and exercise capacity. Inhibition of sphingolipid synthesis confers increased myogenic potential and promotes protein synthesis. Within the sphingolipid pathway, we show that accumulation of dihydroceramides is the culprit disturbing myofibrillar homeostasis. The relevance of sphingolipid pathways in human aging is demonstrated in two cohorts, the UK Biobank and Helsinki Birth Cohort Study in which gene expression-reducing variants of SPTLC1 and DEGS1 are associated with improved and reduced fitness of older individuals, respectively. These findings identify sphingolipid synthesis inhibition as an attractive therapeutic strategy for age-related sarcopenia and co-occurring pathologies.
Collapse
Affiliation(s)
- Pirkka-Pekka Laurila
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Martin Wohlwend
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tanes Imamura de Lima
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Peiling Luan
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sébastien Herzig
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences and Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Barbara Crisol
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Maroun Bou-Sleiman
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Eleonora Porcu
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Michal K Handzlik
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Qi Wang
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Suresh Jain
- Intonation Research Laboratories, Secunderabad, India
| | - Davide D'Amico
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Minna Salonen
- Chronic Disease Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Christian M Metallo
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zoltan Kutalik
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- University Center for Primary Care and Public Health, University of Lausanne, Lausanne, Switzerland
| | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Center for Explorative Lipidomics, BioTechMed-Graz, Graz, Austria
| | - Nicolas Place
- Institute of Sport Sciences and Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Jari Lahti
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
- Turku Institute for Advanced Studies, University of Turku, Turku, Finland
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, National University Singapore, Yong Loo Lin School of Medicine, Singapore, Singapore
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
7
|
Olsson K, Cheng AJ, Al-Ameri M, Tardif N, Melin M, Rooyackers O, Lanner JT, Westerblad H, Gustafsson T, Bruton JD, Rullman E. Sphingomyelinase activity promotes atrophy and attenuates force in human muscle fibres and is elevated in heart failure patients. J Cachexia Sarcopenia Muscle 2022; 13:2551-2561. [PMID: 35852046 PMCID: PMC9530516 DOI: 10.1002/jcsm.13029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/26/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Activation of sphingomyelinase (SMase) as a result of a general inflammatory response has been implicated as a mechanism underlying disease-related loss of skeletal muscle mass and function in several clinical conditions including heart failure. Here, for the first time, we characterize the effects of SMase activity on human muscle fibre contractile function and assess skeletal muscle SMase activity in heart failure patients. METHODS The effects of SMase on force production and intracellular Ca2+ handling were investigated in single intact human muscle fibres. Additional mechanistic studies were performed in single mouse toe muscle fibres. RNA sequencing was performed in human muscle bundles exposed to SMase. Intramuscular SMase activity was measured from heart failure patients (n = 61, age 69 ± 0.8 years, NYHA III-IV, ejection fraction 25 ± 1.0%, peak VO2 14.4 ± 0.6 mL × kg × min) and healthy age-matched control subjects (n = 10, age 71 ± 2.2 years, ejection fraction 60 ± 1.2%, peak VO2 25.8 ± 1.1 mL × kg × min). SMase activity was related to circulatory factors known to be associated with progression and disease severity in heart failure. RESULTS Sphingomyelinase reduced muscle fibre force production (-30%, P < 0.05) by impairing sarcoplasmic reticulum (SR) Ca2+ release (P < 0.05) and reducing myofibrillar Ca2+ sensitivity. In human muscle bundles exposed to SMase, RNA sequencing analysis revealed 180 and 291 genes as up-regulated and down-regulated, respectively, at a FDR of 1%. Gene-set enrichment analysis identified 'proteasome degradation' as an up-regulated pathway (average fold-change 1.1, P = 0.008), while the pathway 'cytoplasmic ribosomal proteins' (average fold-change 0.8, P < 0.0001) and factors involving proliferation of muscle cells (average fold-change 0.8, P = 0.0002) where identified as down-regulated. Intramuscular SMase activity was ~20% higher (P < 0.05) in human heart failure patients than in age-matched healthy controls and was positively correlated with markers of disease severity and progression, and with several circulating inflammatory proteins, including TNF-receptor 1 and 2. In a longitudinal cohort of heart failure patients (n = 6, mean follow-up time 2.5 ± 0.2 years), SMase activity was demonstrated to increase by 30% (P < 0.05) with duration of disease. CONCLUSIONS The present findings implicate activation of skeletal muscle SMase as a mechanism underlying human heart failure-related loss of muscle mass and function. Moreover, our findings strengthen the idea that SMase activation may underpin disease-related loss of muscle mass and function in other clinical conditions, acting as a common patophysiological mechanism for the myopathy often reported in diseases associated with a systemic inflammatory response.
Collapse
Affiliation(s)
- Karl Olsson
- Department of Laboratory Medicine, Section of Clinical Physiology, Karolinska Institutet, Huddinge, Sweden
| | - Arthur J Cheng
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Solna, Sweden.,Muscle Health Research Centre, School of Kinesiology and Health Science, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Mamdoh Al-Ameri
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Nicolas Tardif
- Division of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Huddinge, Sweden.,Anesthesiology and intensive care, Department of Clinical Science Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| | - Michael Melin
- Department of Laboratory Medicine, Section of Clinical Physiology, Karolinska Institutet, Huddinge, Sweden
| | - Olav Rooyackers
- Division of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Huddinge, Sweden.,Anesthesiology and intensive care, Department of Clinical Science Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Solna, Sweden
| | - Håkan Westerblad
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Solna, Sweden
| | - Thomas Gustafsson
- Department of Laboratory Medicine, Section of Clinical Physiology, Karolinska Institutet and Department of Clinical Physiology Karolinska Univ Hospital, Huddinge, Sweden
| | - Joseph D Bruton
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Solna, Sweden
| | - Eric Rullman
- Department of Laboratory Medicine, Section of Clinical Physiology, Karolinska Institutet and Department of Clinical Physiology Karolinska Univ Hospital, Huddinge, Sweden
| |
Collapse
|
8
|
LIM CHANGHYUN, NUNES EVERSONA, CURRIER BRADS, MCLEOD JONATHANC, THOMAS AARONCQ, PHILLIPS STUARTM. An Evidence-Based Narrative Review of Mechanisms of Resistance Exercise-Induced Human Skeletal Muscle Hypertrophy. Med Sci Sports Exerc 2022; 54:1546-1559. [PMID: 35389932 PMCID: PMC9390238 DOI: 10.1249/mss.0000000000002929] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skeletal muscle plays a critical role in physical function and metabolic health. Muscle is a highly adaptable tissue that responds to resistance exercise (RE; loading) by hypertrophying, or during muscle disuse, RE mitigates muscle loss. Resistance exercise training (RET)-induced skeletal muscle hypertrophy is a product of external (e.g., RE programming, diet, some supplements) and internal variables (e.g., mechanotransduction, ribosomes, gene expression, satellite cells activity). RE is undeniably the most potent nonpharmacological external variable to stimulate the activation/suppression of internal variables linked to muscular hypertrophy or countering disuse-induced muscle loss. Here, we posit that despite considerable research on the impact of external variables on RET and hypertrophy, internal variables (i.e., inherent skeletal muscle biology) are dominant in regulating the extent of hypertrophy in response to external stimuli. Thus, identifying the key internal skeletal muscle-derived variables that mediate the translation of external RE variables will be pivotal to determining the most effective strategies for skeletal muscle hypertrophy in healthy persons. Such work will aid in enhancing function in clinical populations, slowing functional decline, and promoting physical mobility. We provide up-to-date, evidence-based perspectives of the mechanisms regulating RET-induced skeletal muscle hypertrophy.
Collapse
Affiliation(s)
- CHANGHYUN LIM
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - EVERSON A. NUNES
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
- Department of Physiological Science, Federal University of Santa Catarina, Florianópolis, Santa-Catarina, BRAZIL
| | - BRAD S. CURRIER
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - JONATHAN C. MCLEOD
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - AARON C. Q. THOMAS
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - STUART M. PHILLIPS
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| |
Collapse
|
9
|
Meacci E, Pierucci F, Garcia-Gil M. Skeletal Muscle and COVID-19: The Potential Involvement of Bioactive Sphingolipids. Biomedicines 2022; 10:biomedicines10051068. [PMID: 35625805 PMCID: PMC9138286 DOI: 10.3390/biomedicines10051068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 virus infection is the cause of the coronavirus disease 2019 (COVID-19), which is still spreading over the world. The manifestation of this disease can range from mild to severe and can be limited in time (weeks) or persist for months in about 30–50% of patients. COVID-19 is considered a multiple organ dysfunction syndrome and the musculoskeletal system manifestations are beginning to be considered of absolute importance in both COVID-19 patients and in patients recovering from the SARS-CoV-2 infection. Musculoskeletal manifestations of COVID-19 and other coronavirus infections include loss of muscle mass, muscle weakness, fatigue or myalgia, and muscle injury. The molecular mechanisms by which SARS-CoV-2 can cause damage to skeletal muscle (SkM) cells are not yet well understood. Sphingolipids (SLs) represent an important class of eukaryotic lipids with structural functions as well as bioactive molecules able to modulate crucial processes, including inflammation and viral infection. In the last two decades, several reports have highlighted the role of SLs in modulating SkM cell differentiation, regeneration, aging, response to insulin, and contraction. This review summarizes the consequences of SARS-CoV-2 infection on SkM and the potential involvement of SLs in the tissue responses to virus infection. In particular, we highlight the role of sphingosine 1-phosphate signaling in order to aid the prediction of novel targets for preventing and/or treating acute and long-term musculoskeletal manifestations of virus infection in COVID-19.
Collapse
Affiliation(s)
- Elisabetta Meacci
- Unit of Biochemical Sciences and Molecular Biology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale GB Morgagni 50, 50121 Florence, Italy;
- Interuniversity Institute of Myology, University of Florence, 50121 Florence, Italy
- Correspondence: ; Tel.: +39-055-2751231
| | - Federica Pierucci
- Unit of Biochemical Sciences and Molecular Biology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale GB Morgagni 50, 50121 Florence, Italy;
| | - Mercedes Garcia-Gil
- Unit of Physiology, Department of Biology, University of Pisa, Via S. Zeno 31, 56127 Pisa, Italy;
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56127 Pisa, Italy
| |
Collapse
|
10
|
Lee MH, Lee JH, Kim WJ, Kim SH, Kim SY, Kim HS, Kim TJ. Linoleic Acid Attenuates Denervation-Induced Skeletal Muscle Atrophy in Mice through Regulation of Reactive Oxygen Species-Dependent Signaling. Int J Mol Sci 2022; 23:4778. [PMID: 35563168 PMCID: PMC9105847 DOI: 10.3390/ijms23094778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/17/2022] [Accepted: 04/25/2022] [Indexed: 02/01/2023] Open
Abstract
Muscle atrophy is a major muscle disease, the symptoms of which include decreased muscle volume leading to insufficient muscular support during exercise. One cause of muscle atrophy is the induction of oxidative stress by reactive oxygen species (ROS). This study aimed to identify the antioxidant mechanism of linoleic acid (LA) in muscle atrophy caused by oxidative stress. H2O2 has been used to induce oxidative stress in myoblasts in vitro. C2C12 myoblasts treated with H2O2 exhibited decreased viability and increased ROS synthesis. However, with LA treatment, the cells tended to recover from oxidative effects similar to those of the control groups. At the molecular level, the expression of superoxide dismutase 1 (SOD1), Bax, heat shock protein 70 (HSP70), and phosphorylated forkhead box protein O1 was increased by oxidative stress, causing apoptosis. LA treatment suppressed these changes. In addition, the expression of MuRF1 and Atrogin-1/MAFbx mRNA increased under oxidative stress but not in the LA-treated group. Sciatic denervation of C57BL/6 mice manifested as atrophy of the skeletal muscle in micro-computed tomography (micro-CT). The protein expression levels of SOD1, HSP70, and MuRF1 did not differ between the atrophied muscle tissues and C2C12 myoblasts under oxidative stress. With LA treatment, muscle atrophy recovered and protein expression was restored to levels similar to those in the control. Therefore, this study suggests that LA may be a candidate substance for preventing muscle atrophy.
Collapse
Affiliation(s)
- Myung-Hun Lee
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Korea; (M.-H.L.); (J.-H.L.); (W.-J.K.); (S.H.K.); (S.-Y.K.)
| | - Jin-Ho Lee
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Korea; (M.-H.L.); (J.-H.L.); (W.-J.K.); (S.H.K.); (S.-Y.K.)
| | - Wan-Joong Kim
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Korea; (M.-H.L.); (J.-H.L.); (W.-J.K.); (S.H.K.); (S.-Y.K.)
| | - Seo Ho Kim
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Korea; (M.-H.L.); (J.-H.L.); (W.-J.K.); (S.H.K.); (S.-Y.K.)
| | - Sun-Young Kim
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Korea; (M.-H.L.); (J.-H.L.); (W.-J.K.); (S.H.K.); (S.-Y.K.)
| | - Han Sung Kim
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Korea;
| | - Tack-Joong Kim
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Korea; (M.-H.L.); (J.-H.L.); (W.-J.K.); (S.H.K.); (S.-Y.K.)
- Research & Development Center, Doctor TJ Co., Ltd., Wonju 26493, Korea
| |
Collapse
|
11
|
Roux-Biejat P, Coazzoli M, Marrazzo P, Zecchini S, Di Renzo I, Prata C, Napoli A, Moscheni C, Giovarelli M, Barbalace MC, Catalani E, Bassi MT, De Palma C, Cervia D, Malaguti M, Hrelia S, Clementi E, Perrotta C. Acid Sphingomyelinase Controls Early Phases of Skeletal Muscle Regeneration by Shaping the Macrophage Phenotype. Cells 2021; 10:3028. [PMID: 34831250 PMCID: PMC8616363 DOI: 10.3390/cells10113028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle regeneration is a complex process involving crosstalk between immune cells and myogenic precursor cells, i.e., satellite cells. In this scenario, macrophage recruitment in damaged muscles is a mandatory step for tissue repair since pro-inflammatory M1 macrophages promote the activation of satellite cells, stimulating their proliferation and then, after switching into anti-inflammatory M2 macrophages, they prompt satellite cells' differentiation into myotubes and resolve inflammation. Here, we show that acid sphingomyelinase (ASMase), a key enzyme in sphingolipid metabolism, is activated after skeletal muscle injury induced in vivo by the injection of cardiotoxin. ASMase ablation shortens the early phases of skeletal muscle regeneration without affecting satellite cell behavior. Of interest, ASMase regulates the balance between M1 and M2 macrophages in the injured muscles so that the absence of the enzyme reduces inflammation. The analysis of macrophage populations indicates that these events depend on the altered polarization of M1 macrophages towards an M2 phenotype. Our results unravel a novel role of ASMase in regulating immune response during muscle regeneration/repair and suggest ASMase as a supplemental therapeutic target in conditions of redundant inflammation that impairs muscle recovery.
Collapse
Affiliation(s)
- Paulina Roux-Biejat
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Marco Coazzoli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Pasquale Marrazzo
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Ilaria Di Renzo
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Cecilia Prata
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, 40126 Bologna, Italy;
| | - Alessandra Napoli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Claudia Moscheni
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Maria Cristina Barbalace
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy; (E.C.); (D.C.)
| | - Maria Teresa Bassi
- Scientific Institute IRCCS “Eugenio Medea”, 23842 Bosisio Parini, Italy;
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, 20129 Milano, Italy;
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy; (E.C.); (D.C.)
| | - Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
- Scientific Institute IRCCS “Eugenio Medea”, 23842 Bosisio Parini, Italy;
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| |
Collapse
|
12
|
Valentino TR, Rule BD, Mobley CB, Nikolova-Karakashian M, Vechetti IJ. Skeletal Muscle Cell Growth Alters the Lipid Composition of Extracellular Vesicles. MEMBRANES 2021; 11:619. [PMID: 34436382 PMCID: PMC8397976 DOI: 10.3390/membranes11080619] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/31/2021] [Accepted: 08/11/2021] [Indexed: 02/01/2023]
Abstract
We sought to characterize the lipid profile of skeletal muscle cell-derived Extracellular Vesicles (EVs) to determine if a hypertrophic stimulus would affect the lipid composition of C2C12 myotube-derived EVs. Analyses included C2C12 murine myoblasts differentiated into myotubes and treated with Insulin-Like Growth Factor 1 (IGF-1) for 24 h to induce hypertrophic growth. EVs were isolated from cell culture media, quantified using Nanoparticle Tracking Analysis (NTA) and analyzed using Transmission Electron Microscopy (TEM). EVs were homogenized and lipids extracted for quantification by Mass Spectrometry followed by downstream lipid class enrichment and lipid chain analysis. IGF-1 treatment elicited an increase in CD63 and CD81 levels (39% and 21%) compared to the controls (16%), respectively. Analysis revealed that skeletal muscle-derived EVs are enriched in bioactive lipids that are likely selectively incorporated into EVs during hypertrophic growth. IGF-1 treatment of myotubes had a significant impact on the levels of diacylglycerol (DG) and ceramide (Cer) in secreted EVs. Specifically, the proportion of unsaturated DG was two- to three-fold higher in EVs derived from IGF-treated cells, as compared to those from control cells. The levels of saturated DG were unaffected. Selective increases were similarly seen in C16- and C24-Cer but not in other species. Levels of free sphingoid bases tended to decrease, while those of sphingosine-1-phosphate was unaffected. Our results suggest that the lipid composition and biogenesis of skeletal muscle-derived EVs, are specific and highly selective during hypertrophic growth.
Collapse
Affiliation(s)
- Taylor R. Valentino
- Department of Physiology, College of Medicine, Lexington, KY 40536, USA; (T.R.V.); (C.B.M.); (M.N.-K.)
| | - Blake D. Rule
- Department of Nutrition and Health Sciences, College of Education and Human Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| | - C. Brooks Mobley
- Department of Physiology, College of Medicine, Lexington, KY 40536, USA; (T.R.V.); (C.B.M.); (M.N.-K.)
| | | | - Ivan J. Vechetti
- Department of Nutrition and Health Sciences, College of Education and Human Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| |
Collapse
|
13
|
Pierucci F, Frati A, Battistini C, Penna F, Costelli P, Meacci E. Control of Skeletal Muscle Atrophy Associated to Cancer or Corticosteroids by Ceramide Kinase. Cancers (Basel) 2021; 13:3285. [PMID: 34209043 PMCID: PMC8269416 DOI: 10.3390/cancers13133285] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/18/2021] [Accepted: 06/25/2021] [Indexed: 01/19/2023] Open
Abstract
Apart from cytokines and chemokines, sphingolipid mediators, particularly sphingosine-1-phosphate (S1P) and ceramide 1-phosphate (C1P), contribute to cancer and inflammation. Cancer, as well as other inflammatory conditions, are associated with skeletal muscle (SkM) atrophy, which is characterized by the unbalance between protein synthesis and degradation. Although the signaling pathways involved in SkM mass wasting are multiple, the regulatory role of simple sphingolipids is limited. Here, we report the impairment of ceramide kinase (CerK), the enzyme responsible for the phosphorylation of ceramide to C1P, associated with the accomplishment of atrophic phenotype in various experimental models of SkM atrophy: in vivo animal model bearing the C26 adenocarcinoma or Lewis lung carcinoma tumors, in human and murine SkM cells treated with the conditioned medium obtained from cancer cells or with the glucocorticoid dexamethasone. Notably, we demonstrate in all the three experimental approaches a drastic decrease of CerK expression. Gene silencing of CerK promotes the up-regulation of atrogin-1/MAFbx expression, which was also observed after cell treatment with C8-ceramide, a biologically active ceramide analogue. Conversely, C1P treatment significantly reduced the corticosteroid's effects. Altogether, these findings provide evidence that CerK, acting as a molecular modulator, may be a new possible target for SkM mass regulation associated with cancer or corticosteroids.
Collapse
Affiliation(s)
- Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”—Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy; (F.P.); (A.F.); (C.B.)
| | - Alessia Frati
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”—Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy; (F.P.); (A.F.); (C.B.)
| | - Chiara Battistini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”—Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy; (F.P.); (A.F.); (C.B.)
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Turin, 10125 Torino, Italy; (F.P.); (P.C.)
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Turin, 10125 Torino, Italy; (F.P.); (P.C.)
| | - Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”—Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy; (F.P.); (A.F.); (C.B.)
| |
Collapse
|
14
|
Joseph J, Doles JD. Disease-associated metabolic alterations that impact satellite cells and muscle regeneration: perspectives and therapeutic outlook. Nutr Metab (Lond) 2021; 18:33. [PMID: 33766031 PMCID: PMC7992337 DOI: 10.1186/s12986-021-00565-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 03/15/2021] [Indexed: 11/10/2022] Open
Abstract
Many chronic disease patients experience a concurrent loss of lean muscle mass. Skeletal muscle is a dynamic tissue maintained by continuous protein turnover and progenitor cell activity. Muscle stem cells, or satellite cells, differentiate (by a process called myogenesis) and fuse to repair and regenerate muscle. During myogenesis, satellite cells undergo extensive metabolic alterations; therefore, pathologies characterized by metabolic derangements have the potential to impair myogenesis, and consequently exacerbate skeletal muscle wasting. How disease-associated metabolic disruptions in satellite cells might be contributing to wasting is an important question that is largely neglected. With this review we highlight the impact of various metabolic disruptions in disease on myogenesis and skeletal muscle regeneration. We also discuss metabolic therapies with the potential to improve myogenesis, skeletal muscle regeneration, and ultimately muscle mass.
Collapse
Affiliation(s)
- Josiane Joseph
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, MN, USA
| | - Jason D Doles
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
15
|
Takehara M, Kobayashi K, Nagahama M. Clostridium perfringens α-toxin inhibits myogenic differentiation of C2C12 myoblasts. Anaerobe 2020; 65:102265. [PMID: 32860931 DOI: 10.1016/j.anaerobe.2020.102265] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/03/2020] [Accepted: 08/24/2020] [Indexed: 10/23/2022]
Abstract
Clostridium perfringens type A is the causative agent of clostridial myonecrosis, and α-toxin has been reported to be responsible for the pathogenesis. Recently, it was reported that regeneration of skeletal muscle after C. perfringens-induced muscle disorders is delayed, but the detailed mechanisms have not been elucidated. Here, we tested whether α-toxin impairs the differentiation of C2C12 myoblasts, a useful cell line to study muscle growth, maturation, and regeneration in vitro. α-Toxin dose-dependently inhibited myotube formation in C2C12 cultures after induction of their differentiation by horse serum. Also, immunoblot analysis revealed that α-toxin dose-dependently decreases the expressions of two skeletal muscle differentiation markers, myogenic differentiation 1 (MyoD) and myogenin. These results demonstrate that α-toxin impairs the myogenic differentiation of C2C12 myoblasts. To reveal the mechanism behind α-toxin-mediated impairment of myogenic differentiation, we focused on ceramide production since α-toxin is known to promote the formation of ceramide by its sphingomyelinase activity. Immunofluorescent analysis revealed that ceramide production is accelerated by treatment with α-toxin. Furthermore, a synthetic cell-permeable ceramide analog, C2-ceramide, inhibited myotube formation in C2C12 cells and decreased the expressions of MyoD and myogenin, suggesting that accelerated ceramide production is involved in the α-toxin-mediated blockage of myogenic differentiation. Together, our results illustrate that the impairment of myogenic differentiation by α-toxin might be crucial for the pathogenesis of C. perfringens to delay regeneration of severely damaged skeletal muscles.
Collapse
Affiliation(s)
- Masaya Takehara
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514, Japan.
| | - Keiko Kobayashi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514, Japan
| | - Masahiro Nagahama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514, Japan.
| |
Collapse
|
16
|
Geiger AE, Daughtry MR, Yen C, Kirkpatrick LT, Shi H, Gerrard DE. Dual effects of obesity on satellite cells and muscle regeneration. Physiol Rep 2020; 8:e14511. [PMID: 32776502 PMCID: PMC7415910 DOI: 10.14814/phy2.14511] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
Obesity is a complex metabolic disorder that often leads to a decrease in insulin sensitivity, chronic inflammation, and overall decline in human health and well-being. In mouse skeletal muscle, obesity has been shown to impair muscle regeneration after injury; however, the mechanism underlying these changes has yet to be determined. To test whether there is a negative impact of obesity on satellite cell (SC) decisions and behaviors, we fed C57BL/6 mice normal chow (NC, control) or a high-fat diet (HFD) for 10 weeks and performed SC proliferation and differentiation assays in vitro. SCs from HFD mice formed colonies with smaller size (p < .001) compared to those from NC mice, and this decreased proliferation was confirmed (p < .05) by BrdU incorporation. Moreover, in vitro assays showed that HFD SCs exhibited diminished (p < .001) fusion capacity compared to NC SCs. In single fiber explants, a higher ratio of SCs experienced apoptotic events (p < .001) in HFD mice compared to that of NC-fed mice. In vivo lineage tracing using H2B-GFP mice showed that SCs from HFD treatment also cycled faster (p < .001) than their NC counterparts. In spite of all these autonomous cellular effects, obesity as triggered by high-fat feeding did not significantly impair muscle regeneration in vivo, as reflected by the comparable cross-sectional area (p > .05) of the regenerating fibers in HFD and NC muscles, suggesting that other factors may mitigate the negative impact of obesity on SCs properties.
Collapse
Affiliation(s)
- Ashley E. Geiger
- Department of Animal and Poultry SciencesVirginia Polytechnic Institute and State UniversityBlacksburgVAUSA
| | - Morgan R. Daughtry
- Department of Animal and Poultry SciencesVirginia Polytechnic Institute and State UniversityBlacksburgVAUSA
| | - Con‐Ning Yen
- Department of Animal and Poultry SciencesVirginia Polytechnic Institute and State UniversityBlacksburgVAUSA
| | - Laila T. Kirkpatrick
- Department of Animal and Poultry SciencesVirginia Polytechnic Institute and State UniversityBlacksburgVAUSA
| | - Hao Shi
- Department of Animal and Poultry SciencesVirginia Polytechnic Institute and State UniversityBlacksburgVAUSA
| | - David E. Gerrard
- Department of Animal and Poultry SciencesVirginia Polytechnic Institute and State UniversityBlacksburgVAUSA
| |
Collapse
|
17
|
Mankhong S, Kim S, Moon S, Kwak HB, Park DH, Kang JH. Experimental Models of Sarcopenia: Bridging Molecular Mechanism and Therapeutic Strategy. Cells 2020; 9:E1385. [PMID: 32498474 PMCID: PMC7348939 DOI: 10.3390/cells9061385] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
Sarcopenia has been defined as a progressive decline of skeletal muscle mass, strength, and functions in elderly people. It is accompanied by physical frailty, functional disability, falls, hospitalization, and mortality, and is becoming a major geriatric disorder owing to the increasing life expectancy and growing older population worldwide. Experimental models are critical to understand the pathophysiology of sarcopenia and develop therapeutic strategies. Although its etiologies remain to be further elucidated, several mechanisms of sarcopenia have been identified, including cellular senescence, proteostasis imbalance, oxidative stress, and "inflammaging." In this article, we address three main aspects. First, we describe the fundamental aging mechanisms. Next, we discuss both in vitro and in vivo experimental models based on molecular mechanisms that have the potential to elucidate the biochemical processes integral to sarcopenia. The use of appropriate models to reflect sarcopenia and/or its underlying pathways will enable researchers to understand sarcopenia and develop novel therapeutic strategies for sarcopenia. Lastly, we discuss the possible molecular targets and the current status of drug candidates for sarcopenia treatment. In conclusion, the development of experimental models for sarcopenia is essential to discover molecular targets that are valuable as biochemical biomarkers and/or therapeutic targets for sarcopenia.
Collapse
Affiliation(s)
- Sakulrat Mankhong
- Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.); (S.M.)
| | - Sujin Kim
- Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.); (S.M.)
- Department of Kinesiology, Inha University, Incheon 22212, Korea; (H.-B.K.); (D.-H.P.)
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
| | - Sohee Moon
- Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.); (S.M.)
| | - Hyo-Bum Kwak
- Department of Kinesiology, Inha University, Incheon 22212, Korea; (H.-B.K.); (D.-H.P.)
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
| | - Dong-Ho Park
- Department of Kinesiology, Inha University, Incheon 22212, Korea; (H.-B.K.); (D.-H.P.)
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
| | - Ju-Hee Kang
- Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.); (S.M.)
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
| |
Collapse
|
18
|
Tan-Chen S, Guitton J, Bourron O, Le Stunff H, Hajduch E. Sphingolipid Metabolism and Signaling in Skeletal Muscle: From Physiology to Physiopathology. Front Endocrinol (Lausanne) 2020; 11:491. [PMID: 32849282 PMCID: PMC7426366 DOI: 10.3389/fendo.2020.00491] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
Sphingolipids represent one of the major classes of eukaryotic lipids. They play an essential structural role, especially in cell membranes where they also possess signaling properties and are capable of modulating multiple cell functions, such as apoptosis, cell proliferation, differentiation, and inflammation. Many sphingolipid derivatives, such as ceramide, sphingosine-1-phosphate, and ganglioside, have been shown to play many crucial roles in muscle under physiological and pathological conditions. This review will summarize our knowledge of sphingolipids and their effects on muscle fate, highlighting the role of this class of lipids in modulating muscle cell differentiation, regeneration, aging, response to insulin, and contraction. We show that modulating sphingolipid metabolism may be a novel and interesting way for preventing and/or treating several muscle-related diseases.
Collapse
Affiliation(s)
- Sophie Tan-Chen
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Jeanne Guitton
- Université Saclay, CNRS UMR 9197, Institut des Neurosciences Paris-Saclay, Orsay, France
| | - Olivier Bourron
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique-Hôpitaux de Paris, Département de Diabétologie et Maladies Métaboliques, Hôpital Pitié-Salpêtrière, Paris, France
| | - Hervé Le Stunff
- Université Saclay, CNRS UMR 9197, Institut des Neurosciences Paris-Saclay, Orsay, France
| | - Eric Hajduch
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- *Correspondence: Eric Hajduch
| |
Collapse
|
19
|
Nguyen JH, Chung JD, Lynch GS, Ryall JG. The Microenvironment Is a Critical Regulator of Muscle Stem Cell Activation and Proliferation. Front Cell Dev Biol 2019; 7:254. [PMID: 31737625 PMCID: PMC6828616 DOI: 10.3389/fcell.2019.00254] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle has a remarkable capacity to regenerate following injury, a property conferred by a resident population of muscle stem cells (MuSCs). In response to injury, MuSCs must double their cellular content to divide, a process requiring significant new biomass in the form of nucleotides, phospholipids, and amino acids. This new biomass is derived from a series of intracellular metabolic cycles and alternative routing of carbon. In this review, we examine the link between metabolism and skeletal muscle regeneration with particular emphasis on the role of the cellular microenvironment in supporting the production of new biomass and MuSC proliferation.
Collapse
Affiliation(s)
- John H Nguyen
- Department of Physiology, Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, Australia
| | - Jin D Chung
- Department of Physiology, Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, Australia
| | - Gordon S Lynch
- Department of Physiology, Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, Australia
| | - James G Ryall
- Department of Physiology, Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
20
|
Beals JW, Burd NA, Moore DR, van Vliet S. Obesity Alters the Muscle Protein Synthetic Response to Nutrition and Exercise. Front Nutr 2019; 6:87. [PMID: 31263701 PMCID: PMC6584965 DOI: 10.3389/fnut.2019.00087] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 05/23/2019] [Indexed: 12/17/2022] Open
Abstract
Improving the health of skeletal muscle is an important component of obesity treatment. Apart from allowing for physical activity, skeletal muscle tissue is fundamental for the regulation of postprandial macronutrient metabolism, a time period that represents when metabolic derangements are most often observed in adults with obesity. In order for skeletal muscle to retain its capacity for physical activity and macronutrient metabolism, its protein quantity and composition must be maintained through the efficient degradation and resynthesis for proper tissue homeostasis. Life-style behaviors such as increasing physical activity and higher protein diets are front-line treatment strategies to enhance muscle protein remodeling by primarily stimulating protein synthesis rates. However, the muscle of individuals with obesity appears to be resistant to the anabolic action of targeted exercise regimes and protein ingestion when compared to normal-weight adults. This indicates impaired muscle protein remodeling in response to the main anabolic stimuli to human skeletal muscle tissue is contributing to poor muscle health with obesity. Deranged anabolic signaling related to insulin resistance, lipid accumulation, and/or systemic/muscle inflammation are likely at the root of the anabolic resistance of muscle protein synthesis rates with obesity. The purpose of this review is to discuss the impact of protein ingestion and exercise on muscle protein remodeling in people with obesity, and the potential mechanisms underlining anabolic resistance of their muscle.
Collapse
Affiliation(s)
- Joseph W Beals
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, United States
| | - Nicholas A Burd
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Daniel R Moore
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| | - Stephan van Vliet
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
21
|
Abdallah D, Skafi N, Hamade E, Borel M, Reibel S, Vitale N, El Jamal A, Bougault C, Laroche N, Vico L, Badran B, Hussein N, Magne D, Buchet R, Brizuela L, Mebarek S. Effects of phospholipase D during cultured osteoblast mineralization and bone formation. J Cell Biochem 2018; 120:5923-5935. [DOI: 10.1002/jcb.27881] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/20/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Dina Abdallah
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS Lyon France
- Lebanese University, Faculty of Sciences, Campus Rafic Hariri‐Hadath‐Beirut‐Liban Genomic and Health Laboratory/PRASE‐EDST Beirut Lebanon
| | - Najwa Skafi
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS Lyon France
- Lebanese University, Faculty of Sciences, Campus Rafic Hariri‐Hadath‐Beirut‐Liban Genomic and Health Laboratory/PRASE‐EDST Beirut Lebanon
| | - Eva Hamade
- Lebanese University, Faculty of Sciences, Campus Rafic Hariri‐Hadath‐Beirut‐Liban Genomic and Health Laboratory/PRASE‐EDST Beirut Lebanon
| | - Mathieu Borel
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS Lyon France
| | | | - Nicolas Vitale
- Centre National de la Recherche Scientifique (CNRS) UPR‐3212 and Université de Strasbourg Strasbourg France
| | - Alaeddine El Jamal
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS Lyon France
| | - Carole Bougault
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS Lyon France
| | - Norbert Laroche
- Univ Lyon, Université Jean Monnet, Faculté de Médecine, Campus Santé Innovation, INSERM UMR 1059, Sainbiose, LBTO Saint‐Etienne France
| | - Laurence Vico
- Univ Lyon, Université Jean Monnet, Faculté de Médecine, Campus Santé Innovation, INSERM UMR 1059, Sainbiose, LBTO Saint‐Etienne France
| | - Bassam Badran
- Lebanese University, Faculty of Sciences, Campus Rafic Hariri‐Hadath‐Beirut‐Liban Genomic and Health Laboratory/PRASE‐EDST Beirut Lebanon
| | - Nader Hussein
- Lebanese University, Faculty of Sciences, Campus Rafic Hariri‐Hadath‐Beirut‐Liban Genomic and Health Laboratory/PRASE‐EDST Beirut Lebanon
| | - David Magne
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS Lyon France
| | - Rene Buchet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS Lyon France
| | - Leyre Brizuela
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS Lyon France
| | - Saida Mebarek
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS Lyon France
| |
Collapse
|
22
|
Rasool S, Geetha T, Broderick TL, Babu JR. High Fat With High Sucrose Diet Leads to Obesity and Induces Myodegeneration. Front Physiol 2018; 9:1054. [PMID: 30258366 PMCID: PMC6143817 DOI: 10.3389/fphys.2018.01054] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/16/2018] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle utilizes both free fatty acids (FFAs) and glucose that circulate in the blood stream. When blood glucose levels acutely increase, insulin stimulates muscle glucose uptake, oxidation, and glycogen synthesis. Under these conditions, skeletal muscle preferentially oxidizes glucose while the oxidation of fatty acids (FAs) oxidation is reciprocally decreased. In metabolic disorders associated with insulin resistance, such as diabetes and obesity, both glucose uptake, and utilization muscle are significantly reduced causing FA oxidation to provide the majority of ATP for metabolic processes and contraction. Although the causes of this metabolic inflexibility or disrupted "glucose-fatty acid cycle" are largely unknown, a diet high in fat and sugar (HFS) may be a contributing factor. This metabolic inflexibility observed in models of obesity or with HFS feeding is detrimental because high rates of FA oxidation in skeletal muscle can lead to the buildup of toxic metabolites of fat metabolism and the accumulation of pro-inflammatory cytokines, which further exacerbate the insulin resistance. Further, HFS leads to skeletal muscle atrophy with a decrease in myofibrillar proteins and phenotypically characterized by loss of muscle mass and strength. Overactivation of ubiquitin proteasome pathway, oxidative stress, myonuclear apoptosis, and mitochondrial dysfunction are some of the mechanisms involved in muscle atrophy induced by obesity or in mice fed with HFS. In this review, we will discuss how HFS diet negatively impacts the various physiological and metabolic mechanisms in skeletal muscle.
Collapse
Affiliation(s)
- Suhail Rasool
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL, United States
| | - Thangiah Geetha
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL, United States
| | - Tom L Broderick
- Laboratory of Diabetes and Exercise Metabolism, Department of Physiology, Midwestern University, Glendale, AZ, United States
| | - Jeganathan R Babu
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL, United States
| |
Collapse
|
23
|
Bernacchioni C, Cencetti F, Ouro A, Bruno M, Gomez-Muñoz A, Donati C, Bruni P. Lysophosphatidic Acid Signaling Axis Mediates Ceramide 1-Phosphate-Induced Proliferation of C2C12 Myoblasts. Int J Mol Sci 2018; 19:ijms19010139. [PMID: 29300303 PMCID: PMC5796088 DOI: 10.3390/ijms19010139] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 12/23/2017] [Accepted: 12/28/2017] [Indexed: 02/07/2023] Open
Abstract
Sphingolipids are not only crucial for membrane architecture but act as critical regulators of cell functions. The bioactive sphingolipid ceramide 1-phosphate (C1P), generated by the action of ceramide kinase, has been reported to stimulate cell proliferation, cell migration and to regulate inflammatory responses via activation of different signaling pathways. We have previously shown that skeletal muscle is a tissue target for C1P since the phosphosphingolipid plays a positive role in myoblast proliferation implying a role in muscle regeneration. Skeletal muscle displays strong capacity of regeneration thanks to the presence of quiescent adult stem cells called satellite cells that upon trauma enter into the cell cycle and start proliferating. However, at present, the exact molecular mechanism by which C1P triggers its mitogenic effect in myoblasts is lacking. Here, we report for the first time that C1P stimulates C2C12 myoblast proliferation via lysophosphatidic acid (LPA) signaling axis. Indeed, C1P subsequently to phospholipase A2 activation leads to LPA1 and LPA3 engagement, which in turn drive Akt (protein kinase B) and ERK1/2 (extracellular signal-regulated kinases 1/2) activation, thus stimulating DNA synthesis. The present findings shed new light on the key role of bioactive sphingolipids in skeletal muscle and provide further support to the notion that these pleiotropic molecules might be useful therapeutic targets for skeletal muscle regeneration.
Collapse
Affiliation(s)
- Caterina Bernacchioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134 Firenze, Italy.
- Istituto Interuniversitario di Miologia (IIM), Italy.
| | - Francesca Cencetti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134 Firenze, Italy.
- Istituto Interuniversitario di Miologia (IIM), Italy.
| | - Alberto Ouro
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48080 Bilbao, Spain.
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| | - Marina Bruno
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134 Firenze, Italy.
| | - Antonio Gomez-Muñoz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48080 Bilbao, Spain.
| | - Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134 Firenze, Italy.
- Istituto Interuniversitario di Miologia (IIM), Italy.
| | - Paola Bruni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134 Firenze, Italy.
- Istituto Interuniversitario di Miologia (IIM), Italy.
| |
Collapse
|
24
|
Baraibar MA, Hyzewicz J, Rogowska-Wrzesinska A, Bulteau AL, Prip-Buus C, Butler-Browne G, Friguet B. Impaired energy metabolism of senescent muscle satellite cells is associated with oxidative modifications of glycolytic enzymes. Aging (Albany NY) 2017; 8:3375-3389. [PMID: 27922824 PMCID: PMC5270674 DOI: 10.18632/aging.101126] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/16/2016] [Indexed: 01/05/2023]
Abstract
Accumulation of oxidized proteins is a hallmark of cellular and organismal aging. Adult muscle stem cell (or satellite cell) replication and differentiation is compromised with age contributing to sarcopenia. However, the molecular events related to satellite cell dysfunction during aging are not completely understood. In the present study we have addressed the potential impact of oxidatively modified proteins on the altered metabolism of senescent human satellite cells. By using a modified proteomics analysis we have found that proteins involved in protein quality control and glycolytic enzymes are the main targets of oxidation (carbonylation) and modification with advanced glycation/lipid peroxidation end products during the replicative senescence of satellite cells. Inactivation of the proteasome appeared to be a likely contributor to the accumulation of such damaged proteins. Metabolic and functional analyses revealed an impaired glucose metabolism in senescent cells. A metabolic shift leading to increased mobilization of non-carbohydrate substrates such as branched chain amino acids or long chain fatty acids was observed. Increased levels of acyl-carnitines indicated an increased turnover of storage and membrane lipids for energy production. Taken together, these results support a link between oxidative protein modifications and the altered cellular metabolism associated with the senescent phenotype of human myoblasts.
Collapse
Affiliation(s)
- Martín A Baraibar
- Sorbonne Universités, UPMC Univ Paris 06, UMR 8256, Biological Adaptation and Ageing- IBPS, CNRS UMR 8256, INSERM U1164, Paris, France
| | - Janek Hyzewicz
- Sorbonne Universités, UPMC Univ Paris 06, UMR 8256, Biological Adaptation and Ageing- IBPS, CNRS UMR 8256, INSERM U1164, Paris, France
| | | | - Anne-Laure Bulteau
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, INSERM U1016, CNRS UMR 8104, Institut Cochin, Paris, France
| | - Carina Prip-Buus
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, INSERM U1016, CNRS UMR 8104, Institut Cochin, Paris, France
| | - Gillian Butler-Browne
- Institut de Myologie, UPMC Univ Paris 06, UMRS INSERM U974, CNRS UMR 7215, CHU Pitié-Salpétrière, Sorbonne Universités, Paris, France
| | - Bertrand Friguet
- Sorbonne Universités, UPMC Univ Paris 06, UMR 8256, Biological Adaptation and Ageing- IBPS, CNRS UMR 8256, INSERM U1164, Paris, France
| |
Collapse
|
25
|
Rodriguez-Cuenca S, Pellegrinelli V, Campbell M, Oresic M, Vidal-Puig A. Sphingolipids and glycerophospholipids - The "ying and yang" of lipotoxicity in metabolic diseases. Prog Lipid Res 2017; 66:14-29. [PMID: 28104532 DOI: 10.1016/j.plipres.2017.01.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/30/2016] [Accepted: 01/05/2017] [Indexed: 12/14/2022]
Abstract
Sphingolipids in general and ceramides in particular, contribute to pathophysiological mechanisms by modifying signalling and metabolic pathways. Here, we present the available evidence for a bidirectional homeostatic crosstalk between sphingolipids and glycerophospholipids, whose dysregulation contributes to lipotoxicity induced metabolic stress. The initial evidence for this crosstalk originates from simulated models designed to investigate the biophysical properties of sphingolipids in plasma membrane representations. In this review, we reinterpret some of the original findings and conceptualise them as a sort of "ying/yang" interaction model of opposed/complementary forces, which is consistent with the current knowledge of lipid homeostasis and pathophysiology. We also propose that the dysregulation of the balance between sphingolipids and glycerophospholipids results in a lipotoxic insult relevant in the pathophysiology of common metabolic diseases, typically characterised by their increased ceramide/sphingosine pools.
Collapse
Affiliation(s)
- S Rodriguez-Cuenca
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge. Cambridge, UK.
| | - V Pellegrinelli
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge. Cambridge, UK
| | - M Campbell
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge. Cambridge, UK
| | - M Oresic
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, FI -20520 Turku, Finland
| | - A Vidal-Puig
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge. Cambridge, UK; Wellcome Trust Sanger Institute, Hinxton, UK.
| |
Collapse
|
26
|
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: 170] [Impact Index Per Article: 21.3] [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
|
27
|
Saini A, Sharples AP, Al-Shanti N, Stewart CE. Omega-3 fatty acid EPA improves regenerative capacity of mouse skeletal muscle cells exposed to saturated fat and inflammation. Biogerontology 2016; 18:109-129. [PMID: 27864687 PMCID: PMC5288450 DOI: 10.1007/s10522-016-9667-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/05/2016] [Indexed: 12/17/2022]
Abstract
Sarcopenic obesity is characterised by high fat mass, low muscle mass and an elevated inflammatory environmental milieu. We therefore investigated the effects of elevated inflammatory cytokine TNF-α (aging/obesity) and saturated fatty acid, palmitate (obesity) on skeletal muscle cells in the presence/absence of EPA, a-3 polyunsaturated fatty acid with proposed anti-inflammatory, anti-obesity activities. In the present study we show that palmitate was lipotoxic, inducing high levels of cell death and blocking myotube formation. Cell death under these conditions was associated with increased caspase activity, suppression of differentiation, reductions in both creatine kinase activity and gene expression of myogenic factors; IGF-II, IGFBP-5, MyoD and myogenin. However, inhibition of caspase activity via administration of Z-VDVAD-FMK (caspase-2), Z-DEVD-FMK (caspase-3) and ZIETD-KMK (caspase 8) was without effect on cell death. By contrast, lipotoxicity associated with elevated palmitate was reduced with the MEK inhibitor PD98059, indicating palmitate induced cell death was MAPK mediated. These lipotoxic conditions were further exacerbated in the presence of inflammation via TNF-α co-administration. Addition of EPA under cytotoxic stress (TNF-α) was shown to partially rescue differentiation with enhanced myotube formation being associated with increased MyoD, myogenin, IGF-II and IGFBP-5 expression. EPA had little impact on the cell death phenotype observed in lipotoxic conditions but did show benefit in restoring differentiation under lipotoxic plus cytotoxic conditions. Under these conditions Id3 (inhibitor of differentiation) gene expression was inversely linked with survival rates, potentially indicating a novel role of EPA and Id3 in the regulation of apoptosis in lipotoxic/cytotoxic conditions. Additionally, signalling studies indicated the combination of lipo- and cyto-toxic effects on the muscle cells acted through ceramide, JNK and MAPK pathways and blocking these pathways using PD98059 (MEK inhibitor) and Fumonisin B1 (ceramide inhibitor) significantly reduced levels of cell death. These findings highlight novel pathways associated with in vitro models of lipotoxicity (palmitate-mediated) and cytotoxicity (inflammatory cytokine mediated) in the potential targeting of molecular modulators of sarcopenic obesity.
Collapse
Affiliation(s)
- Amarjit Saini
- Department of Laboratory Medicine, Clinical Physiology, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, 141 86, Stockholm, Sweden
| | - Adam P Sharples
- Stem Cells, Ageing and Molecular Physiology (SCAMP) Unit, Exercise Metabolism and Adaptation Research Group, Research Institute for Sport & Exercise Sciences, School of Sport and Exercise Sciences, Liverpool John Moores University, Life Science Building, Byrom Street Campus, Liverpool, L3 3AF, UK.
| | - Nasser Al-Shanti
- Neuromuscular and Skeletal Ageing Research Group, Healthcare Science Research Institute, Manchester Metropolitan University, Oxford Road, Manchester, M1 5GD, UK
| | - Claire E Stewart
- Stem Cells, Ageing and Molecular Physiology (SCAMP) Unit, Exercise Metabolism and Adaptation Research Group, Research Institute for Sport & Exercise Sciences, School of Sport and Exercise Sciences, Liverpool John Moores University, Life Science Building, Byrom Street Campus, Liverpool, L3 3AF, UK
| |
Collapse
|
28
|
Zhang Y, Yu B, He J, Chen D. From Nutrient to MicroRNA: a Novel Insight into Cell Signaling Involved in Skeletal Muscle Development and Disease. Int J Biol Sci 2016; 12:1247-1261. [PMID: 27766039 PMCID: PMC5069446 DOI: 10.7150/ijbs.16463] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/19/2016] [Indexed: 12/17/2022] Open
Abstract
Skeletal muscle is a remarkably complicated organ comprising many different cell types, and it plays an important role in lifelong metabolic health. Nutrients, as an external regulator, potently regulate skeletal muscle development through various internal regulatory factors, such as mammalian target of rapamycin (mTOR) and microRNAs (miRNAs). As a nutrient sensor, mTOR, integrates nutrient availability to regulate myogenesis and directly or indirectly influences microRNA expression. MiRNAs, a class of small non-coding RNAs mediating gene silencing, are implicated in myogenesis and muscle-related diseases. Meanwhile, growing evidence has emerged supporting the notion that the expression of myogenic miRNAs could be regulated by nutrients in an epigenetic mechanism. Therefore, this review presents a novel insight into the cell signaling network underlying nutrient-mTOR-miRNA pathway regulation of skeletal myogenesis and summarizes the epigenetic modifications in myogenic differentiation, which will provide valuable information for potential therapeutic intervention.
Collapse
Affiliation(s)
- Yong Zhang
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China.; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China.; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China.; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China.; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, China
| |
Collapse
|
29
|
Salaun E, Lefeuvre-Orfila L, Cavey T, Martin B, Turlin B, Ropert M, Loreal O, Derbré F. Myriocin prevents muscle ceramide accumulation but not muscle fiber atrophy during short-term mechanical unloading. J Appl Physiol (1985) 2015; 120:178-87. [PMID: 26542521 DOI: 10.1152/japplphysiol.00720.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/04/2015] [Indexed: 01/24/2023] Open
Abstract
Bedridden patients in intensive care unit or after surgery intervention commonly develop skeletal muscle weakness. The latter is promoted by a variety of prolonged hospitalization-associated conditions. Muscle disuse is the most ubiquitous and contributes to rapid skeletal muscle atrophy and progressive functional strength reduction. Disuse causes a reduction in fatty acid oxidation, leading to its accumulation in skeletal muscle. We hypothesized that muscle fatty acid accumulation could stimulate ceramide synthesis and promote skeletal muscle weakness. Therefore, the present study was designed to determine the effects of sphingolipid metabolism on skeletal muscle atrophy induced by 7 days of disuse. For this purpose, male Wistar rats were treated with myriocin, an inhibitor of de novo synthesis of ceramides, and subjected to hindlimb unloading (HU) for 7 days. Soleus muscles were assayed for fiber diameter, ceramide levels, protein degradation, and apoptosis signaling. Serum and liver were removed to evaluate the potential hepatoxicity of myriocin treatment. We found that HU increases content of saturated C16:0 and C18:0 ceramides and decreases soleus muscle weight and fiber diameter. HU increased the level of polyubiquitinated proteins and induced apoptosis in skeletal muscle. Despite a prevention of C16:0 and C18:0 muscle accumulation, myriocin treatment did not prevent skeletal muscle atrophy and concomitant induction of apoptosis and proteolysis. Moreover, myriocin treatment increased serum transaminases and induced hepatocyte necrosis. These data highlight that inhibition of de novo synthesis of ceramides during immobilization is not an efficient strategy to prevent skeletal muscle atrophy and exerts adverse effects like hepatotoxicity.
Collapse
Affiliation(s)
- Erwann Salaun
- Laboratory "Movement Sport and Health Sciences," University Rennes 2-ENS Rennes, Bruz, France
| | - Luz Lefeuvre-Orfila
- Laboratory "Movement Sport and Health Sciences," University Rennes 2-ENS Rennes, Bruz, France
| | - Thibault Cavey
- INSERM UMR 991, Iron and the Liver Team Rennes, Faculty of Medicine, University of Rennes 1, Rennes, France; Laboratory of Biochemistry, University Hospital Pontchaillou, Rennes, France
| | - Brice Martin
- Laboratory "Movement Sport and Health Sciences," University Rennes 2-ENS Rennes, Bruz, France
| | - Bruno Turlin
- INSERM UMR 991, Iron and the Liver Team Rennes, Faculty of Medicine, University of Rennes 1, Rennes, France; Department of Pathology, University Hospital Pontchaillou, Rennes, France
| | - Martine Ropert
- INSERM UMR 991, Iron and the Liver Team Rennes, Faculty of Medicine, University of Rennes 1, Rennes, France; Laboratory of Biochemistry, University Hospital Pontchaillou, Rennes, France
| | - Olivier Loreal
- INSERM UMR 991, Iron and the Liver Team Rennes, Faculty of Medicine, University of Rennes 1, Rennes, France
| | - Frédéric Derbré
- Laboratory "Movement Sport and Health Sciences," University Rennes 2-ENS Rennes, Bruz, France;
| |
Collapse
|
30
|
Lee YH, Kim WJ, Lee MH, Kim SY, Seo DH, Kim HS, Gelinsky M, Kim TJ. Anti-skeletal muscle atrophy effect of Oenothera odorata root extract via reactive oxygen species-dependent signaling pathways in cellular and mouse model. Biosci Biotechnol Biochem 2015; 80:80-8. [PMID: 26613402 DOI: 10.1080/09168451.2015.1075861] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Skeletal muscle atrophy can be defined as a decrease of muscle volume caused by injury or lack of use. This condition is associated with reactive oxygen species (ROS), resulting in various muscular disorders. We acquired 2D and 3D images using micro-computed tomography in gastrocnemius and soleus muscles of sciatic-denervated mice. We confirmed that sciatic denervation-small animal model reduced muscle volume. However, the intraperitoneal injection of Oenothera odorata root extract (EVP) delayed muscle atrophy compared to a control group. We also investigated the mechanism of muscle atrophy's relationship with ROS. EVP suppressed expression of SOD1, and increased expression of HSP70, in both H2O2-treated C2C12 myoblasts and sciatic-denervated mice. Moreover, EVP regulated apoptotic signals, including caspase-3, Bax, Bcl-2, and ceramide. These results indicate that EVP has a positive effect on reducing the effect of ROS on muscle atrophy.
Collapse
Affiliation(s)
- Yong-Hyeon Lee
- a Division of Biological Science and Technology, Yonsei-Fraunhofer Medical Device Lab , College of Science and Technology, Yonsei University , Wonju , Korea
| | - Wan-Joong Kim
- a Division of Biological Science and Technology, Yonsei-Fraunhofer Medical Device Lab , College of Science and Technology, Yonsei University , Wonju , Korea
| | - Myung-Hun Lee
- a Division of Biological Science and Technology, Yonsei-Fraunhofer Medical Device Lab , College of Science and Technology, Yonsei University , Wonju , Korea
| | - Sun-Young Kim
- a Division of Biological Science and Technology, Yonsei-Fraunhofer Medical Device Lab , College of Science and Technology, Yonsei University , Wonju , Korea
| | - Dong-Hyun Seo
- b Department of Biomedical Engineering, Yonsei-Fraunhofer Medical Device Lab , College of Health Science, Yonsei University , Wonju , Korea
| | - Han-Sung Kim
- b Department of Biomedical Engineering, Yonsei-Fraunhofer Medical Device Lab , College of Health Science, Yonsei University , Wonju , Korea
| | - Michael Gelinsky
- c Center for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Hospital, Technische Universität Dresden , Dresden , Germany
| | - Tack-Joong Kim
- a Division of Biological Science and Technology, Yonsei-Fraunhofer Medical Device Lab , College of Science and Technology, Yonsei University , Wonju , Korea
| |
Collapse
|
31
|
Effects of Aging and Experimentally Induced Modifications of Signal Pathways on Insulin-Induced Shifts of Glucose Metabolism in the Rat Neocortex. NEUROPHYSIOLOGY+ 2015. [DOI: 10.1007/s11062-015-9491-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
32
|
Babenko NA, Kharchenko VS. Modulation of Insulin Sensitivity of Hepatocytes by the Pharmacological Downregulation of Phospholipase D. Int J Endocrinol 2015; 2015:794838. [PMID: 26089893 PMCID: PMC4458285 DOI: 10.1155/2015/794838] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/30/2015] [Accepted: 05/11/2015] [Indexed: 12/13/2022] Open
Abstract
Background. The role of phospholipase D (PLD) as a positive modulator of glucose uptake activation by insulin in muscle and adipose cells has been demonstrated. The role of PLD in the regulation of glucose metabolism by insulin in the primary hepatocytes has been determined in this study. Methods. For this purpose, we studied effects of inhibitors of PLD on glucose uptake and glycogen synthesis stimulation by insulin. To determine the PLD activity, the method based on determination of products of transphosphatidylation reaction, phosphatidylethanol or phosphatidylbutanol, was used. Results. Inhibition of PLD by a general antagonist (1-butanol) or specific inhibitor, halopemide, or N-hexanoylsphingosine, or by cellular ceramides accumulated in doxorubicin-treated hepatocytes decreased insulin-stimulated glucose metabolism. Doxorubicin-induced hepatocytes resistance to insulin action could be abolished by inhibition of ceramide production. Halopemide could nullify this effect. Addition of propranolol, as well as inhibitors of phosphatidylinositol 3-kinase (PI3-kinase) (wortmannin, LY294002) or suppressors of Akt phosphorylation/activity, luteolin-7-O-glucoside or apigenin-7-O-glucoside, to the culture media could block cell response to insulin action. Conclusion. PLD plays an important role in the insulin signaling in the hepatocytes. PLD is activated downstream of PI3-kinase and Akt and is highly sensitive to ceramide content in the liver cells.
Collapse
Affiliation(s)
- Nataliya A. Babenko
- Department of Physiology of Ontogenesis, Biology Research Institute, Karazin Kharkov National University, Svobody Square 4, Kharkov 61022, Ukraine
- *Nataliya A. Babenko:
| | - Vitalina S. Kharchenko
- Department of Physiology of Ontogenesis, Biology Research Institute, Karazin Kharkov National University, Svobody Square 4, Kharkov 61022, Ukraine
| |
Collapse
|
33
|
Abstract
The importance of skeletal muscle for metabolic health and obesity prevention is gradually gaining recognition. As a result, interventions are being developed to increase or maintain muscle mass and metabolic function in adult and elderly populations. These interventions include exercise, hormonal and nutritional therapies. Nonetheless, growing evidence suggests that maternal malnutrition and obesity during pregnancy and lactation impede skeletal muscle development and growth in the offspring, with long-term functional consequences lasting into adult life. Here we review the role of skeletal muscle in health and obesity, providing an insight into how this tissue develops and discuss evidence that maternal obesity affects its development, growth and function into adult life. Such evidence warrants the need to develop early life interventions to optimise skeletal muscle development and growth in the offspring and thereby maximise metabolic health into adult life.
Collapse
|
34
|
Kakoi H, Maeda S, Shinohara N, Matsuyama K, Imamura K, Kawamura I, Nagano S, Setoguchi T, Yokouchi M, Ishidou Y, Komiya S. Bone morphogenic protein (BMP) signaling up-regulates neutral sphingomyelinase 2 to suppress chondrocyte maturation via the Akt protein signaling pathway as a negative feedback mechanism. J Biol Chem 2014; 289:8135-50. [PMID: 24505141 DOI: 10.1074/jbc.m113.509331] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although bone morphogenic protein (BMP) signaling promotes chondrogenesis, it is not clear whether BMP-induced chondrocyte maturation is cell-autonomously terminated. Loss of function of Smpd3 in mice results in an increase in mature hypertrophic chondrocytes. Here, we report that in chondrocytes the Runx2-dependent expression of Smpd3 was increased by BMP-2 stimulation. Neutral sphingomyelinase 2 (nSMase2), encoded by the Smpd3 gene, was detected both in prehypertrophic and hypertrophic chondrocytes of mouse embryo bone cartilage. An siRNA for Smpd3, as well as the nSMase inhibitor GW4869, significantly enhanced BMP-2-induced differentiation and maturation of chondrocytes. Conversely, overexpression of Smpd3 or C2-ceramide, which mimics the function of nSMase2, inhibited chondrogenesis. Upon induction of Smpd3 siRNA or GW4869, phosphorylation of both Akt and S6 proteins was increased. The accelerated chondrogenesis induced by Smpd3 silencing was negated by application of the Akt inhibitor MK2206 or the mammalian target of rapamycin inhibitor rapamycin. Importantly, in mouse bone culture, GW4869 treatment significantly promoted BMP-2-induced hypertrophic maturation and calcification of chondrocytes, which subsequently was eliminated by C2-ceramide. Smpd3 knockdown decreased the apoptosis of terminally matured ATDC5 chondrocytes, probably as a result of decreased ceramide production. In addition, we found that expression of hyaluronan synthase 2 (Has2) was elevated by a loss of Smpd3, which was restored by MK2206. Indeed, expression of Has2 protein decreased in nSMase2-positive hypertrophic chondrocytes in the bones of mouse embryos. Our data suggest that the Smpd3/nSMase2-ceramide-Akt signaling axis negatively regulates BMP-induced chondrocyte maturation and Has2 expression to control the rate of endochondral ossification as a negative feedback mechanism.
Collapse
|
35
|
Akhmedov D, Berdeaux R. The effects of obesity on skeletal muscle regeneration. Front Physiol 2013; 4:371. [PMID: 24381559 PMCID: PMC3865699 DOI: 10.3389/fphys.2013.00371] [Citation(s) in RCA: 164] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 11/28/2013] [Indexed: 12/18/2022] Open
Abstract
Obesity and metabolic disorders such as type 2 diabetes mellitus are accompanied by increased lipid deposition in adipose and non-adipose tissues including liver, pancreas, heart and skeletal muscle. Recent publications report impaired regenerative capacity of skeletal muscle following injury in obese mice. Although muscle regeneration has not been thoroughly studied in obese and type 2 diabetic humans and mechanisms leading to decreased muscle regeneration in obesity remain elusive, the initial findings point to the possibility that muscle satellite cell function is compromised under conditions of lipid overload. Elevated toxic lipid metabolites and increased pro-inflammatory cytokines as well as insulin and leptin resistance that occur in obese animals may contribute to decreased regenerative capacity of skeletal muscle. In addition, obesity-associated alterations in the metabolic state of skeletal muscle fibers and satellite cells may directly impair the potential for satellite cell-mediated repair. Here we discuss recent studies that expand our understanding of how obesity negatively impacts skeletal muscle maintenance and regeneration.
Collapse
Affiliation(s)
- Dmitry Akhmedov
- Department of Integrative Biology and Pharmacology and Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston Houston, TX, USA
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology and Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston Houston, TX, USA
| |
Collapse
|
36
|
Age-Related Changes in the Phospholipase D-Dependent Signal Pathway of Insulin in the Rat Neocortex. NEUROPHYSIOLOGY+ 2013. [DOI: 10.1007/s11062-013-9346-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
37
|
Ge Y, Chen J. Mammalian target of rapamycin (mTOR) signaling network in skeletal myogenesis. J Biol Chem 2012; 287:43928-35. [PMID: 23115234 DOI: 10.1074/jbc.r112.406942] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mammalian (or mechanistic) target of rapamycin (mTOR) regulates a wide range of cellular and developmental processes by coordinating signaling responses to mitogens, nutrients, and various stresses. Over the last decade, mTOR has emerged as a master regulator of skeletal myogenesis, controlling multiple stages of the myofiber formation process. In this minireview, we present an emerging view of the signaling network underlying mTOR regulation of myogenesis, which contrasts with the well established mechanisms in the regulation of cell and muscle growth. Current questions for future studies are also highlighted.
Collapse
Affiliation(s)
- Yejing Ge
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | | |
Collapse
|
38
|
Babenko NA, Kharchenko VS. Ceramides inhibit phospholipase D-dependent insulin signaling in liver cells of old rats. BIOCHEMISTRY (MOSCOW) 2012; 77:180-6. [DOI: 10.1134/s0006297912020095] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
39
|
TNF-α- and tumor-induced skeletal muscle atrophy involves sphingolipid metabolism. Skelet Muscle 2012; 2:2. [PMID: 22257771 PMCID: PMC3344678 DOI: 10.1186/2044-5040-2-2] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 01/18/2012] [Indexed: 11/11/2022] Open
Abstract
Background Muscle atrophy associated with various pathophysiological conditions represents a major health problem, because of its contribution to the deterioration of patient status and its effect on mortality. Although the involvement of pro-inflammatory cytokines in this process is well recognized, the role of sphingolipid metabolism alterations induced by the cytokines has received little attention. Results We addressed this question both in vitro using differentiated myotubes treated with TNF-α, and in vivo in a murine model of tumor-induced cachexia. Myotube atrophy induced by TNF-α was accompanied by a substantial increase in cell ceramide levels, and could be mimicked by the addition of exogenous ceramides. It could be prevented by the addition of ceramide-synthesis inhibitors that targeted either the de novo pathway (myriocin), or the sphingomyelinases (GW4869 and 3-O-methylsphingomyelin). In the presence of TNF-α, ceramide-synthesis inhibitors significantly increased protein synthesis and decreased proteolysis. In parallel, they lowered the expression of both the Atrogin-1 and LC3b genes, involved in muscle protein degradation by proteasome and in autophagic proteolysis, respectively, and increased the proportion of inactive, phosphorylated Foxo3 transcription factor. Furthermore, these inhibitors increased the expression and/or phosphorylation levels of key factors regulating protein metabolism, including phospholipase D, an activator of mammalian target of rapamycin (mTOR), and the mTOR substrates S6K1 and Akt. In vivo, C26 carcinoma implantation induced a substantial increase in muscle ceramide, together with drastic muscle atrophy. Treatment of the animals with myriocin reduced the expression of the atrogenes Foxo3 and Atrogin-1, and partially protected muscle tissue from atrophy. Conclusions Ceramide accumulation induced by TNF-α or tumor development participates in the mechanism of muscle-cell atrophy, and sphingolipid metabolism is a logical target for pharmacological or nutritional interventions aiming at preserving muscle mass in pathological situations.
Collapse
|
40
|
Nikolova-Karakashian MN, Reid MB. Sphingolipid metabolism, oxidant signaling, and contractile function of skeletal muscle. Antioxid Redox Signal 2011; 15:2501-17. [PMID: 21453197 PMCID: PMC3176343 DOI: 10.1089/ars.2011.3940] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 03/31/2011] [Indexed: 12/17/2022]
Abstract
SIGNIFICANCE Sphingolipids are a class of bioactive lipids that regulate diverse cell functions. Ceramide, sphingosine, and sphingosine-1-phosphate accumulate in tissues such as liver, brain, and lung under conditions of cellular stress, including oxidative stress. The activity of some sphingolipid metabolizing enzymes, chiefly the sphingomyelinases, is stimulated during inflammation and in response to oxidative stress. Ceramide, the sphingomyelinase product, as well as the ceramide metabolite, sphingosine-1-phosphate, can induce the generation of more reactive oxygen species, propagating further inflammation. RECENT ADVANCES This review article summarizes information on sphingolipid biochemistry and signaling pertinent to skeletal muscle and describes the potential influence of sphingolipids on contractile function. CRITICAL ISSUES It encompasses topics related to (1) the pathways for complex sphingolipid biosynthesis and degradation, emphasizing sphingolipid regulation in various muscle fiber types and subcellular compartments; (2) the emerging evidence that implicates ceramide, sphingosine, and sphingosine-1-phosphate as regulators of muscle oxidant activity, and (3) sphingolipid effects on contractile function and fatigue. FUTURE DIRECTIONS We propose that prolonged inflammatory conditions alter ceramide, sphingosine, and sphingosine-1-phosphate levels in skeletal muscle and that these changes promote the weakness, premature fatigue, and cachexia that plague individuals with heart failure, cancer, diabetes, and other chronic inflammatory diseases.
Collapse
|
41
|
Gangoiti P, Bernacchioni C, Donati C, Cencetti F, Ouro A, Gómez-Muñoz A, Bruni P. Ceramide 1-phosphate stimulates proliferation of C2C12 myoblasts. Biochimie 2011; 94:597-607. [PMID: 21945811 PMCID: PMC3314975 DOI: 10.1016/j.biochi.2011.09.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 09/12/2011] [Indexed: 12/11/2022]
Abstract
Recent studies have established specific cellular functions for different bioactive sphingolipids in skeletal muscle cells. Ceramide 1-phosphate (C1P) is an important bioactive sphingolipid that has been involved in cell growth and survival. However its possible role in the regulation of muscle cell homeostasis has not been so far investigated. In this study, we show that C1P stimulates myoblast proliferation, as determined by measuring the incorporation of tritiated thymidine into DNA, and progression of the myoblasts through the cell cycle. C1P induced phosphorylation of glycogen synthase kinase-3β and the product of retinoblastoma gene, and enhanced cyclin D1 protein levels. The mitogenic action of C1P also involved activation of phosphatidylinositol 3-kinase/Akt, ERK1/2 and the mammalian target of rapamycin. These effects of C1P were independent of interaction with a putative Gi-coupled C1P receptor as pertussis toxin, which maintains Gi protein in the inactive form, did not affect C1P-stimulated myoblast proliferation. By contrast, C1P was unable to inhibit serum starvation- or staurosporine-induced apoptosis in the myoblasts, and did not affect myogenic differentiation. Collectively, these results add up to the current knowledge on cell types targeted by C1P, which so far has been mainly confined to fibroblasts and macrophages, and extend on the mechanisms by which C1P exerts its mitogenic effects. Moreover, the biological activities of C1P described in this report establish that this phosphosphingolipid may be a relevant cue in the regulation of skeletal muscle regeneration, and that C1P-metabolizing enzymes might be important targets for developing cellular therapies for treatment of skeletal muscle degenerative diseases, or tissue injury.
Collapse
Affiliation(s)
- Patricia Gangoiti
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country, 48080 Bilbao, Spain
| | | | | | | | | | | | | |
Collapse
|
42
|
Han X, Yu R, Ji L, Zhen D, Tao S, Li S, Sun Y, Huang L, Feng Z, Li X, Han G, Schmidt M, Han L. InlB-mediated Listeria monocytogenes internalization requires a balanced phospholipase D activity maintained through phospho-cofilin. Mol Microbiol 2011; 81:860-80. [PMID: 21722201 DOI: 10.1111/j.1365-2958.2011.07726.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Internalization of Listeria monocytogenes into non-phagocytic cells is tightly controlled by host cell actin dynamics and cell membrane alterations. However, knowledge about the impact of phosphatidylcholine cleavage driven by host cell phospholipase D (PLD) on Listeria internalization into epithelial cells is limited. Here, we report that L. monocytogenes activates PLD in Vero cells during the internalization. With immunostaining it was shown that both PLD1 and PLD2 surrounded partially or completely the phagocytic cup of most L. monocytogenes. Either up- or down-regulation of PLD expression (activity) diminished Listeria internalization. Both PLD1 and PLD2 in Vero cells were required for efficient Listeria internalization, and could substitute for each other in the regulation of Listeria internalization. Further, exogenous InlB activated host cell PLD1 and PLD2 via the Met receptor, and restored host PLD activation by InlB-deficient L. monocytogenes. InlB-induced PLD activation and Listeria internalization were tightly controlled by phospho-cycling of cofilin. PLD1, but not PLD2, was involved in cofilin-mediated PLD activation and Listeria internalization. These data indicate that cofilin-dependent PLD activation induced by InlB may represent a novel regulation mechanism for efficient Listeria internalization into epithelial cells.
Collapse
Affiliation(s)
- Xuelin Han
- Department for Hospital Infection Control & Research, Institute of Disease Control & Prevention of PLA, Academy of Military Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Jaafar R, Zeiller C, Pirola L, Di Grazia A, Naro F, Vidal H, Lefai E, Némoz G. Phospholipase D regulates myogenic differentiation through the activation of both mTORC1 and mTORC2 complexes. J Biol Chem 2011; 286:22609-21. [PMID: 21525000 DOI: 10.1074/jbc.m110.203885] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
How phospholipase D (PLD) is involved in myogenesis remains unclear. At the onset of myogenic differentiation of L6 cells induced by the PLD agonist vasopressin in the absence of serum, mTORC1 complex was rapidly activated, as reflected by phosphorylation of S6 kinase1 (S6K1). Both the long (p85) and short (p70) S6K1 isoforms were phosphorylated in a PLD1-dependent way. Short rapamycin treatment specifically inhibiting mTORC1 suppressed p70 but not p85 phosphorylation, suggesting that p85 might be directly activated by phosphatidic acid. Vasopressin stimulation also induced phosphorylation of Akt on Ser-473 through PLD1-dependent activation of mTORC2 complex. In this model of myogenesis, mTORC2 had a positive role mostly unrelated to Akt activation, whereas mTORC1 had a negative role, associated with S6K1-induced Rictor phosphorylation. The PLD requirement for differentiation can thus be attributed to its ability to trigger via mTORC2 activation the phosphorylation of an effector that could be PKCα. Moreover, PLD is involved in a counter-regulation loop expected to limit the response. This study thus brings new insights in the intricate way PLD and mTOR cooperate to control myogenesis.
Collapse
|
44
|
Cowart LA. A novel role for sphingolipid metabolism in oxidant-mediated skeletal muscle fatigue. Focus on "Sphingomyelinase stimulates oxidant signaling to weaken skeletal muscle and promote fatigue". Am J Physiol Cell Physiol 2010; 299:C549-51. [PMID: 20573998 DOI: 10.1152/ajpcell.00236.2010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
45
|
Bach AS, Enjalbert S, Comunale F, Bodin S, Vitale N, Charrasse S, Gauthier-Rouvière C. ADP-ribosylation factor 6 regulates mammalian myoblast fusion through phospholipase D1 and phosphatidylinositol 4,5-bisphosphate signaling pathways. Mol Biol Cell 2010; 21:2412-24. [PMID: 20505075 PMCID: PMC2903670 DOI: 10.1091/mbc.e09-12-1063] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Here we show that ARF6 is associated with the multiproteic complex that contains M-cadherin, Trio, and Rac1 and accumulates at sites of myoblast fusion. ARF6 silencing inhibits the association of Trio and Rac1 with M-cadherin. Moreover, we demonstrate that ARF6 regulates myoblast fusion through Phospholipase D activation and PI(4,5)P2 production. Myoblast fusion is an essential step during myoblast differentiation that remains poorly understood. M-cadherin–dependent pathways that signal through Rac1 GTPase activation via the Rho-guanine nucleotide exchange factor (GEF) Trio are important for myoblast fusion. The ADP-ribosylation factor (ARF)6 GTPase has been shown to bind to Trio and to regulate Rac1 activity. Moreover, Loner/GEP100/BRAG2, a GEF of ARF6, has been involved in mammalian and Drosophila myoblast fusion, but the specific role of ARF6 has been not fully analyzed. Here, we show that ARF6 activity is increased at the time of myoblast fusion and is required for its implementation in mouse C2C12 myoblasts. Specifically, at the onset of myoblast fusion, ARF6 is associated with the multiproteic complex that contains M-cadherin, Trio, and Rac1 and accumulates at sites of myoblast fusion. ARF6 silencing inhibits the association of Trio and Rac1 with M-cadherin. Moreover, we demonstrate that ARF6 regulates myoblast fusion through phospholipase D (PLD) activation and phosphatidylinositol 4,5-bis-phosphate production. Together, these data indicate that ARF6 is a critical regulator of C2C12 myoblast fusion and participates in the regulation of PLD activities that trigger both phospholipids production and actin cytoskeleton reorganization at fusion sites.
Collapse
Affiliation(s)
- Anne-Sophie Bach
- Universités Montpellier 2 et 1, Centre de Recherche en Biochimie Macromoléculaire, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5237, Institut Fédératif de Recherche 122 1919 Route de Mende, 34293 Montpellier, France
| | | | | | | | | | | | | |
Collapse
|
46
|
Kim JE, Ahn MW, Baek SH, Lee IK, Kim YW, Kim JY, Dan JM, Park SY. AMPK activator, AICAR, inhibits palmitate-induced apoptosis in osteoblast. Bone 2008; 43:394-404. [PMID: 18502715 DOI: 10.1016/j.bone.2008.03.021] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 03/06/2008] [Accepted: 03/16/2008] [Indexed: 12/28/2022]
Abstract
Osteoblast apoptosis reduces bone mineral density. Apoptosis can be induced in a variety of cells by palmitate, which is one of the most common saturated fatty acids in dietary fat. The AMPK activator, AICAR, has been shown to inhibit palmitate-induced apoptosis. However, the role of palmitate in osteoblast apoptosis is currently unknown. This study examined whether palmitate could induce apoptosis in osteoblasts, and if so, whether AICAR could alleviate palmitate-induced apoptosis. Palmitate reduced cell survival and induced apoptosis in a dose- and time-dependent manner in human fetal osteoblasts (hFOB) 1.19. While the long-chain acyl-CoA synthetase inhibitor, triacsin C, inhibited palmitate-induced apoptosis, anti-oxidants and ceramide synthesis inhibitors did not attenuate the apoptosis. AICAR prevented palmitate-induced apoptosis and the inhibition of AICAR-mediated increase in fatty acid oxidation by etomoxir did not affect the prevention of apoptosis by AICAR. Constitutively-active AMPK also inhibited palmitate-induced apoptosis. Treatment with an AMPK inhibitor (compound C) and a dominant-negative AMPK adenovirus suppressed the inhibitory effect of AICAR on apoptosis. Palmitate impaired the activation of ERK by fetal bovine serum, which was blocked by AICAR. Moreover, AICAR increased ERK activation, and ERK inhibitors, PD98059 and U0126, as well as a dominant-negative MEK1, abolished the inhibitory effect of AICAR on palmitate-induced apoptosis. AICAR also inhibited palmitate-induced apoptosis in osteoblastic differentiated cells from human bone marrow, which was accompanied by recovered ERK activity. These results suggest that palmitate induces apoptosis in osteoblasts through the impaired activation of ERK, and the activation of AMPK inhibits palmitate-induced apoptosis by activating ERK.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, South Korea; Aging-associated Vascular Disease Research Center, Yeungnam University, Daegu, South Korea
| | - Myun-Whan Ahn
- Department of Orthopedics, College of Medicine, Yeungnam University, Daegu, South Korea
| | - Suk-Hwan Baek
- Aging-associated Vascular Disease Research Center, Yeungnam University, Daegu, South Korea; Department of Biochemistry and Molecular Biology, College of Medicine, Yeungnam University, Daegu, South Korea
| | - In Kyu Lee
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, South Korea
| | - Yong-Woon Kim
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, South Korea
| | - Jong-Yeon Kim
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, South Korea
| | - Jin-Myoung Dan
- Department of Orthopedics, College of Medicine, Pochun Cha University, Kumi, South Korea
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, South Korea; Aging-associated Vascular Disease Research Center, Yeungnam University, Daegu, South Korea.
| |
Collapse
|
47
|
Prasad VVTS, Nithipatikom K, Harder DR. Ceramide elevates 12-hydroxyeicosatetraenoic acid levels and upregulates 12-lipoxygenase in rat primary hippocampal cell cultures containing predominantly astrocytes. Neurochem Int 2008; 53:220-9. [PMID: 18680775 DOI: 10.1016/j.neuint.2008.07.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Revised: 06/01/2008] [Accepted: 07/01/2008] [Indexed: 10/21/2022]
Abstract
We report, exogenous addition of ceramide significantly increases 12-hydroxyeicosatetraenoic acid [12-(S)-HETE] levels, in a dose-dependent manner. 12-(S)-HETE levels, in 20, 30 and 40microM ceramide exposed rat primary hippocampal cell cultures containing predominantly astrocytes and few neurons and other glial cells (the cultured hippocampal cells were predominantly astrocytes amounting to over 99% of total cells with few neurons and other glial cells) amounted to 207, 260 and 408% of the controls, respectively. However, dihydroceramide, an inactive analog of ceramide did not alter the levels of 12-(S)-HETE. Ceramide also increased the mRNA and protein expression, and activity of 12-lipoxygease (12-LOX) needed for the synthesis of 12(S)-HETE. These results indicate a possible link between ceramide and 12-LOX pathway. However, ceramide did not alter expression of 5-lipoxygenase (5-LOX), another member of the lipoxygenase family. However, ceramide upregulated expression of cytosolic phospholipase-A(2) (cPLA(2)) and cyclooxygenase-2 (COX-2). Further, ceramide caused a significant increase in the levels of reactive oxygen species (ROS). Ceramide-mediated generation of ROS was inhibited by baicalien but not by indomethacin. In addition, ceramide treated cells exhibited increased mRNA expression of DNA damage induced transcript3 (Ddit3). This report which demonstrate induction of pro-carcinogenic 12-LOX pathway by an anticancer ceramide, may be relevant to cancer biologists studying drug resistant tumors and devising potent anticancer therapeutic strategies to treat drug resistant tumors. These results indicate possibility of 12-LOX involvement in ceramide-mediated generation of ROS and cellular oxidative stress. Induction of 12-LOX pathway by ceramide may have implications in understanding pathophysiology of neurodegenerative diseases involving ROS generation and inflammation.
Collapse
|
48
|
Sbrana F, Sassoli C, Meacci E, Nosi D, Squecco R, Paternostro F, Tiribilli B, Zecchi-Orlandini S, Francini F, Formigli L. Role for stress fiber contraction in surface tension development and stretch-activated channel regulation in C2C12 myoblasts. Am J Physiol Cell Physiol 2008; 295:C160-72. [DOI: 10.1152/ajpcell.00014.2008] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Membrane-cytoskeleton interaction regulates transmembrane currents through stretch-activated channels (SACs); however, the mechanisms involved have not been tested in living cells. We combined atomic force microscopy, confocal immunofluorescence, and patch-clamp analysis to show that stress fibers (SFs) in C2C12 myoblasts behave as cables that, tensed by myosin II motor, activate SACs by modifying the topography and the viscoelastic (Young's modulus and hysteresis) and electrical passive (membrane capacitance, Cm) properties of the cell surface. Stimulation with sphingosine 1-phosphate to elicit SF formation, the inhibition of Rho-dependent SF formation by Y-27632 and of myosin II-driven SF contraction by blebbistatin, showed that not SF polymerization alone but the generation of tensional forces by SF contraction were involved in the stiffness response of the cell surface. Notably, this event was associated with a significant reduction in the amplitude of the cytoskeleton-mediated corrugations in the cell surface topography, suggesting a contribution of SF contraction to plasma membrane stretching. Moreover, Cm, used as an index of cell surface area, showed a linear inverse relationship with cell stiffness, indicating participation of the actin cytoskeleton in plasma membrane remodeling and the ability of SF formation to cause internalization of plasma membrane patches to reduce Cm and increase membrane tension. SF contraction also increased hysteresis. Together, these data provide the first experimental evidence for a crucial role of SF contraction in SAC activation. The related changes in cell viscosity may prevent SAC from abnormal activation.
Collapse
|
49
|
Yoon MS, Chen J. PLD regulates myoblast differentiation through the mTOR-IGF2 pathway. J Cell Sci 2008; 121:282-9. [PMID: 18198186 DOI: 10.1242/jcs.022566] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
A mammalian target of rapamycin (mTOR) pathway is essential for the differentiation of cultured skeletal myoblasts in response to growth factor withdrawal. Previously, phospholipase D (PLD) has been found to play a role in cell growth regulation and mitogenic activation of mTOR signaling. However, a role for PLD in the autocrine regulation of myoblast differentiation is not known. Here we show that upon induction of differentiation in mouse C2C12 satellite cells, the expression of both PLD1 and PLD2 is upregulated. C2C12 differentiation is markedly inhibited by 1-butanol, an inhibitor of the PLD-catalyzed transphosphatidylation reaction, and also by the knockdown of PLD1, but not PLD2. Further investigation has revealed that PLD1 is unlikely to regulate myogenesis through modulation of the actin cytoskeleton as previously suggested. Instead, PLD1 positively regulates mTOR signaling leading to the production of IGF2, an autocrine factor instrumental for the initiation of satellite cell differentiation. Furthermore, exogenous IGF2 fully rescues the differentiation defect resulting from PLD1 knockdown. Hence, PLD1 is critically involved in skeletal myogenesis by regulating the mTOR-IGF2 pathway.
Collapse
Affiliation(s)
- Mee-Sup Yoon
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, 601 S. Goodwin Avenue B107, Urbana, IL 61801, USA
| | | |
Collapse
|
50
|
Editors' Picks. J Invest Dermatol 2007. [DOI: 10.1038/sj.jid.5700839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|