1
|
Mosoni L, Germond A, Coudy-Gandilhon C, Malige M, Claustre A, Delabrise C, Djelloul-Mazouz M, Delorme Y, Hermet J, Fafournoux P, Combaret L, Polge C, Maurin AC, Taillandier D. Knockout of the Muscle-Specific E3 Ligase MuRF1 Affects Liver Lipid Metabolism upon Dexamethasone Treatment in Mice. ACS OMEGA 2024; 9:45610-45623. [PMID: 39554453 PMCID: PMC11561631 DOI: 10.1021/acsomega.4c08501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/19/2024]
Abstract
In order to preserve muscle mass during catabolic states, investigators are actively searching for a specific inhibitor of MuRF1, the only known E3 ligase that can target muscle contractile proteins for their degradation. However, what would be the consequences of such inhibitors on other organs, both in the short and long term? Indeed, skeletal muscles can provide amino acids for liver gluconeogenesis, which is a crucial adaptation for maintaining glucose homeostasis upon elevated energy demands (e.g., during prolonged starvation). Comparing 3-month-old wild-type and MuRF1-KO mice, we measured tissue weights, liver glycogen, lipid and protein content, and liver biochemical composition using Fourier transform infrared (FTIR) spectrometry in control animals and in dexamethasone (Dex)-treated animals. Dex induces a catabolic situation with muscle atrophy and lipid deposits in the liver. In response to Dex treatment, liver glycogen, lipid, and protein content increased in wild type (WT) and MuRF1-KO mice. We found that MuRF1 deletion differentially affected organ weights, the liver of KO mice being hypertrophied upon Dex treatment when compared to WT mice. Upon Dex treatment, muscle mass was preserved in MuRF1-KO mice, and by contrast, liver lipid content increased more in these animals than in WT mice. PLS-DA analysis of FTIR showed that the levels of 13 markers were significantly altered in KO vs WT mice, witnessing profound alterations of lipid, protein, and glycogen content in the liver due to the absence of MuRF1. Using Nile red and oil red lipid staining, we also found that both membrane-linked lipids and intracellular lipid droplets were altered due to the absence of MuRF1. Altogether, it seems that when the liver is deprived of the possibility of obtaining amino acids from muscle upon Dex treatment, there is a concomitant increase in tissue weight and anabolic activity.
Collapse
Affiliation(s)
- Laurent Mosoni
- Université
Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Arno Germond
- UR370,
QuaPA, Qualité des Produits Animaux, INRAE, F-63000 Clermont-Ferrand, France
| | - Cécile Coudy-Gandilhon
- Université
Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Mélodie Malige
- Université
Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Agnès Claustre
- Université
Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Coralie Delabrise
- Université
Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Mehdi Djelloul-Mazouz
- Université
Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Yoann Delorme
- Université
Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Julien Hermet
- Université
Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Pierre Fafournoux
- Université
Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Lydie Combaret
- Université
Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Cécile Polge
- Université
Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Anne-Catherine Maurin
- Université
Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Daniel Taillandier
- Université
Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| |
Collapse
|
2
|
Lu F, Zhang S, Dong S, Wang M, Pang K, Zhao Y, Huang J, Kang J, Liu N, Zhang X, Zhao D, Lu F, Zhang W. Exogenous hydrogen sulfide enhances myogenic differentiation of C2C12 myoblasts under high palmitate stress. Heliyon 2024; 10:e38661. [PMID: 39416846 PMCID: PMC11481675 DOI: 10.1016/j.heliyon.2024.e38661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
Skeletal muscle atrophy was one of main complications of type 2 diabetes mellitus. Hydrogen sulfide (H2S) is involved in various physiological functions, such as anti-hypertension and anti-oxidant. Skeletal muscle atrophy caused by type 2 diabetes could lead to the regeneration of muscle fibers. Wnt signaling pathway plays a crucial important role in this process. H2S maybe regulate the Wnt signaling pathway to alleviate skeletal muscle atrophy, however, this role has not been clarified. The aim of this study is to investigate the potential regulatory role of H2S in the Wnt signaling pathway. C2C12 myoblasts treated with 500 μmol palmitate as an in vitro model. Western blot was used to detect the levels of CSE, PKM1, β-catenin, MuRF1, MYOG, MYF6 and MYOD1. In addition, MuRF1 was mutated at Cys44 and MuRF1 S-sulfhydration was detected by biotin switch assay. The interaction between PKM1 and MuRF1 was assessed via Co-immunoprecipitation. Differentiation of C2C12 myoblasts was evaluated using LAMININ staining. These data showed the levels of CSE, β-catenin, PKM1, MYOG, MYF6 and MYOD1 were decreased in pal group, compared with control and pal + NaHS groups. MuRF1 Cys44 mutants increased the protein levels of β-catenin, MYOG, MYF6 and MYOD1 in pal group. Our results suggest that H2S regulates the S-sulfhydration levels of MuRF1 at Cys44, influencing the ubiquitination levels of PKM1 and ultimately promoting myoblast differentiation.
Collapse
Affiliation(s)
- Fangping Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
- Department of Pathophysiology, Mudanjiang Medical University, Mudanjiang, China
| | - Shiwu Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shiyun Dong
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Mengyi Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Kemiao Pang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yajun Zhao
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Jiayi Huang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Jiaxin Kang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Ning Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Xueya Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Dechao Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fanghao Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| |
Collapse
|
3
|
Uenaka E, Ojima K, Suzuki T, Kobayashi K, Muroya S, Nishimura T. Murf1 alters myosin replacement rates in cultured myotubes in a myosin isoform-dependent manner. In Vitro Cell Dev Biol Anim 2024; 60:748-759. [PMID: 38758432 DOI: 10.1007/s11626-024-00916-0] [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: 01/25/2024] [Accepted: 04/23/2024] [Indexed: 05/18/2024]
Abstract
Skeletal muscle tissue increases or decreases its volume by synthesizing or degrading myofibrillar proteins. The ubiquitin-proteasome system plays a pivotal role during muscle atrophy, where muscle ring finger proteins (Murf) function as E3 ubiquitin ligases responsible for identifying and targeting substrates for degradation. Our previous study demonstrated that overexpression of Ozz, an E3 specific to embryonic myosin heavy chain (Myh3), precisely reduced the Myh3 replacement rate in the thick filaments of myotubes (E. Ichimura et al., Physiol Rep. 9:e15003, 2021). These findings strongly suggest that E3 plays a critical role in regulating myosin replacement. Here, we hypothesized that the Murf isoforms, which recognize Myhs as substrates, reduced the myosin replacement rates through the enhanced Myh degradation by Murfs. First, fluorescence recovery after a photobleaching experiment was conducted to assess whether Murf isoforms affected the GFP-Myh3 replacement. In contrast to Murf2 or Murf3 overexpression, Murf1 overexpression selectively facilitated the GFP-Myh3 myosin replacement. Next, to examine the effects of Murf1 overexpression on the replacement of myosin isoforms, Cherry-Murf1 was coexpressed with GFP-Myh1, GFP-Myh4, or GFP-Myh7 in myotubes. Intriguingly, Murf1 overexpression enhanced the myosin replacement of GFP-Myh4 but did not affect those of GFP-Myh1 or GFP-Myh7. Surprisingly, overexpression of Murf1 did not enhance the ubiquitination of proteins. These results indicate that Murf1 selectively regulated myosin replacement in a Myh isoform-dependent fashion, independent of enhanced ubiquitination. This suggests that Murf1 may have a role beyond functioning as a ubiquitin ligase E3 in thick filament myosin replacement.
Collapse
Affiliation(s)
- Emi Uenaka
- Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, 9 Kita, 9 Nishi, Sapporo, Hokkaido, 060-8589, Japan
- Space Environment and Energy Laboratories, Nippon Telegraph and Telephone Corporation, Musashino, Tokyo, 180-8585, Japan
| | - Koichi Ojima
- Muscle Biology Research Unit, Division of Animal Products Research, Institute of Livestock and Grassland Science, NARO, 2 Ikenodai, Tsukuba, Ibaraki, 305-0901, Japan
| | - Takahiro Suzuki
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Motooka 744, Nishi-Ku, Fukuoka, 819-0395, Japan
| | - Ken Kobayashi
- Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, 9 Kita, 9 Nishi, Sapporo, Hokkaido, 060-8589, Japan
| | - Susumu Muroya
- Muscle Biology Research Unit, Division of Animal Products Research, Institute of Livestock and Grassland Science, NARO, 2 Ikenodai, Tsukuba, Ibaraki, 305-0901, Japan
- Laboratory of Meat Science and Production, Faculty of Veterinary Medicine, Kagoshima University, 1-21-24, Korimoto, Kagoshima, 890-0065, Japan
| | - Takanori Nishimura
- Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, 9 Kita, 9 Nishi, Sapporo, Hokkaido, 060-8589, Japan.
| |
Collapse
|
4
|
Chen J, Feng X, Zhou X, Li Y. Role of the tripartite motif-containing (TRIM) family of proteins in insulin resistance and related disorders. Diabetes Obes Metab 2024; 26:3-15. [PMID: 37726973 DOI: 10.1111/dom.15294] [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: 06/13/2023] [Revised: 08/27/2023] [Accepted: 09/05/2023] [Indexed: 09/21/2023]
Abstract
Emerging evidence suggests that the ubiquitin-mediated degradation of insulin-signalling-related proteins may be involved in the development of insulin resistance and its related disorders. Tripartite motif-containing (TRIM) proteins, a superfamily belonging to the E3 ubiquitin ligases, are capable of controlling protein levels and function by ubiquitination, which is essential for the modulation of insulin sensitivity. Recent research has indicated that some of these TRIMs act as key regulatory factors of metabolic disorders such as type 2 diabetes mellitus, obesity, nonalcoholic fatty liver disease, and atherosclerosis. This review provides a comprehensive overview of the latest evidence linking TRIMs to the regulation of insulin resistance and its related disorders, their roles in regulating multiple signalling pathways or cellular processes, such as insulin signalling pathways, peroxisome proliferator-activated receptor signalling pathways, glucose and lipid metabolism, the inflammatory response, and cell cycle control, as well as recent advances in the development of TRIM-targeted drugs.
Collapse
Affiliation(s)
- Jianrong Chen
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Clinical Research Centre for Endocrine and Metabolic disease, Nanchang, China
- Jiangxi Branch of National Clinical Research Centre for Metabolic disease, Nanchang, China
| | - Xianjie Feng
- Evidence-based Medicine Research Centre, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Xu Zhou
- Evidence-based Medicine Research Centre, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yong Li
- Department of Anaesthesiology, Medical Centre of Anaesthesiology and Pain, First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
5
|
Li J, Hu Y, Li J, Wang H, Wu H, Zhao C, Tan T, Zhang L, Zhu D, Liu X, Li N, Hu X. Loss of MuRF1 in Duroc pigs promotes skeletal muscle hypertrophy. Transgenic Res 2023; 32:153-167. [PMID: 37071377 DOI: 10.1007/s11248-023-00342-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/03/2023] [Indexed: 04/19/2023]
Abstract
Muscle mass development depends on increased protein synthesis and reduced muscle protein degradation. Muscle ring-finger protein-1 (MuRF1) plays a key role in controlling muscle atrophy. Its E3 ubiquitin ligase activity recognizes and degrades skeletal muscle proteins through the ubiquitin-proteasome system. The loss of Murf1, which encodes MuRF1, in mice leads to the accumulation of skeletal muscle proteins and alleviation of muscle atrophy. However, the function of Murf1 in agricultural animals remains unclear. Herein, we bred F1 generation Murf1+/- and F2 generation Murf1-/- Duroc pigs from F0 Murf1-/- pigs to investigate the effect of Murf1 knockout on skeletal muscle development. We found that the Murf1+/- pigs retained normal levels of muscle growth and reproduction, and their percentage of lean meat increased by 6% compared to that of the wild type (WT) pigs. Furthermore, the meat color, pH, water-holding capacity, and tenderness of the Murf1+/- pigs were similar to those of the WT pigs. The drip loss rate and intramuscular fat decreased slightly in the Murf1+/- pigs. However, the cross-sectional area of the myofibers in the longissimus dorsi increased in the adult Murf1+/- pigs. The skeletal muscle proteins MYBPC3 and actin, which are targeted by MuRF1, accumulated in the Murf1+/- and Murf1-/- pigs. Our findings show that inhibiting muscle protein degradation in MuRF1-deficient Duroc pigs increases the size of their myofibers and their percentage of lean meat without influencing their growth or pork quality. Our study demonstrates that Murf1 is a target gene for promoting skeletal muscle hypertrophy in pig breeding.
Collapse
Affiliation(s)
- Jiaping Li
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Yiqing Hu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
- National Center for Cardiovascular Diseases, Beijing, People's Republic of China
- National Institute of Biological Sciences, Beijing, People's Republic of China
| | - Jiajia Li
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Haitao Wang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Hanyu Wu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Chengcheng Zhao
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Tan Tan
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
- Development Center of Science and Technology, Ministry of Agriculture and Rural Affairs, Beijing, People's Republic of China
| | - Li Zhang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, 225300, China
| | - Di Zhu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Xu Liu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Ning Li
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China.
| | - Xiaoxiang Hu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China.
| |
Collapse
|
6
|
Kincheloe GN, Roberson PA, Toro AL, Stanley BA, Stanley AE, Jefferson LS, Dennis MD, Kimball SR. Loss of 4E-BPs prevents the hindlimb immobilization-induced decrease in protein synthesis in skeletal muscle. J Appl Physiol (1985) 2023; 134:72-83. [PMID: 36454675 PMCID: PMC9799152 DOI: 10.1152/japplphysiol.00563.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/30/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022] Open
Abstract
The present study was designed to test the hypothesis that upregulating protein synthesis attenuates the loss of muscle mass in a model of disuse atrophy. The studies compared the effect of unilateral hindlimb immobilization in wild-type (WT) mice and double-knockout (DKO) mice lacking the translational regulators 4E-BP1 and 4E-BP2. Immobilization-induced downregulation of protein synthesis occurred in both groups of mice, but protein synthesis was higher in gastrocnemius muscle from the immobilized hindlimb of fasted DKO compared with WT mice. Surprisingly, although protein synthesis was partially elevated in DKO compared with WT mice, atrophy occurred to the same extent in both groups of animals. This may be partially due to impaired leucine-induced stimulation of protein synthesis in DKO compared with WT mice due to downregulated eukaryotic initiation factor eIF4E expression in muscle of DKO compared with WT mice. Expression of the E3 ubiquitin ligases MAFbx and MuRF-1 mRNAs and total protein ubiquitylation was upregulated in the immobilized compared with the nonimmobilized hindlimb of both WT and DKO mice, with little difference in the magnitude of the upregulation between genotypes. Analysis of newly synthesized proteins revealed downregulation of several glycolytic enzymes in the gastrocnemius of DKO mice compared with WT mice, as well as in the immobilized compared with the nonimmobilized hindlimb. Overall, the results suggest that the elevated rate of protein synthesis during hindlimb immobilization in fasted DKO mice is insufficient to prevent disuse-induced muscle atrophy, probably due to induction of compensatory mechanisms including downregulation of eIF4E expression.NEW & NOTEWORTHY Basal rates of protein synthesis are elevated in skeletal muscle in the immobilized leg of mice lacking the translational repressors, 4E-BP1 and 4E-BP2 (knockout mice), compared with wild-type mice. However, disuse-induced muscle atrophy occurs to the same extent in both wild-type and knockout mice suggesting that compensatory mechanisms are induced that overcome the upregulation of muscle protein synthesis. Proteomic analysis revealed that mRNAs encoding several glycolytic enzymes are differentially translated in wild-type and knockout mice.
Collapse
Affiliation(s)
- Gregory N Kincheloe
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Paul A Roberson
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Allyson L Toro
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Bruce A Stanley
- Mass Spectrometry & Proteomics Core, Penn State College of Medicine, Hershey, Pennsylvania
| | - Anne E Stanley
- Mass Spectrometry & Proteomics Core, Penn State College of Medicine, Hershey, Pennsylvania
| | - Leonard S Jefferson
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Michael D Dennis
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
7
|
Skeletal muscle mitochondrial remodeling in heart failure: An update on mechanisms and therapeutic opportunities. Biomed Pharmacother 2022; 155:113833. [DOI: 10.1016/j.biopha.2022.113833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022] Open
|
8
|
Sugiura K, Hirasaka K, Maeda T, Uchida T, Kishimoto K, Oarada M, Labeit S, Ulla A, Sakakibara I, Nakao R, Sairyo K, Nikawa T. MuRF1 deficiency prevents age-related fat weight gain, possibly through accumulation of PDK4 in skeletal muscle mitochondria in older mice. J Orthop Res 2022; 40:1026-1038. [PMID: 34185335 DOI: 10.1002/jor.25131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 06/09/2021] [Accepted: 06/24/2021] [Indexed: 02/04/2023]
Abstract
Recent studies show that muscle mass and metabolic function are interlinked. Muscle RING finger 1 (MuRF1) is a critical muscle-specific ubiquitin ligase associated with muscle atrophy. Yet, the molecular target of MuRF1 in atrophy and aging remains unclear. We examined the role of MuRF1 in aging, using MuRF1-deficient (MuRF1-/- ) mice in vivo, and MuRF1-overexpressing cell in vitro. MuRF1 deficiency partially prevents age-induced skeletal muscle loss in mice. Interestingly, body weight and fat mass of more than 7-month-old MuRF1-/- mice were lower than in MuRF1+/+ mice. Serum and muscle metabolic parameters and results of indirect calorimetry suggest significantly higher energy expenditure and enhanced lipid metabolism in 3-month-old MuRF1-/- mice than in MuRF1+/+ mice, resulting in suppressed adipose tissue gain during aging. Pyruvate dehydrogenase kinase 4 (PDK4) is crucial for a switch from glucose to lipid metabolism, and the interaction between MuRF1 and PDK4 was examined. PDK4 protein levels were elevated in mitochondria from the skeletal muscle in MuRF1-/- mice. In vitro, MuRF1 interacted with PDK4 but did not induce degradation through ubiquitination. Instead, SUMO posttranscriptional modification (SUMOylation) of PDK4 was detected in MuRF1-overexpressing cells, in contrast to cells without the RING domain of MuRF1. MuRF1 deficiency enhances lipid metabolism possibly by upregulating PDK4 localization into mitochondrial through prevention of SUMOylation. Inhibition of MuRF1-mediated PDK4 SUMOylation is a potential therapeutic target for age-related dysfunction of lipid metabolism and muscle atrophy.
Collapse
Affiliation(s)
- Kosuke Sugiura
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan.,Department of Orthopedics, Institute of Medical Biosciences, Tokushima University Graduate School, Tokushima, Japan
| | - Katsuya Hirasaka
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan.,Division of Marine Energy Utilization, Organization for Marine Science and Technology, Nagasaki University, Nagasaki, Japan
| | - Tasuku Maeda
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Takayuki Uchida
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Koji Kishimoto
- Department of Bioscience and Bioindustry, Tokushima University Graduate School, Tokushima, Japan
| | - Motoko Oarada
- Department of Nutrition Health, Faculty of Nutritional Science Sagami Women's University, Sagamihara, Kanagawa, Japan
| | - Siegfried Labeit
- Department of Integrative Pathophysiology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Anayt Ulla
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Iori Sakakibara
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Reiko Nakao
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Koichi Sairyo
- Department of Orthopedics, Institute of Medical Biosciences, Tokushima University Graduate School, Tokushima, Japan
| | - Takeshi Nikawa
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
9
|
Balakrishnan R, Thurmond DC. Mechanisms by Which Skeletal Muscle Myokines Ameliorate Insulin Resistance. Int J Mol Sci 2022; 23:4636. [PMID: 35563026 PMCID: PMC9102915 DOI: 10.3390/ijms23094636] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 12/17/2022] Open
Abstract
The skeletal muscle is the largest organ in the body and secretes circulating factors, including myokines, which are involved in various cellular signaling processes. Skeletal muscle is vital for metabolism and physiology and plays a crucial role in insulin-mediated glucose disposal. Myokines have autocrine, paracrine, and endocrine functions, serving as critical regulators of myogenic differentiation, fiber-type switching, and maintaining muscle mass. Myokines have profound effects on energy metabolism and inflammation, contributing to the pathophysiology of type 2 diabetes (T2D) and other metabolic diseases. Myokines have been shown to increase insulin sensitivity, thereby improving glucose disposal and regulating glucose and lipid metabolism. Many myokines have now been identified, and research on myokine signaling mechanisms and functions is rapidly emerging. This review summarizes the current state of the field regarding the role of myokines in tissue cross-talk, including their molecular mechanisms, and their potential as therapeutic targets for T2D.
Collapse
Affiliation(s)
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA;
| |
Collapse
|
10
|
Zhu S, Gu H, Peng C, Xia F, Cao H, Cui H. Regulation of Glucose, Fatty Acid and Amino Acid Metabolism by Ubiquitination and SUMOylation for Cancer Progression. Front Cell Dev Biol 2022; 10:849625. [PMID: 35392171 PMCID: PMC8981989 DOI: 10.3389/fcell.2022.849625] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/01/2022] [Indexed: 12/19/2022] Open
Abstract
Ubiquitination and SUMOylation, which are posttranslational modifications, play prominent roles in regulating both protein expression and function in cells, as well as various cellular signal transduction pathways. Metabolic reprogramming often occurs in various diseases, especially cancer, which has become a new entry point for understanding cancer mechanisms and developing treatment methods. Ubiquitination or SUMOylation of protein substrates determines the fate of modified proteins. Through accurate and timely degradation and stabilization of the substrate, ubiquitination and SUMOylation widely control various crucial pathways and different proteins involved in cancer metabolic reprogramming. An understanding of the regulatory mechanisms of ubiquitination and SUMOylation of cell proteins may help us elucidate the molecular mechanism underlying cancer development and provide an important theory for new treatments. In this review, we summarize the processes of ubiquitination and SUMOylation and discuss how ubiquitination and SUMOylation affect cancer metabolism by regulating the key enzymes in the metabolic pathway, including glucose, lipid and amino acid metabolism, to finally reshape cancer metabolism.
Collapse
Affiliation(s)
- Shunqin Zhu
- State Key Laboratory of Silkworm Genome Biology, School of Life Sciences, Southwest University, Chongqing, China
- Cancer Center, Reproductive Medicine Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Hongyu Gu
- State Key Laboratory of Silkworm Genome Biology, School of Life Sciences, Southwest University, Chongqing, China
- Cancer Center, Reproductive Medicine Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Cheng Peng
- State Key Laboratory of Silkworm Genome Biology, School of Life Sciences, Southwest University, Chongqing, China
- Cancer Center, Reproductive Medicine Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Fanwei Xia
- State Key Laboratory of Silkworm Genome Biology, School of Life Sciences, Southwest University, Chongqing, China
| | - Huan Cao
- State Key Laboratory of Silkworm Genome Biology, School of Life Sciences, Southwest University, Chongqing, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, School of Life Sciences, Southwest University, Chongqing, China
- Cancer Center, Reproductive Medicine Center, Medical Research Institute, Southwest University, Chongqing, China
- *Correspondence: Hongjuan Cui,
| |
Collapse
|
11
|
Goodman CA, Parker BL, Gregorevic P. Expanding the MuRF1 Universe with Quantitative Ubiquitylomics. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab058. [PMID: 35330789 PMCID: PMC8788820 DOI: 10.1093/function/zqab058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/02/2021] [Indexed: 01/07/2023]
Affiliation(s)
| | - Benjamin L Parker
- Centre for Muscle Research (CMR), Department of Physiology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Paul Gregorevic
- Centre for Muscle Research (CMR), Department of Physiology, The University of Melbourne, Parkville, Victoria 3010, Australia,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3168, Australia,Department of Neurology, The University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
12
|
Neurohr JM, Paulson ET, Kinsey ST. A higher mitochondrial content is associated with greater oxidative damage, oxidative defenses, protein synthesis and ATP turnover in resting skeletal muscle. J Exp Biol 2021; 224:jeb242462. [PMID: 34581401 PMCID: PMC8541733 DOI: 10.1242/jeb.242462] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 09/15/2021] [Indexed: 01/13/2023]
Abstract
An unavoidable consequence of aerobic metabolism is the production of reactive oxygen species (ROS). Mitochondria have historically been considered the primary source of ROS; however, recent literature has highlighted the uncertainty in primary ROS production sites and it is unclear how variation in mitochondrial density influences ROS-induced damage and protein turnover. Fish skeletal muscle is composed of distinct, highly aerobic red muscle and anaerobic white muscle, offering an excellent model system in which to evaluate the relationship of tissue aerobic capacity and ROS-induced damage under baseline conditions. The present study used a suite of indices to better understand potential consequences of aerobic tissue capacity in red and white muscle of the pinfish, Lagodon rhomboides. Red muscle had a 7-fold greater mitochondrial volume density than white muscle, and more oxidative damage despite also having higher activity of the antioxidant enzymes superoxide dismutase and catalase. The dominant protein degradation system appears to be tissue dependent. Lysosomal degradation markers and autophagosome volume density were greater in white muscle, while ubiquitin expression and 20S proteasome activity were significantly greater in red muscle. However, ubiquitin ligase expression was significantly higher in white muscle. Red muscle had a more than 2-fold greater rate of translation and total ATP turnover than white muscle, results that may be due in part to the higher mitochondrial density and the associated increase in oxidative damage. Together, these results support the concept that an elevated aerobic capacity is associated with greater oxidative damage and higher costs of protein turnover.
Collapse
Affiliation(s)
| | | | - Stephen T. Kinsey
- University of North Carolina Wilmington, Department of Biology and Marine Biology, 601 S. College Road, Wilmington, NC 28403, USA
| |
Collapse
|
13
|
Haberecht-Müller S, Krüger E, Fielitz J. Out of Control: The Role of the Ubiquitin Proteasome System in Skeletal Muscle during Inflammation. Biomolecules 2021; 11:biom11091327. [PMID: 34572540 PMCID: PMC8468834 DOI: 10.3390/biom11091327] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
The majority of critically ill intensive care unit (ICU) patients with severe sepsis develop ICU-acquired weakness (ICUAW) characterized by loss of muscle mass, reduction in myofiber size and decreased muscle strength leading to persisting physical impairment. This phenotype results from a dysregulated protein homeostasis with increased protein degradation and decreased protein synthesis, eventually causing a decrease in muscle structural proteins. The ubiquitin proteasome system (UPS) is the predominant protein-degrading system in muscle that is activated during diverse muscle atrophy conditions, e.g., inflammation. The specificity of UPS-mediated protein degradation is assured by E3 ubiquitin ligases, such as atrogin-1 and MuRF1, which target structural and contractile proteins, proteins involved in energy metabolism and transcription factors for UPS-dependent degradation. Although the regulation of activity and function of E3 ubiquitin ligases in inflammation-induced muscle atrophy is well perceived, the contribution of the proteasome to muscle atrophy during inflammation is still elusive. During inflammation, a shift from standard- to immunoproteasome was described; however, to which extent this contributes to muscle wasting and whether this changes targeting of specific muscular proteins is not well described. This review summarizes the function of the main proinflammatory cytokines and acute phase response proteins and their signaling pathways in inflammation-induced muscle atrophy with a focus on UPS-mediated protein degradation in muscle during sepsis. The regulation and target-specificity of the main E3 ubiquitin ligases in muscle atrophy and their mode of action on myofibrillar proteins will be reported. The function of the standard- and immunoproteasome in inflammation-induced muscle atrophy will be described and the effects of proteasome-inhibitors as treatment strategies will be discussed.
Collapse
Affiliation(s)
- Stefanie Haberecht-Müller
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Elke Krüger
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17475 Greifswald, Germany;
- Correspondence: (E.K.); (J.F.)
| | - Jens Fielitz
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, 17475 Greifswald, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, 17475 Greifswald, Germany
- Correspondence: (E.K.); (J.F.)
| |
Collapse
|
14
|
Baehr LM, Hughes DC, Lynch SA, Van Haver D, Maia TM, Marshall AG, Radoshevich L, Impens F, Waddell DS, Bodine SC. Identification of the MuRF1 Skeletal Muscle Ubiquitylome Through Quantitative Proteomics. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab029. [PMID: 34179788 PMCID: PMC8218097 DOI: 10.1093/function/zqab029] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023]
Abstract
MuRF1 (TRIM63) is a muscle-specific E3 ubiquitin ligase and component of the ubiquitin proteasome system. MuRF1 is transcriptionally upregulated under conditions that cause muscle loss, in both rodents and humans, and is a recognized marker of muscle atrophy. In this study, we used in vivo electroporation to determine whether MuRF1 overexpression alone can cause muscle atrophy and, in combination with ubiquitin proteomics, identify the endogenous MuRF1 substrates in skeletal muscle. Overexpression of MuRF1 in adult mice increases ubiquitination of myofibrillar and sarcoplasmic proteins, increases expression of genes associated with neuromuscular junction instability, and causes muscle atrophy. A total of 169 ubiquitination sites on 56 proteins were found to be regulated by MuRF1. MuRF1-mediated ubiquitination targeted both thick and thin filament contractile proteins, as well as, glycolytic enzymes, deubiquitinases, p62, and VCP. These data reveal a potential role for MuRF1 in not only the breakdown of the sarcomere but also the regulation of metabolism and other proteolytic pathways in skeletal muscle.
Collapse
Affiliation(s)
| | | | - Sarah A Lynch
- Department of Biology, University of North Florida, 1 UNF Drive, Jacksonville, FL 32224, USA
| | - Delphi Van Haver
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium,VIB Center for Medical Biotechnology, Ghent, Belgium,VIB Proteomics Core, Ghent, Belgium
| | - Teresa Mendes Maia
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium,VIB Center for Medical Biotechnology, Ghent, Belgium,VIB Proteomics Core, Ghent, Belgium
| | - Andrea G Marshall
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Lilliana Radoshevich
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Francis Impens
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium,VIB Center for Medical Biotechnology, Ghent, Belgium,VIB Proteomics Core, Ghent, Belgium
| | - David S Waddell
- Department of Biology, University of North Florida, 1 UNF Drive, Jacksonville, FL 32224, USA
| | | |
Collapse
|
15
|
The emerging role of the sympathetic nervous system in skeletal muscle motor innervation and sarcopenia. Ageing Res Rev 2021; 67:101305. [PMID: 33610815 DOI: 10.1016/j.arr.2021.101305] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/06/2021] [Accepted: 02/15/2021] [Indexed: 12/30/2022]
Abstract
Examining neural etiologic factors'role in the decline of neuromuscular function with aging is essential to our understanding of the mechanisms underlying sarcopenia, the age-dependent decline in muscle mass, force and power. Innervation of the skeletal muscle by both motor and sympathetic axons has been established, igniting interest in determining how the sympathetic nervous system (SNS) affect skeletal muscle composition and function throughout the lifetime. Selective expression of the heart and neural crest derivative 2 gene in peripheral SNs increases muscle mass and force regulating skeletal muscle sympathetic and motor innervation; improving acetylcholine receptor stability and NMJ transmission; preventing inflammation and myofibrillar protein degradation; increasing autophagy; and probably enhancing protein synthesis. Elucidating the role of central SNs will help to define the coordinated response of the visceral and neuromuscular system to physiological and pathological challenges across ages. This review discusses the following questions: (1) Does the SNS regulate skeletal muscle motor innervation? (2) Does the SNS regulate presynaptic and postsynaptic neuromuscular junction (NMJ) structure and function? (3) Does sympathetic neuron (SN) regulation of NMJ transmission decline with aging? (4) Does maintenance of SNs attenuate aging sarcopenia? and (5) Do central SN group relays influence sympathetic and motor muscle innervation?
Collapse
|
16
|
Labeit S, Hirner S, Bogomolovas J, Cruz A, Myrzabekova M, Moriscot A, Bowen TS, Adams V. Regulation of Glucose Metabolism by MuRF1 and Treatment of Myopathy in Diabetic Mice with Small Molecules Targeting MuRF1. Int J Mol Sci 2021; 22:2225. [PMID: 33672385 PMCID: PMC7926706 DOI: 10.3390/ijms22042225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 01/07/2023] Open
Abstract
The muscle-specific ubiquitin ligase MuRF1 regulates muscle catabolism during chronic wasting states, although its roles in general metabolism are less-studied. Here, we metabolically profiled MuRF1-deficient knockout mice. We also included knockout mice for MuRF2 as its closely related gene homolog. MuRF1 and MuRF2-KO (knockout) mice have elevated serum glucose, elevated triglycerides, and reduced glucose tolerance. In addition, MuRF2-KO mice have a reduced tolerance to a fat-rich diet. Western blot and enzymatic studies on MuRF1-KO skeletal muscle showed perturbed FoxO-Akt signaling, elevated Akt-Ser-473 activation, and downregulated oxidative mitochondrial metabolism, indicating potential mechanisms for MuRF1,2-dependent glucose and fat metabolism regulation. Consistent with this, the adenoviral re-expression of MuRF1 in KO mice normalized Akt-Ser-473, serum glucose, and triglycerides. Finally, we tested the MuRF1/2 inhibitors MyoMed-205 and MyoMed-946 in a mouse model for type 2 diabetes mellitus (T2DM). After 28 days of treatment, T2DM mice developed progressive muscle weakness detected by wire hang tests, but this was attenuated by the MyoMed-205 treatment. While MyoMed-205 and MyoMed-946 had no significant effects on serum glucose, they did normalize the lymphocyte-granulocyte counts in diabetic sera as indicators of the immune response. Thus, small molecules directed to MuRF1 may be useful in attenuating skeletal muscle strength loss in T2DM conditions.
Collapse
Affiliation(s)
- Siegfried Labeit
- Department of Anesthesiology, Medical Faculty Mannheim, University of Heidelberg, 68169 Mannheim, Germany;
- Myomedix GmbH, 69151 Neckargemünd, Germany
| | - Stephanie Hirner
- Department of Anesthesiology, Medical Faculty Mannheim, University of Heidelberg, 68169 Mannheim, Germany;
| | | | - André Cruz
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, 05508-000 Sao Paulo, Brazil; (A.C.); (A.M.)
| | - Moldir Myrzabekova
- Scientific Research Institute of Biology and Biotechnology Problems, al-Farabi Kasakh National University, Almaty 050040, Kazakhstan;
| | - Anselmo Moriscot
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, 05508-000 Sao Paulo, Brazil; (A.C.); (A.M.)
| | | | - Volker Adams
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany;
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, 01307 Dresden, Germany
| |
Collapse
|
17
|
Li C, Wu Q, Li Z, Wang Z, Tu Y, Chen C, Sun S, Sun S. Exosomal microRNAs in cancer-related sarcopenia: Tumor-derived exosomal microRNAs in muscle atrophy. Exp Biol Med (Maywood) 2021; 246:1156-1166. [PMID: 33554647 DOI: 10.1177/1535370221990322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cancer-associated sarcopenia is a complex metabolic syndrome marked by muscle mass wasting. Muscle wasting is a serious complication that is a primary contributor to cancer-related mortality. The underlying molecular mechanisms of cancer-associated sarcopenia have not been completely described to date. In general, evidence shows that the main pathophysiological alterations in sarcopenia are associated with the degradation of cellular components, an exceptional inflammatory secretome and mitochondrial dysfunction. Importantly, we highlight the prospect that several miRNAs carried by tumor-derived exosomes that have shown the ability to promote inflammatory secretion, activate catabolism, and even participate in the regulation of cellular degradation pathways can be delivered to and exert effects on muscle cells. In this review, we aim to describe the current knowledge about the functions of exosomal miRNAs in the induction of cancer-associated muscle wasting and propose potential treatment strategies.
Collapse
Affiliation(s)
- Chenyuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | - Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | - Zhiyu Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | - Zhong Wang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | - Yi Tu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| |
Collapse
|
18
|
Pablo Tortola C, Fielitz B, Li Y, Rüdebusch J, Luft FC, Fielitz J. Activation of Tripartite Motif Containing 63 Expression by Transcription Factor EB and Transcription Factor Binding to Immunoglobulin Heavy Chain Enhancer 3 Is Regulated by Protein Kinase D and Class IIa Histone Deacetylases. Front Physiol 2021; 11:550506. [PMID: 33519497 PMCID: PMC7838639 DOI: 10.3389/fphys.2020.550506] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 12/09/2020] [Indexed: 01/07/2023] Open
Abstract
Rationale The ubiquitin–proteasome system (UPS) is responsible for skeletal muscle atrophy. We showed earlier that the transcription factor EB (TFEB) plays a role by increasing E3 ubiquitin ligase muscle really interesting new gene-finger 1(MuRF1)/tripartite motif-containing 63 (TRIM63) expression. MuRF 1 ubiquitinates structural proteins and mediates their UPS-dependent degradation. We now investigated how TFEB-mediated TRIM63 expression is regulated. Objective Because protein kinase D1 (PKD1), histone deacetylase 5 (HDAC5), and TFEB belong to respective families with close structural, regulatory, and functional properties, we hypothesized that these families comprise a network regulating TRIM63 expression. Methods and Results We found that TFEB and transcription factor for immunoglobulin heavy-chain enhancer 3 (TFE3) activate TRIM63 expression. The class IIa HDACs HDAC4, HDAC5, and HDAC7 inhibited this activity. Furthermore, we could map the HDAC5 and TFE3 physical interaction. PKD1, PKD2, and PKD3 reversed the inhibitory effect of all tested class IIa HDACs toward TFEB and TFE3. PKD1 mediated nuclear export of all HDACs and lifted TFEB and TFE3 repression. We also mapped the PKD2 and HDAC5 interaction. We found that the inhibitory effect of PKD1 and PKD2 toward HDAC4, HDAC5, and HDAC7 was mediated by their phosphorylation and 14-3-3 mediated nuclear export. Conclusion TFEB and TFE3 activate TRIM63 expression. Both transcription factors are controlled by HDAC4, HDAC5, HDAC7, and all PKD-family members. We propose that the multilevel PKD/HDAC/TFEB/TFE3 network tightly controls TRIM63 expression.
Collapse
Affiliation(s)
- Cristina Pablo Tortola
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Britta Fielitz
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Yi Li
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Rüdebusch
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jens Fielitz
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| |
Collapse
|
19
|
Goodman CA, Davey JR, Hagg A, Parker BL, Gregorevic P. Dynamic Changes to the Skeletal Muscle Proteome and Ubiquitinome Induced by the E3 Ligase, ASB2β. Mol Cell Proteomics 2021; 20:100050. [PMID: 33516941 PMCID: PMC8042406 DOI: 10.1016/j.mcpro.2021.100050] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 02/06/2023] Open
Abstract
Ubiquitination is a posttranslational protein modification that has been shown to have a range of effects, including regulation of protein function, interaction, localization, and degradation. We have previously shown that the muscle-specific ubiquitin E3 ligase, ASB2β, is downregulated in models of muscle growth and that overexpression ASB2β is sufficient to induce muscle atrophy. To gain insight into the effects of increased ASB2β expression on skeletal muscle mass and function, we used liquid chromatography coupled to tandem mass spectrometry to investigate ASB2β-mediated changes to the skeletal muscle proteome and ubiquitinome, via a parallel analysis of remnant diGly-modified peptides. The results show that viral vector-mediated ASB2β overexpression in murine muscles causes progressive muscle atrophy and impairment of force-producing capacity, while ASB2β knockdown induces mild muscle hypertrophy. ASB2β-induced muscle atrophy and dysfunction were associated with the early downregulation of mitochondrial and contractile protein abundance and the upregulation of proteins involved in proteasome-mediated protein degradation (including other E3 ligases), protein synthesis, and the cytoskeleton/sarcomere. The overexpression ASB2β also resulted in marked changes in protein ubiquitination; however, there was no simple relationship between changes in ubiquitination status and protein abundance. To investigate proteins that interact with ASB2β and, therefore, potential ASB2β targets, Flag-tagged wild-type ASB2β, and a mutant ASB2β lacking the C-terminal SOCS box domain (dSOCS) were immunoprecipitated from C2C12 myotubes and subjected to label-free proteomic analysis to determine the ASB2β interactome. ASB2β was found to interact with a range of cytoskeletal and nuclear proteins. When combined with the in vivo ubiquitinomic data, our studies have identified novel putative ASB2β target substrates that warrant further investigation. These findings provide novel insight into the complexity of proteome and ubiquitinome changes that occur during E3 ligase-mediated skeletal muscle atrophy and dysfunction.
Collapse
Affiliation(s)
- Craig A Goodman
- Department of Physiology, Centre for Muscle Research (CMR), The University of Melbourne, Victoria, Australia; Australian Institute for Musculoskeletal Science (AIMSS), Sunshine Hospital, The University of Melbourne, St Albans, Victoria, Australia
| | - Jonathan R Davey
- Department of Physiology, Centre for Muscle Research (CMR), The University of Melbourne, Victoria, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Adam Hagg
- Department of Physiology, Centre for Muscle Research (CMR), The University of Melbourne, Victoria, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Benjamin L Parker
- Department of Physiology, Centre for Muscle Research (CMR), The University of Melbourne, Victoria, Australia; Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney, NSW, Australia.
| | - Paul Gregorevic
- Department of Physiology, Centre for Muscle Research (CMR), The University of Melbourne, Victoria, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia; Department of Neurology, The University of Washington School of Medicine, Seattle, Washington, USA.
| |
Collapse
|
20
|
Wiedmer P, Jung T, Castro JP, Pomatto LC, Sun PY, Davies KJ, Grune T. Sarcopenia - Molecular mechanisms and open questions. Ageing Res Rev 2021; 65:101200. [PMID: 33130247 DOI: 10.1016/j.arr.2020.101200] [Citation(s) in RCA: 206] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022]
Abstract
Sarcopenia represents a muscle-wasting syndrome characterized by progressive and generalized degenerative loss of skeletal muscle mass, quality, and strength occurring during normal aging. Sarcopenia patients are mainly suffering from the loss in muscle strength and are faced with mobility disorders reducing their quality of life and are, therefore, at higher risk for morbidity (falls, bone fracture, metabolic diseases) and mortality. Several molecular mechanisms have been described as causes for sarcopenia that refer to very different levels of muscle physiology. These mechanisms cover e. g. function of hormones (e. g. IGF-1 and Insulin), muscle fiber composition and neuromuscular drive, myo-satellite cell potential to differentiate and proliferate, inflammatory pathways as well as intracellular mechanisms in the processes of proteostasis and mitochondrial function. In this review, we describe sarcopenia as a muscle-wasting syndrome distinct from other atrophic diseases and summarize the current view on molecular causes of sarcopenia development as well as open questions provoking further research efforts for establishing efficient lifestyle and therapeutic interventions.
Collapse
|
21
|
Adams V, Gußen V, Zozulya S, Cruz A, Moriscot A, Linke A, Labeit S. Small-Molecule Chemical Knockdown of MuRF1 in Melanoma Bearing Mice Attenuates Tumor Cachexia Associated Myopathy. Cells 2020; 9:E2272. [PMID: 33050629 PMCID: PMC7600862 DOI: 10.3390/cells9102272] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
: Patients with malignant tumors frequently suffer during disease progression from a syndrome referred to as cancer cachexia (CaCax): CaCax includes skeletal muscle atrophy and weakness, loss of bodyweight, and fat tissues. Currently, there are no FDA (Food and Drug Administration) approved treatments available for CaCax. Here, we studied skeletal muscle atrophy and dysfunction in a murine CaCax model by injecting B16F10 melanoma cells into mouse thighs and followed mice during melanoma outgrowth. Skeletal muscles developed progressive weakness as detected by wire hang tests (WHTs) during days 13-23. Individual muscles analyzed at day 24 had atrophy, mitochondrial dysfunction, augmented metabolic reactive oxygen species (ROS) stress, and a catabolically activated ubiquitin proteasome system (UPS), including upregulated MuRF1. Accordingly, we tested as an experimental intervention of recently identified small molecules, Myomed-205 and -946, that inhibit MuRF1 activity and MuRF1/MuRF2 expression. Results indicate that MuRF1 inhibitor fed attenuated induction of MuRF1 in tumor stressed muscles. In addition, the compounds augmented muscle performance in WHTs and attenuated muscle weight loss. Myomed-205 and -946 also rescued citrate synthase and complex-1 activities in tumor-stressed muscles, possibly suggesting that mitochondrial-metabolic and muscle wasting effects in this CaCax model are mechanistically connected. Inhibition of MuRF1 during tumor cachexia may represent a suitable strategy to attenuate skeletal muscle atrophy and dysfunction.
Collapse
Affiliation(s)
- Volker Adams
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, 1307 Dresden, Germany; (V.G.); (A.L.)
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, 01067 Dresden, Germany
| | - Victoria Gußen
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, 1307 Dresden, Germany; (V.G.); (A.L.)
| | - Sergey Zozulya
- Department of Drug Research, Enamine-Bienta Ltd., 02000 Kiev, Ukraine;
| | - André Cruz
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil; (A.C.); (A.M.)
| | - Anselmo Moriscot
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil; (A.C.); (A.M.)
| | - Axel Linke
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, 1307 Dresden, Germany; (V.G.); (A.L.)
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, 01067 Dresden, Germany
| | - Siegfried Labeit
- Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany;
| |
Collapse
|
22
|
Peris-Moreno D, Taillandier D, Polge C. MuRF1/TRIM63, Master Regulator of Muscle Mass. Int J Mol Sci 2020; 21:ijms21186663. [PMID: 32933049 PMCID: PMC7555135 DOI: 10.3390/ijms21186663] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
The E3 ubiquitin ligase MuRF1/TRIM63 was identified 20 years ago and suspected to play important roles during skeletal muscle atrophy. Since then, numerous studies have been conducted to decipher the roles, molecular mechanisms and regulation of this enzyme. This revealed that MuRF1 is an important player in the skeletal muscle atrophy process occurring during catabolic states, making MuRF1 a prime candidate for pharmacological treatments against muscle wasting. Indeed, muscle wasting is an associated event of several diseases (e.g., cancer, sepsis, diabetes, renal failure, etc.) and negatively impacts the prognosis of patients, which has stimulated the search for MuRF1 inhibitory molecules. However, studies on MuRF1 cardiac functions revealed that MuRF1 is also cardioprotective, revealing a yin and yang role of MuRF1, being detrimental in skeletal muscle and beneficial in the heart. This review discusses data obtained on MuRF1, both in skeletal and cardiac muscles, over the past 20 years, regarding the structure, the regulation, the location and the different functions identified, and the first inhibitors reported, and aim to draw the picture of what is known about MuRF1. The review also discusses important MuRF1 characteristics to consider for the design of future drugs to maintain skeletal muscle mass in patients with different pathologies.
Collapse
|
23
|
Yoshida T, Delafontaine P. Mechanisms of IGF-1-Mediated Regulation of Skeletal Muscle Hypertrophy and Atrophy. Cells 2020; 9:cells9091970. [PMID: 32858949 PMCID: PMC7564605 DOI: 10.3390/cells9091970] [Citation(s) in RCA: 301] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/18/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) is a key growth factor that regulates both anabolic and catabolic pathways in skeletal muscle. IGF-1 increases skeletal muscle protein synthesis via PI3K/Akt/mTOR and PI3K/Akt/GSK3β pathways. PI3K/Akt can also inhibit FoxOs and suppress transcription of E3 ubiquitin ligases that regulate ubiquitin proteasome system (UPS)-mediated protein degradation. Autophagy is likely inhibited by IGF-1 via mTOR and FoxO signaling, although the contribution of autophagy regulation in IGF-1-mediated inhibition of skeletal muscle atrophy remains to be determined. Evidence has suggested that IGF-1/Akt can inhibit muscle atrophy-inducing cytokine and myostatin signaling via inhibition of the NF-κΒ and Smad pathways, respectively. Several miRNAs have been found to regulate IGF-1 signaling in skeletal muscle, and these miRs are likely regulated in different pathological conditions and contribute to the development of muscle atrophy. IGF-1 also potentiates skeletal muscle regeneration via activation of skeletal muscle stem (satellite) cells, which may contribute to muscle hypertrophy and/or inhibit atrophy. Importantly, IGF-1 levels and IGF-1R downstream signaling are suppressed in many chronic disease conditions and likely result in muscle atrophy via the combined effects of altered protein synthesis, UPS activity, autophagy, and muscle regeneration.
Collapse
Affiliation(s)
- Tadashi Yoshida
- Heart and Vascular Institute, John W. Deming Department of Medicine, Tulane University School of Medicine, 1430 Tulane Ave SL-48, New Orleans, LA 70112, USA
- Department of Physiology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
- Correspondence: (T.Y.); (P.D.)
| | - Patrice Delafontaine
- Heart and Vascular Institute, John W. Deming Department of Medicine, Tulane University School of Medicine, 1430 Tulane Ave SL-48, New Orleans, LA 70112, USA
- Department of Physiology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
- Correspondence: (T.Y.); (P.D.)
| |
Collapse
|
24
|
Scalabrin M, Adams V, Labeit S, Bowen TS. Emerging Strategies Targeting Catabolic Muscle Stress Relief. Int J Mol Sci 2020; 21:E4681. [PMID: 32630118 PMCID: PMC7369951 DOI: 10.3390/ijms21134681] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle wasting represents a common trait in many conditions, including aging, cancer, heart failure, immobilization, and critical illness. Loss of muscle mass leads to impaired functional mobility and severely impedes the quality of life. At present, exercise training remains the only proven treatment for muscle atrophy, yet many patients are too ill, frail, bedridden, or neurologically impaired to perform physical exertion. The development of novel therapeutic strategies that can be applied to an in vivo context and attenuate secondary myopathies represents an unmet medical need. This review discusses recent progress in understanding the molecular pathways involved in regulating skeletal muscle wasting with a focus on pro-catabolic factors, in particular, the ubiquitin-proteasome system and its activating muscle-specific E3 ligase RING-finger protein 1 (MuRF1). Mechanistic progress has provided the opportunity to design experimental therapeutic concepts that may affect the ubiquitin-proteasome system and prevent subsequent muscle wasting, with novel advances made in regards to nutritional supplements, nuclear factor kappa-light-chain-enhancer of activated B cells (NFB) inhibitors, myostatin antibodies, β2 adrenergic agonists, and small-molecules interfering with MuRF1, which all emerge as a novel in vivo treatment strategies for muscle wasting.
Collapse
Affiliation(s)
- Mattia Scalabrin
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK;
| | - Volker Adams
- Department of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany;
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, 01067 Dresden, Germany
| | - Siegfried Labeit
- Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany;
- Myomedix GmbH, Im Biengarten 36, 69151 Neckargemünd, Germany
| | - T. Scott Bowen
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK;
| |
Collapse
|
25
|
Adams V, Bowen TS, Werner S, Barthel P, Amberger C, Konzer A, Graumann J, Sehr P, Lewis J, Provaznik J, Benes V, Büttner P, Gasch A, Mangner N, Witt CC, Labeit D, Linke A, Labeit S. Small-molecule-mediated chemical knock-down of MuRF1/MuRF2 and attenuation of diaphragm dysfunction in chronic heart failure. J Cachexia Sarcopenia Muscle 2019; 10:1102-1115. [PMID: 31140761 PMCID: PMC6818456 DOI: 10.1002/jcsm.12448] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Chronic heart failure (CHF) leads to diaphragm myopathy that significantly impairs quality of life and worsens prognosis. In this study, we aimed to assess the efficacy of a recently discovered small-molecule inhibitor of MuRF1 in treating CHF-induced diaphragm myopathy and loss of contractile function. METHODS Myocardial infarction was induced in mice by ligation of the left anterior descending coronary artery. Sham-operated animals (sham) served as controls. One week post-left anterior descending coronary artery ligation animals were randomized into two groups-one group was fed control rodent chow, whereas the other group was fed a diet containing 0.1% of the compound ID#704946-a recently described MuRF1-interfering small molecule. Echocardiography confirmed development of CHF after 10 weeks. Functional and molecular analysis of the diaphragm was subsequently performed. RESULTS Chronic heart failure induced diaphragm fibre atrophy and contractile dysfunction by ~20%, as well as decreased activity of enzymes involved in mitochondrial energy production (P < 0.05). Treatment with compound ID#704946 in CHF mice had beneficial effects on the diaphragm: contractile function was protected, while mitochondrial enzyme activity and up-regulation of the MuRF1 and MuRF2 was attenuated after infarct. CONCLUSIONS Our murine CHF model presented with diaphragm fibre atrophy, impaired contractile function, and reduced mitochondrial enzyme activities. Compound ID#704946 rescued from this partially, possibly by targeting MuRF1/MuRF2. However, at this stage of our study, we refrain to claim specific mechanism(s) and targets of compound ID#704946, because the nature of changes after 12 weeks of feeding is likely to be complex and is not necessarily caused by direct mechanistic effects.
Collapse
Affiliation(s)
- Volker Adams
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, Dresden, Germany
| | - T Scott Bowen
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Sarah Werner
- University Clinic of Cardiology, Heart Center Leipzig, Leipzig, Germany
| | - Peggy Barthel
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, Dresden, Germany
| | | | - Anne Konzer
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Rhine-Main, Germany
| | - Johannes Graumann
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Rhine-Main, Germany
| | - Peter Sehr
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Joe Lewis
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jan Provaznik
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Vladimir Benes
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Petra Büttner
- University Clinic of Cardiology, Heart Center Leipzig, Leipzig, Germany
| | - Alexander Gasch
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Norman Mangner
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, Dresden, Germany
| | - Christian C Witt
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Dittmar Labeit
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.,Myomedix GmbH, Neckargemünd, Germany
| | - Axel Linke
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, Dresden, Germany
| | - Siegfried Labeit
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.,Myomedix GmbH, Neckargemünd, Germany
| |
Collapse
|
26
|
Mizgier ML, Fernández-Verdejo R, Cherfan J, Pinget M, Bouzakri K, Galgani JE. Insights on the Role of Putative Muscle-Derived Factors on Pancreatic Beta Cell Function. Front Physiol 2019; 10:1024. [PMID: 31440170 PMCID: PMC6694406 DOI: 10.3389/fphys.2019.01024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/24/2019] [Indexed: 01/02/2023] Open
Abstract
Skeletal muscle is a main target of insulin action that plays a pivotal role in postprandial glucose disposal. Importantly, skeletal muscle insulin sensitivity relates inversely with pancreatic insulin secretion, which prompted the hypothesis of the existence of a skeletal muscle-pancreas crosstalk mediated through an endocrine factor. The observation that changes in skeletal muscle glucose metabolism are accompanied by altered insulin secretion supports this hypothesis. Meanwhile, a muscle-derived circulating factor affecting in vivo insulin secretion remains elusive. This factor may correspond to peptides/proteins (so called myokines), exosomes and their cargo, and metabolites. We hereby review the most remarkable evidence encouraging the possibility of such inter-organ communication, with special focus on muscle-derived factors that may potentially mediate such skeletal muscle-pancreas crosstalk.
Collapse
Affiliation(s)
- Maria L Mizgier
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Rodrigo Fernández-Verdejo
- Departamento de Ciencias de la Salud, Nutrición y Dietética, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Julien Cherfan
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Michel Pinget
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Karim Bouzakri
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Jose E Galgani
- Departamento de Ciencias de la Salud, Nutrición y Dietética, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
27
|
Rao VV, Sangiah U, Mary KA, Akira S, Mohanty A. Role of Akirin1 in the regulation of skeletal muscle fiber-type switch. J Cell Biochem 2019; 120:11284-11304. [PMID: 30746755 DOI: 10.1002/jcb.28406] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/19/2018] [Accepted: 01/07/2019] [Indexed: 01/24/2023]
Abstract
Akirin1 is a highly conserved ubiquitously expressed nuclear protein. Owing to its strong nuclear localization signal and protein-protein interaction properties, Akirin1 has been speculated to regulate transcription of target genes as a cofactor. Previous studies have reported Akirin1 as a downstream target of myostatin, a potent negative regulator of myogenesis. Mice lacking myostatin displayed enhanced Akirin1 gene expression. Further, in vitro evidence has shown Akirin1 overexpression leads to hypertrophy in C2 C 12 myotubes. In this study, we used Akirin1 knockout mice as a model system to further elucidate the function of Akirin1 in fully differentiated skeletal muscle. Akirin1 knockout mice did not show any obvious phenotypic difference when compared with wild type. However, promoter-reporter assay suggested that Akirin1 regulated the transcription of muscle-specific RING finger 1 (MuRF-1), an important E3 ubiquitin ligase in skeletal muscle. Furthermore, ablation of Akirin1 resulted in increased type IIa and decreased type I muscle fibers, which was further supported by an increase in Myh2 and decrease in Myh7 gene expression. Also, histochemical studies for succinate dehydrogenase activity revealed a less oxidative muscle in the absence of Akirin1. Together, our study suggests a novel role of Akirin1 in maintaining the muscle fiber type and regulation of the metabolic activity of the skeletal muscle.
Collapse
Affiliation(s)
- Vanitha Venkoba Rao
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Umamaheswari Sangiah
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kavitha Arockia Mary
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shizuo Akira
- Department of Host Defense, Osaka University, Suita, Japan
| | - Abhishek Mohanty
- Department of Molecular Oncology, MVR Cancer Center and Research Institute, Kozhikode, India
| |
Collapse
|
28
|
Sharlo K, Paramonova I, Turtikova O, Tyganov S, Shenkman B. Plantar mechanical stimulation prevents calcineurin-NFATc1 inactivation and slow-to-fast fiber type shift in rat soleus muscle under hindlimb unloading. J Appl Physiol (1985) 2019; 126:1769-1781. [PMID: 31046517 DOI: 10.1152/japplphysiol.00029.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The prevailing myosin isoform [myosin heavy chain (MyHC)] in a skeletal muscle determines contractile properties of the muscle. Under actual or simulated microgravity conditions such as human bed rest or rat hindlimb unloading, decrease in expression of MyHC of the slow type [MyHC I(β)] has been observed. It was demonstrated that increasing sensory input by performing plantar mechanical stimulation (PMS) on the soles of the feet results in an increase in neuromuscular activation of the lower limb muscles and may prevent slow-to-fast fiber type shift. The calcineurin-nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) signaling pathway is the main cascade regulating MyHC I(β) expression. The present study was aimed to analyze the states of the calcineurin-NFATc1 signaling cascade under conditions of PMS during rat hindlimb unloading. Male Wistar rats were randomly assigned to vivarium control groups and 1-day unloading (1HS), 3-day unloading (3HS), 1HS+PMS, and 3HS+PMS groups. We found that both 1 day and 3 days of unloading caused decrease in MyHC I(β) mRNA expression and decrease in glycogen synthase kinase-3β phosphorylation (Ser 9) that brought about the kinase activation, and these effects of unloading were prevented by PMS. Three days of unloading also caused increase in expression of calsarcin-2 (myozenin-I), which was found to be the endogenous calcineurin inhibitor. The level of calsarcin-2 expression in the 3HS+PMS group did not differ from the control group. Therefore, we conclude that PMS upregulates the calcineurin-NFATc1 signaling pathway and prevents unloading-induced MyHC I(β) decrease. NEW & NOTEWORTHY It is widely accepted that changes in the myosin phenotype during functional unloading (disuse) are determined by a decreased expression of the myosin heavy chain (MyHC) I(β) gene, and this decrease leads to changes of contractile and fatigue characteristics of soleus muscle. The calcineurin-nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) pathway is one of the most important signaling cascades regulating slow MyHC isoform expression. The present study is the first to show that plantar mechanical stimulation upregulates calcineurin-NFATc1 signaling in soleus muscles of hindlimb-unloaded rats.
Collapse
Affiliation(s)
- Kristina Sharlo
- Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| | - Inna Paramonova
- Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| | - Olga Turtikova
- Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| | - Sergey Tyganov
- Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| | - Boris Shenkman
- Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| |
Collapse
|
29
|
Sun X, Zhao D, Lu F, Peng S, Yu M, Liu N, Sun Y, Du H, Wang B, Chen J, Dong S, Lu F, Zhang W. Hydrogen sulfide regulates muscle RING finger-1 protein S-sulfhydration at Cys 44 to prevent cardiac structural damage in diabetic cardiomyopathy. Br J Pharmacol 2019; 177:836-856. [PMID: 30734268 DOI: 10.1111/bph.14601] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 12/04/2018] [Accepted: 12/19/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Hydrogen sulfide (H2 S) plays important roles as a gasotransmitter in pathologies. Increased expression of the E3 ubiquitin ligase, muscle RING finger-1 (MuRF1), may be involved in diabetic cardiomyopathy. Here we have investigated whether and how exogenous H2 S alleviates cardiac muscle degradation through modifications of MuRF1 S-sulfhydration in db/db mice. EXPERIMENTAL APPROACH Neonatal rat cardiomyocytes were treated with high glucose (40 mM), oleate (100 μM), palmitate (400 μM), and NaHS (100 μM) for 72 hr. MuRF1 was silenced with siRNA technology and mutation at Cys44 . Endoplasmic reticulum stress markers, MuRF1 expression, and ubiquitination level were measured. db/db mice were injected with NaHS (39 μmol·kg-1 ) for 20 weeks. Echocardiography, cardiac ultrastructure, cystathionine-γ-lyase, cardiac structure proteins expression, and S-sulfhydration production were measured. KEY RESULTS H2 S levels and cystathionine-γ-lyase protein expression in myocardium were decreased in db/db mice. Exogenous H2 S reversed endoplasmic reticulum stress, including impairment of the function of cardiomyocytes and structural damage in db/db mice. Exogenous H2 S could suppress the levels of myosin heavy chain 6 and myosin light chain 2 ubiquitination in cardiac tissues of db/db mice, and MuRF1 was modified by S-sulfhydration, following treatment with exogenous H2 S, to reduce the interaction between MuRF1 and myosin heavy chain 6 and myosin light chain 2. CONCLUSIONS AND IMPLICATIONS Our findings suggest that H2 S regulates MuRF1 S-sulfhydration at Cys44 to prevent myocardial degradation in the cardiac tissues of db/db mice. LINKED ARTICLES This article is part of a themed section on Hydrogen Sulfide in Biology & Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.4/issuetoc.
Collapse
Affiliation(s)
- Xiaojiao Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Dechao Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fangping Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shuo Peng
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Miao Yu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Ning Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yu Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Haining Du
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Bingzhu Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Jian Chen
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shiyun Dong
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Fanghao Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China.,Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University Ministry of Education, Harbin, China
| |
Collapse
|
30
|
Rodrigues ACZ, Messi ML, Wang ZM, Abba MC, Pereyra A, Birbrair A, Zhang T, O’Meara M, Kwan P, Lopez EIS, Willis MS, Mintz A, Files DC, Furdui C, Oppenheim RW, Delbono O. The sympathetic nervous system regulates skeletal muscle motor innervation and acetylcholine receptor stability. Acta Physiol (Oxf) 2019; 225:e13195. [PMID: 30269419 PMCID: PMC7224611 DOI: 10.1111/apha.13195] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 09/23/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022]
Abstract
AIM Symptoms of autonomic failure are frequently the presentation of advanced age and neurodegenerative diseases that impair adaptation to common physiologic stressors. The aim of this work was to examine the interaction between the sympathetic and motor nervous system, the involvement of the sympathetic nervous system (SNS) in neuromuscular junction (NMJ) presynaptic motor function, the stability of postsynaptic molecular organization, and the skeletal muscle composition and function. METHODS Since muscle weakness is a symptom of diseases characterized by autonomic dysfunction, we studied the impact of regional sympathetic ablation on muscle motor innervation by using transcriptome analysis, retrograde tracing of the sympathetic outflow to the skeletal muscle, confocal and electron microscopy, NMJ transmission by electrophysiological methods, protein analysis, and state of the art microsurgical techniques, in C57BL6, MuRF1KO and Thy-1 mice. RESULTS We found that the SNS regulates motor nerve synaptic vesicle release, skeletal muscle transcriptome, muscle force generated by motor nerve activity, axonal neurofilament phosphorylation, myelin thickness, and myofibre subtype composition and CSA. The SNS also modulates the levels of postsynaptic membrane acetylcholine receptor by regulating the Gαi2 -Hdac4-Myogenin-MuRF1pathway, which is prevented by the overexpression of the guanine nucleotide-binding protein Gαi2 (Q205L), a constitutively active mutant G protein subunit. CONCLUSION The SNS regulates NMJ transmission, maintains optimal Gαi2 expression, and prevents any increase in Hdac4, myogenin, MuRF1, and miR-206. SNS ablation leads to upregulation of MuRF1, muscle atrophy, and downregulation of postsynaptic AChR. Our findings are relevant to clinical conditions characterized by progressive decline of sympathetic innervation, such as neurodegenerative diseases and aging.
Collapse
Affiliation(s)
- Anna C. Z. Rodrigues
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Maria L. Messi
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Zhong-Min Wang
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Martin C. Abba
- Basic and Applied Immunological Research Center (CINIBA), School of Medicine, National University of La Plata, Buenos Aires, Argentina
| | - Andrea Pereyra
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Alexander Birbrair
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tan Zhang
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Meaghan O’Meara
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ping Kwan
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Elsa I. S. Lopez
- Department of Internal Medicine, Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Monte S. Willis
- Department of Pathology, McAllister Heart Institute, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina
| | - Akiva Mintz
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - D. Clark Files
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Internal Medicine, Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Internal Medicine, Pulmonary, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Cristina Furdui
- Department of Internal Medicine, Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ronald W. Oppenheim
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Osvaldo Delbono
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
31
|
Nowak M, Suenkel B, Porras P, Migotti R, Schmidt F, Kny M, Zhu X, Wanker EE, Dittmar G, Fielitz J, Sommer T. DCAF8, a novel MuRF1 interaction partner, promotes muscle atrophy. J Cell Sci 2019; 132:jcs.233395. [DOI: 10.1242/jcs.233395] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/22/2019] [Indexed: 12/29/2022] Open
Abstract
The muscle-specific RING-finger protein MuRF1 constitutes a bona fide ubiquitin ligase that routes proteins like Myosin heavy chain (MyHC) to proteasomal degradation during muscle atrophy. In two unbiased screens we identified DCAF8 as a new MuRF1 binding partner. MuRF1 physically interacts with DCAF8 and both proteins localize to overlapping structures in muscle cells. Noteworthy, similar to MuRF1, DCAF8 levels increase during atrophy and the down-regulation of either protein substantially impedes muscle wasting and MyHC degradation in C2C12 myotubes, a model system for muscle differentiation and atrophy. DCAF proteins typically serve as substrate receptors in Cullin 4-type (Cul4) ubiquitin ligases (CRL) and we demonstrate that DCAF8 and MuRF1 associate with the subunits of such a protein complex. Because genetic downregulation of DCAF8 and inhibition of Cullin activity also impair myotube atrophy in C2C12 cells, our data imply that the DCAF8 promotes muscle wasting by targeting proteins like MyHC as an integral substrate receptor of a CRL4A ubiquitin ligase.
Collapse
Affiliation(s)
- Marcel Nowak
- Intracellular Proteolysis, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Strasse 10, 13125 Berlin-Buch, Germany
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin, MDC, Lindenberger Weg 80, 13125 Berlin-Buch, Germany
- Present address: DUNN Labortechnik GmbH, Thelenberg 6, 53567, Asbach, Germany
| | - Benjamin Suenkel
- Intracellular Proteolysis, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Strasse 10, 13125 Berlin-Buch, Germany
| | - Pablo Porras
- Proteomics and Molecular Mechanisms of Neurodegenerative Diseases, MDC, USA
- Present address: European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, CB10 1SD, UK
| | - Rebekka Migotti
- Mass Spectrometric Core Unit, MDC, USA
- Present address: ProPharma Group, Siemensdamm 62, 13627 Berlin, Germany
| | - Franziska Schmidt
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin, MDC, Lindenberger Weg 80, 13125 Berlin-Buch, Germany
- Present address: BCRT Flow and Mass Cytometry Lab, Charité – Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Melanie Kny
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin, MDC, Lindenberger Weg 80, 13125 Berlin-Buch, Germany
| | - Xiaoxi Zhu
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin, MDC, Lindenberger Weg 80, 13125 Berlin-Buch, Germany
| | - Erich E. Wanker
- Proteomics and Molecular Mechanisms of Neurodegenerative Diseases, MDC, USA
| | - Gunnar Dittmar
- Mass Spectrometric Core Unit, MDC, USA
- Present address: Proteome and Genome Research Laboratory, Luxembourg Institute of Health, 1a Rue Thomas Edison, L-1445 Strassen, Luxembourg, Europe
| | - Jens Fielitz
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin, MDC, Lindenberger Weg 80, 13125 Berlin-Buch, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Fleischmann Strasse 41, 17475 Greifswald, Germany
| | - Thomas Sommer
- Intracellular Proteolysis, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Strasse 10, 13125 Berlin-Buch, Germany
- Institute of Biology, Humboldt-University Berlin, Invalidenstrasse 43, 10115 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Oudenarder Straße 16, 13347 Berlin, Germany
| |
Collapse
|
32
|
Samir SM, Abbas AM, Safwat SM, Elserougy HG. Effect of L-carnitine on diabetes-induced changes of skeletal muscles in rats. J Basic Clin Physiol Pharmacol 2018; 29:47-59. [PMID: 28981442 DOI: 10.1515/jbcpp-2016-0185] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 06/18/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Patients with diabetes mellitus (DM) are at risk of experiencing chronic complications such as retinopathy, nephropathy and myopathy. We aimed to evaluate the effects of L-carnitine on type II DM (T2DM)-induced biochemical, contractile and pathological changes in skeletal muscles of rats. METHODS Thirty-two male Sprague Dawley rats were divided into the control, control+L-carnitine, T2DM and T2DM+L-carnitine groups. Plasma levels of glucose, insulin, malondialdehyde and antioxidants such as reduced glutathione, catalase and superoxide dismutase, haemoglobin A1c (HbA1c), insulin sensitivity index (ISI) as well as the contractile properties of the gastrocnemius muscle were measured. Also, histopathological studies and immunohistochemical examination of the gastrocnemius muscle using the MuRF1 (muscle RING-finger protein-1) marker were performed. RESULTS In diabetic rats, malondialdehyde, glucose, insulin, HbA1c and MuRF1 were increased, whereas ISI and antioxidants were decreased and the contractile properties deteriorated. L-carnitine decreased malondialdehyde, glucose, insulin, HbA1c and MuRF1 and increased ISI and antioxidants. Also, L-carnitine improves the contractile properties in diabetic rats. Histopathological studies confirm our data. CONCLUSIONS We conclude that L-carnitine exhibits protective effects on skeletal muscles of T2DM rats through its hypoglycemic and antioxidant actions as well as its inhibitory effect on protein degradation.
Collapse
Affiliation(s)
- Shereen M Samir
- Medical Physiology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amr M Abbas
- Medical Physiology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Sally M Safwat
- Medical Physiology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Hanaa G Elserougy
- Medical Physiology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
33
|
Tewari N, Awad S, Duška F, Williams JP, Bennett A, Macdonald IA, Lobo DN. Postoperative inflammation and insulin resistance in relation to body composition, adiposity and carbohydrate treatment: A randomised controlled study. Clin Nutr 2018; 38:204-212. [PMID: 29454501 PMCID: PMC6380471 DOI: 10.1016/j.clnu.2018.01.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 01/25/2018] [Indexed: 12/17/2022]
Abstract
Background & aims The aims of this study were to identify whether differences in distribution of adipose tissue and skeletal muscle in obese and non-obese individuals contribute to the magnitude of the postoperative inflammatory response and insulin resistance, with and without preoperative treatment with carbohydrate drinks. Methods Thirty-two adults (16 obese/16 non-obese) undergoing elective major open abdominal surgery participated in this 2 × 2 factorial, randomised, double-blind, placebo-controlled study. Participants received Nutricia preOp® or placebo (800 ml on the night before surgery/400 ml 2–3 h preoperatively) after stratifying for obesity. Insulin sensitivity was measured using the hyperinsulinaemic-euglycaemic clamp preoperatively and on the 1st postoperative day. Vastus lateralis, omental and subcutaneous fat biopsies were taken pre- and postoperatively and analysed after RNA extraction. The primary endpoint was within subject differences in insulin sensitivity. Results Major abdominal surgery was associated with a 42% reduction in insulin sensitivity from mean(SD) M value of 37.3(11.8) μmol kg−1 fat free mass (FFM) to 21.7(7.4) μmol kg−1 FFM, but this was not influenced by obesity or preoperative carbohydrate treatment. Activation of the triggering receptor expressed on myeloid cells (TREM1) pathway was seen in response to surgery in omental fat samples. In postoperative muscle samples, gene expression differences indicated activation of the peroxisome proliferator-activated receptor (PPAR-α)/retinoid X-receptor (RXR-α) pathway in obese but not in non-obese participants. There were no significant changes in gene expression pathways associated with carbohydrate treatment. Conclusion The reduction in insulin sensitivity associated with major abdominal surgery was confirmed but there were no differences associated with preoperative carbohydrates or obesity.
Collapse
Affiliation(s)
- Nilanjana Tewari
- Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Sherif Awad
- Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK; The East-Midlands Bariatric and Metabolic Institute (EMBMI), Derby Teaching Hospitals NHS Foundation Trust, Royal Derby Hospital, Derby DE22 3NE, UK
| | - František Duška
- Department of Anaesthesia and Intensive Care, Kralovske Vinohrady University Hospital and The Third Faculty of Medicine, Prague, Czech Republic; Department of Critical Care, Nottingham University Hospitals NHS Trust, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Julian P Williams
- Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Andrew Bennett
- FRAME Laboratory, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK; School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Ian A Macdonald
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK; MRC/ARUK Centre for Musculoskeletal Ageing Research, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Dileep N Lobo
- Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK; MRC/ARUK Centre for Musculoskeletal Ageing Research, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| |
Collapse
|
34
|
Polge C, Cabantous S, Deval C, Claustre A, Hauvette A, Bouchenot C, Aniort J, Béchet D, Combaret L, Attaix D, Taillandier D. A muscle-specific MuRF1-E2 network requires stabilization of MuRF1-E2 complexes by telethonin, a newly identified substrate. J Cachexia Sarcopenia Muscle 2018; 9:129-145. [PMID: 29271608 PMCID: PMC5803617 DOI: 10.1002/jcsm.12249] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/21/2017] [Accepted: 09/05/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Muscle wasting is observed in the course of many diseases and also during physiological conditions (disuse, ageing). Skeletal muscle mass is largely controlled by the ubiquitin-proteasome system and thus by the ubiquitinating enzymes (E2s and E3s) that target substrates for subsequent degradation. MuRF1 is the only E3 ubiquitin ligase known to target contractile proteins (α-actin, myosins) during catabolic situations. However, MuRF1 depends on E2 ubiquitin-conjugating enzymes for ubiquitin chain formation on the substrates. MuRF1-E2 couples are therefore putative targets for preventing muscle wasting. METHODS We focused on 14 E2 enzymes that are either expressed in skeletal muscle or up-regulated during atrophying conditions. In this work, we demonstrated that only highly sensitive and complementary interactomic approaches (surface plasmon resonance, yeast three-hybrid, and split green fluorescent protein) allowed the identification of MuRF1 E2 partners. RESULTS Five E2 enzymes physically interacted with MuRF1, namely, E2E1, E2G1, E2J1, E2J2, and E2L3. Moreover, we demonstrated that MuRF1-E2E1 and MuRF1-E2J1 interactions are facilitated by telethonin, a newly identified MuRF1 substrate. We next showed that the five identified E2s functionally interacted with MuRF1 since, in contrast to the non-interacting E2D2, their co-expression in HEK293T cells with MuRF1 led to increased telethonin degradation. Finally, we showed that telethonin governed the affinity between MuRF1 and E2E1 or E2J1. CONCLUSIONS We report here the first MuRF1-E2s network, which may prove valuable for deciphering the precise mechanisms involved in the atrophying muscle programme and for proposing new therapeutical approaches.
Collapse
Affiliation(s)
- Cécile Polge
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Stéphanie Cabantous
- Cancer Research Center of Toulouse, INSERM UMR 1037F‐31037ToulouseFrance
- Université de ToulouseF‐31062ToulouseFrance
| | - Christiane Deval
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Agnès Claustre
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Antoine Hauvette
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Catherine Bouchenot
- Cancer Research Center of Toulouse, INSERM UMR 1037F‐31037ToulouseFrance
- Université de ToulouseF‐31062ToulouseFrance
| | - Julien Aniort
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
- Service de Néphrologie Réanimation Médicale, Pôle Respiratoire, Endocrinologie‐Diabétologie, Urologie, Néphrologie‐Dialyse, Nutrition Clinique, InfectiologieRéanimation Médicale, Hygiène Hospitalière (REUNNIRH)F‐63000Clermont‐FerrandFrance
| | - Daniel Béchet
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Lydie Combaret
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Didier Attaix
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Daniel Taillandier
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| |
Collapse
|
35
|
Crawford Parks TE, Ravel-Chapuis A, Bondy-Chorney E, Renaud JM, Côté J, Jasmin BJ. Muscle-specific expression of the RNA-binding protein Staufen1 induces progressive skeletal muscle atrophy via regulation of phosphatase tensin homolog. Hum Mol Genet 2017; 26:1821-1838. [PMID: 28369467 DOI: 10.1093/hmg/ddx085] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 03/02/2017] [Indexed: 12/14/2022] Open
Abstract
Converging lines of evidence have now highlighted the key role for post-transcriptional regulation in the neuromuscular system. In particular, several RNA-binding proteins are known to be misregulated in neuromuscular disorders including myotonic dystrophy type 1, spinal muscular atrophy and amyotrophic lateral sclerosis. In this study, we focused on the RNA-binding protein Staufen1, which assumes multiple functions in both skeletal muscle and neurons. Given our previous work that showed a marked increase in Staufen1 expression in various physiological and pathological conditions including denervated muscle, in embryonic and undifferentiated skeletal muscle, in rhabdomyosarcomas as well as in myotonic dystrophy type 1 muscle samples from both mouse models and humans, we investigated the impact of sustained Staufen1 expression in postnatal skeletal muscle. To this end, we generated a skeletal muscle-specific transgenic mouse model using the muscle creatine kinase promoter to drive tissue-specific expression of Staufen1. We report that sustained Staufen1 expression in postnatal skeletal muscle causes a myopathy characterized by significant morphological and functional deficits. These deficits are accompanied by a marked increase in the expression of several atrophy-associated genes and by the negative regulation of PI3K/AKT signaling. We also uncovered that Staufen1 mediates PTEN expression through indirect transcriptional and direct post-transcriptional events thereby providing the first evidence for Staufen1-regulated PTEN expression. Collectively, our data demonstrate that Staufen1 is a novel atrophy-associated gene, and highlight its potential as a biomarker and therapeutic target for neuromuscular disorders and conditions.
Collapse
Affiliation(s)
- Tara E Crawford Parks
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Emma Bondy-Chorney
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jean-Marc Renaud
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jocelyn Côté
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
36
|
Mizgier ML, Cataldo LR, Gutierrez J, Santos JL, Casas M, Llanos P, Contreras-Ferrat AE, Moro C, Bouzakri K, Galgani JE. Effect of Human Myotubes-Derived Media on Glucose-Stimulated Insulin Secretion. J Diabetes Res 2017; 2017:1328573. [PMID: 28286777 PMCID: PMC5329672 DOI: 10.1155/2017/1328573] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 01/01/2017] [Accepted: 01/17/2017] [Indexed: 12/11/2022] Open
Abstract
Fasting to postprandial transition requires a tight adjustment of insulin secretion to its demand, so tissue (e.g., skeletal muscle) glucose supply is assured while hypo-/hyperglycemia are prevented. High muscle glucose disposal after meals is pivotal for adapting to increased glycemia and might drive insulin secretion through muscle-released factors (e.g., myokines). We hypothesized that insulin influences myokine secretion and then increases glucose-stimulated insulin secretion (GSIS). In conditioned media from human myotubes incubated with/without insulin (100 nmol/L) for 24 h, myokines were qualitatively and quantitatively characterized using an antibody-based array and ELISA-based technology, respectively. C57BL6/J mice islets and Wistar rat beta cells were incubated for 24 h with control and conditioned media from noninsulin- and insulin-treated myotubes prior to GSIS determination. Conditioned media from insulin-treated versus nontreated myotubes had higher RANTES but lower IL6, IL8, and MCP1 concentration. Qualitative analyses revealed that conditioned media from noninsulin- and insulin-treated myotubes expressed 32 and 23 out of 80 myokines, respectively. Islets incubated with conditioned media from noninsulin-treated myotubes had higher GSIS versus control islets (p < 0.05). Meanwhile, conditioned media from insulin-treated myotubes did not influence GSIS. In beta cells, GSIS was similar across conditions. In conclusion, factors being present in noninsulin-stimulated muscle cell-derived media appear to influence GSIS in mice islets.
Collapse
Affiliation(s)
- Maria L. Mizgier
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis R. Cataldo
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Gutierrez
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José L. Santos
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariana Casas
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Paola Llanos
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Ariel E. Contreras-Ferrat
- Exercise Science Laboratory, School of Kinesiology, Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Cedric Moro
- INSERM UMR1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université Paul Sabatier, Toulouse, France
| | - Karim Bouzakri
- Departement de Génétique et Développement, CMU, Université de Genève, Genève, Switzerland
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Jose E. Galgani
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- UDA-Ciencias de la Salud, Carrera de Nutrición y Dietética, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Jose E. Galgani:
| |
Collapse
|
37
|
Galgani JE, Gómez C, Mizgier ML, Gutierrez J, Santos JL, Olmos P, Mari A. Assessment of the Role of Metabolic Determinants on the Relationship between Insulin Sensitivity and Secretion. PLoS One 2016; 11:e0168352. [PMID: 28002466 PMCID: PMC5176173 DOI: 10.1371/journal.pone.0168352] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 11/30/2016] [Indexed: 12/21/2022] Open
Abstract
Background Insulin secretion correlates inversely with insulin sensitivity, which may suggest the existence of a crosstalk between peripheral organs and pancreas. Such interaction might be mediated through glucose oxidation that may drive the release of circulating factors with action on insulin secretion. Aim To evaluate the association between whole-body carbohydrate oxidation and circulating factors with insulin secretion to consecutive oral glucose loading in non-diabetic individuals. Methods Carbohydrate oxidation was measured after an overnight fast and for 6 hours after two 3-h apart 75-g oral glucose tolerance tests (OGTT) in 53 participants (24/29 males/females; 34±9 y; 27±4 kg/m2). Insulin secretion was estimated by deconvolution of serum C-peptide concentration, β cell function by mathematical modelling and insulin sensitivity from an OGTT. Circulating lactate, free-fatty acids (FFA) and candidate chemokines were assessed before and after OGTT. The effect of recombinant RANTES (regulated on activation, normal T cell expressed and secreted) and IL8 (interleukin 8) on insulin secretion from isolated mice islets was also measured. Results Carbohydrate oxidation assessed over the 6-h period did not relate with insulin secretion (r = -0.11; p = 0.45) or β cell function indexes. Circulating lactate and FFA showed no association with 6-h insulin secretion. Circulating chemokines concentration increased upon oral glucose stimulation. Insulin secretion associated with plasma IL6 (r = 0.35; p<0.05), RANTES (r = 0.30; p<0.05) and IL8 (r = 0.41; p<0.05) determined at 60 min OGTT. IL8 was independently associated with in vivo insulin secretion; however, it did not affect in vitro insulin secretion. Conclusion Whole-body carbohydrate oxidation appears to have no influence on insulin secretion or putative circulating mediators. IL8 may be a potential factor influencing insulin secretion.
Collapse
Affiliation(s)
- Jose E. Galgani
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- UDA-Ciencias de la Salud, Carrera de Nutrición y Dietética, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- * E-mail:
| | - Carmen Gómez
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Maria L. Mizgier
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Gutierrez
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jose L. Santos
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Olmos
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Andrea Mari
- Istituto di Neuroscienze, Consiglio Nazionale delle Ricerche, Padova, Italy
| |
Collapse
|
38
|
Reduced expression of MyHC slow isoform in rat soleus during unloading is accompanied by alterations of endogenous inhibitors of calcineurin/NFAT signaling pathway. J Muscle Res Cell Motil 2015; 37:7-16. [PMID: 26589960 DOI: 10.1007/s10974-015-9428-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/15/2015] [Indexed: 10/22/2022]
Abstract
Under muscle disuse conditions decrease of expression of MyHC of slow type, and sometimes of type IIa, as well as upregulation of expression of IIb and IId/x isoforms were observed. Through dephosphorylation and entry of NFAT molecules to the nucleus calcineurin/NFATc1 signaling pathway promotes upregulation of the slow MyHC expression. We supposed that downregulation of calcineurin pathway took place during unloading. The study was aimed to analyze the states of the myonuclear NFAT inhibitors calsarcin I (CSI) and calsarcin II (CSII) (also referred to as myozenin II and I) and GSK3β in rat soleus during hindlimb suspension (HS). Male Wistar rats were subjected to 3, 7 and 14 day of HS. We found that after 3 days of HS the content of CSII mRNA twofold increased in soleus as compared to the controls. This level was increased by more than fivefold (as compared to control) after 2 weeks of HS. The increase of CSII mRNA expression may be explained as the mechanism of stabilization of fast phenotype. We found that from the 3 day till 14 day of HS the content of MuRF-1 and MuRF-2 in the nuclear fraction fourfold to fivefold increased in HS soleus. We supposed that nuclear import of the MuRFs allows to promote CSII expression during unloading. We also observed the decline of the phosphorylated GSK3β content in the nuclear extract of the soleus tissue. Thus decline of slow MyHC expression characteristic for the unloading conditions is accompanied with the increased expression and activation of the factors known to prevent NFAT accumulation in the myonuclei.
Collapse
|
39
|
Du Bois P, Pablo Tortola C, Lodka D, Kny M, Schmidt F, Song K, Schmidt S, Bassel-Duby R, Olson EN, Fielitz J. Angiotensin II Induces Skeletal Muscle Atrophy by Activating TFEB-Mediated MuRF1 Expression. Circ Res 2015; 117:424-36. [PMID: 26137861 DOI: 10.1161/circresaha.114.305393] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 07/02/2015] [Indexed: 11/16/2022]
Abstract
RATIONALE Skeletal muscle wasting with accompanying cachexia is a life threatening complication in congestive heart failure. The molecular mechanisms are imperfectly understood, although an activated renin-angiotensin aldosterone system has been implicated. Angiotensin (Ang) II induces skeletal muscle atrophy in part by increased muscle-enriched E3 ubiquitin ligase muscle RING-finger-1 (MuRF1) expression, which may involve protein kinase D1 (PKD1). OBJECTIVE To elucidate the molecular mechanism of Ang II-induced skeletal muscle wasting. METHODS AND RESULTS A cDNA expression screen identified the lysosomal hydrolase-coordinating transcription factor EB (TFEB) as novel regulator of the human MuRF1 promoter. TFEB played a key role in regulating Ang II-induced skeletal muscle atrophy by transcriptional control of MuRF1 via conserved E-box elements. Inhibiting TFEB with small interfering RNA prevented Ang II-induced MuRF1 expression and atrophy. The histone deacetylase-5 (HDAC5), which was directly bound to and colocalized with TFEB, inhibited TFEB-induced MuRF1 expression. The inhibition of TFEB by HDAC5 was reversed by PKD1, which was associated with HDAC5 and mediated its nuclear export. Mice lacking PKD1 in skeletal myocytes were resistant to Ang II-induced muscle wasting. CONCLUSION We propose that elevated Ang II serum concentrations, as occur in patients with congestive heart failure, could activate the PKD1/HDAC5/TFEB/MuRF1 pathway to induce skeletal muscle wasting.
Collapse
Affiliation(s)
- Philipp Du Bois
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Cristina Pablo Tortola
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Doerte Lodka
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Melanie Kny
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Franziska Schmidt
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Kunhua Song
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Sibylle Schmidt
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Rhonda Bassel-Duby
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Eric N Olson
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Jens Fielitz
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.).
| |
Collapse
|
40
|
Abstract
Muscle RING finger 1 (MuRF1) and muscle atrophy F-box (MAFbx)/atrogin-1 were identified more than 10 years ago as two muscle-specific E3 ubiquitin ligases that are increased transcriptionally in skeletal muscle under atrophy-inducing conditions, making them excellent markers of muscle atrophy. In the past 10 years much has been published about MuRF1 and MAFbx with respect to their mRNA expression patterns under atrophy-inducing conditions, their transcriptional regulation, and their putative substrates. However, much remains to be learned about the physiological role of both genes in the regulation of mass and other cellular functions in striated muscle. Although both MuRF1 and MAFbx are enriched in skeletal, cardiac, and smooth muscle, this review will focus on the current understanding of MuRF1 and MAFbx in skeletal muscle, highlighting the critical questions that remain to be answered.
Collapse
Affiliation(s)
- Sue C Bodine
- Departments of Neurobiology, Physiology, and Behavior and Physiology and Membrane Biology, University of California Davis, Davis, California; and Northern California Veterans Affairs Health Systems, Mather, California
| | - Leslie M Baehr
- Membrane Biology, University of California Davis, Davis, California; and
| |
Collapse
|
41
|
Stefanetti RJ, Lamon S, Wallace M, Vendelbo MH, Russell AP, Vissing K. Regulation of ubiquitin proteasome pathway molecular markers in response to endurance and resistance exercise and training. Pflugers Arch 2014; 467:1523-1537. [DOI: 10.1007/s00424-014-1587-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 07/02/2014] [Accepted: 07/24/2014] [Indexed: 12/30/2022]
|
42
|
Mizgier ML, Casas M, Contreras-Ferrat A, Llanos P, Galgani JE. Potential role of skeletal muscle glucose metabolism on the regulation of insulin secretion. Obes Rev 2014; 15:587-97. [PMID: 24618283 DOI: 10.1111/obr.12166] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/31/2014] [Accepted: 02/09/2014] [Indexed: 01/03/2023]
Abstract
Pancreatic beta cells sense glucose flux and release as much insulin as required in order to maintain glycaemia within a narrow range. Insulin secretion is regulated by many factors including glucose, incretins, and sympathetic and parasympathetic tones among other physiological factors. To identify the mechanisms linking obesity-related insulin resistance with impaired insulin secretion represents a central challenge. Recently, it has been argued that a crosstalk between skeletal muscle and the pancreas may regulate insulin secretion. Considering that skeletal muscle is the largest organ in non-obese subjects and a major site of insulin- and exercise-stimulated glucose disposal, it appears plausible that muscle might interact with the pancreas and modulate insulin secretion for appropriate peripheral intracellular glucose utilization. There is growing evidence that muscle can secrete so-called myokines that can have auto/para/endocrine actions. Although it is unclear in which direction they act, interleukin-6 seems to be a possible muscle-derived candidate protein mediating such inter-organ communication. We herein review some of the putative skeletal muscle-derived factors mediating this interaction. In addition, the evidence coming from in vitro, animal and human studies that support such inter-organ crosstalk is thoroughly discussed.
Collapse
Affiliation(s)
- M L Mizgier
- Departmento de Nutrición, Diabetes y Metabolismo, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | |
Collapse
|
43
|
Wu CL, Cornwell EW, Jackman RW, Kandarian SC. NF-κB but not FoxO sites in the MuRF1 promoter are required for transcriptional activation in disuse muscle atrophy. Am J Physiol Cell Physiol 2014; 306:C762-7. [PMID: 24553183 DOI: 10.1152/ajpcell.00361.2013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The muscle-specific ring finger protein 1 (MuRF1) gene is required for most types of skeletal muscle atrophy yet we have little understanding of its transcriptional regulation. The purpose of this study is to identify whether NF-κB and/or FoxO response elements in the MuRF1 promoter are required for MuRF1 gene activation during skeletal muscle atrophy due to the removal of hindlimb weight bearing ("unloading"). Both NF-κB -dependent and FoxO-dependent luciferase reporter activities were significantly increased at 5 days of unloading. Using a 4.4-kb MuRF1 promoter reporter construct, a fourfold increase in reporter (i.e., luciferase) activity was found in rat soleus muscles after 5 days of hindlimb unloading. This activation was abolished by mutagenesis of either of the two distal putative NF-κB sites or all three putative NF-κB sites but not by mutagenesis of all four putative FoxO sites. This work provides the first direct evidence that NF-κB sites, but not FoxO sites, are required for MuRF1 promoter activation in muscle disuse atrophy in vivo.
Collapse
Affiliation(s)
- Chia-Ling Wu
- Department of Health Sciences, Boston University, Boston, Massachusetts
| | | | | | | |
Collapse
|
44
|
Stefanetti RJ, Zacharewicz E, Della Gatta P, Garnham A, Russell AP, Lamon S. Ageing has no effect on the regulation of the ubiquitin proteasome-related genes and proteins following resistance exercise. Front Physiol 2014; 5:30. [PMID: 24550841 PMCID: PMC3907707 DOI: 10.3389/fphys.2014.00030] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 01/14/2014] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle atrophy is a critical component of the ageing process. Age-related muscle wasting is due to disrupted muscle protein turnover, a process mediated in part by the ubiquitin proteasome pathway (UPP). Additionally, older subjects have been observed to have an attenuated anabolic response, at both the molecular and physiological levels, following a single-bout of resistance exercise (RE). We investigated the expression levels of the UPP-related genes and proteins involved in muscle protein degradation in 10 older (60–75 years) vs. 10 younger (18–30 years) healthy male subjects at basal as well as 2 h after a single-bout of RE. MURF1, atrogin-1 and FBXO40, their substrate targets PKM2, myogenin, MYOD, MHC and EIF3F as well as MURF1 and atrogin-1 transcriptional regulators FOXO1 and FOXO3 gene and/or protein expression levels were measured via real time PCR and western blotting, respectively. At basal, no age-related difference was observed in the gene/protein levels of atrogin-1, MURF1, myogenin, MYOD and FOXO1/3. However, a decrease in FBXO40 mRNA and protein levels was observed in older subjects, while PKM2 protein was increased. In response to RE, MURF1, atrogin-1 and FBXO40 mRNA were upregulated in both the younger and older subjects, with changes observed in protein levels. In conclusion, UPP-related gene/protein expression is comparably regulated in healthy young and old male subjects at basal and following RE. These findings suggest that UPP signaling plays a limited role in the process of age-related muscle wasting. Future studies are required to investigate additional proteolytic mechanisms in conjunction with skeletal muscle protein breakdown (MPB) measurements following RE in older vs. younger subjects.
Collapse
Affiliation(s)
- Renae J Stefanetti
- Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| | - Evelyn Zacharewicz
- Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| | - Paul Della Gatta
- Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| | - Andrew Garnham
- Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| | - Aaron P Russell
- Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| | - Séverine Lamon
- Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| |
Collapse
|
45
|
Stefanetti RJ, Lamon S, Rahbek SK, Farup J, Zacharewicz E, Wallace MA, Vendelbo MH, Russell AP, Vissing K. Influence of divergent exercise contraction mode and whey protein supplementation on atrogin-1, MuRF1, and FOXO1/3A in human skeletal muscle. J Appl Physiol (1985) 2014; 116:1491-502. [PMID: 24458747 DOI: 10.1152/japplphysiol.00136.2013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Knowledge from human exercise studies on regulators of muscle atrophy is lacking, but it is important to understand the underlying mechanisms influencing skeletal muscle protein turnover and net protein gain. This study examined the regulation of muscle atrophy-related factors, including atrogin-1 and MuRF1, their upstream transcription factors FOXO1 and FOXO3A and the atrogin-1 substrate eIF3-f, in response to unilateral isolated eccentric (ECC) vs. concentric (CONC) exercise and training. Exercise was performed with whey protein hydrolysate (WPH) or isocaloric carbohydrate (CHO) supplementation. Twenty-four subjects were divided into WPH and CHO groups and completed both single-bout exercise and 12 wk of training. Single-bout ECC exercise decreased atrogin-1 and FOXO3A mRNA compared with basal and CONC exercise, while MuRF1 mRNA was upregulated compared with basal. ECC exercise downregulated FOXO1 and phospho-FOXO1 protein compared with basal, and phospho-FOXO3A was downregulated compared with CONC. CONC single-bout exercise mediated a greater increase in MuRF1 mRNA and increased FOXO1 mRNA compared with basal and ECC. CONC exercise downregulated FOXO1, FOXO3A, and eIF3-f protein compared with basal. Following training, an increase in basal phospho-FOXO1 was observed. While WPH supplementation with ECC and CONC training further increased muscle hypertrophy, it did not have an additional effect on mRNA or protein levels of the targets measured. In conclusion, atrogin-1, MuRF1, FOXO1/3A, and eIF3-f mRNA, and protein levels, are differentially regulated by exercise contraction mode but not WPH supplementation combined with hypertrophy-inducing training. This highlights the complexity in understanding the differing roles these factors play in healthy muscle adaptation to exercise.
Collapse
Affiliation(s)
- Renae J Stefanetti
- Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Séverine Lamon
- Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Stine K Rahbek
- Section of Sport Science, Department of Public Health, Aarhus University, Aarhus, Denmark; and
| | - Jean Farup
- Section of Sport Science, Department of Public Health, Aarhus University, Aarhus, Denmark; and
| | - Evelyn Zacharewicz
- Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Marita A Wallace
- Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Mikkel H Vendelbo
- Department of Internal Medicine and Endocrinology, Aarhus University Hospital, Aarhus, Denmark
| | - Aaron P Russell
- Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Kristian Vissing
- Section of Sport Science, Department of Public Health, Aarhus University, Aarhus, Denmark; and
| |
Collapse
|
46
|
Kuo T, Harris CA, Wang JC. Metabolic functions of glucocorticoid receptor in skeletal muscle. Mol Cell Endocrinol 2013; 380:79-88. [PMID: 23523565 PMCID: PMC4893778 DOI: 10.1016/j.mce.2013.03.003] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 03/01/2013] [Accepted: 03/03/2013] [Indexed: 12/12/2022]
Abstract
Glucocorticoids (GCs) exert key metabolic influences on skeletal muscle. GCs increase protein degradation and decrease protein synthesis. The released amino acids are mobilized from skeletal muscle to liver, where they serve as substrates for hepatic gluconeogenesis. This metabolic response is critical for mammals' survival under stressful conditions, such as fasting and starvation. GCs suppress insulin-stimulated glucose uptake and utilization and glycogen synthesis, and play a permissive role for catecholamine-induced glycogenolysis, thus preserving the level of circulating glucose, the major energy source for the brain. However, chronic or excess exposure of GCs can induce muscle atrophy and insulin resistance. GCs convey their signal mainly through the intracellular glucocorticoid receptor (GR). While GR can act through different mechanisms, one of its major actions is to regulate the transcription of its primary target genes through genomic glucocorticoid response elements (GREs) by directly binding to DNA or tethering onto other DNA-binding transcription factors. These GR primary targets trigger physiological and pathological responses of GCs. Much progress has been made to understand how GCs regulate protein and glucose metabolism. In this review, we will discuss how GR primary target genes confer metabolic functions of GCs, and the mechanisms governing the transcriptional regulation of these targets. Comprehending these processes not only contributes to the fundamental understanding of mammalian physiology, but also will provide invaluable insight for improved GC therapeutics.
Collapse
Affiliation(s)
- Taiyi Kuo
- Department of Nutritional Science & Toxicology, University of California at Berkeley, Berkeley, CA 94720, United States
- Graduate Program of Endocrinology, University of California at Berkeley, Berkeley, CA 94720, United States
| | - Charles A. Harris
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, United States
- Department of Medicine, University of California at San Francisco, San Francisco, CA 94143, United States
| | - Jen-Chywan Wang
- Department of Nutritional Science & Toxicology, University of California at Berkeley, Berkeley, CA 94720, United States
- Graduate Program of Endocrinology, University of California at Berkeley, Berkeley, CA 94720, United States
- Corresponding author. Address: Department of Nutritional Science and Toxicology, 315 Morgan Hall, University of California at Berkeley, Berkeley, CA 94720-3104, United States. Tel.: +1 510 643 1039. (J.-C. Wang)
| |
Collapse
|
47
|
Furlow JD, Watson ML, Waddell DS, Neff ES, Baehr LM, Ross AP, Bodine SC. Altered gene expression patterns in muscle ring finger 1 null mice during denervation- and dexamethasone-induced muscle atrophy. Physiol Genomics 2013; 45:1168-85. [PMID: 24130153 DOI: 10.1152/physiolgenomics.00022.2013] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Muscle atrophy can result from inactivity or unloading on one hand or the induction of a catabolic state on the other. Muscle-specific ring finger 1 (MuRF1), a member of the tripartite motif family of E3 ubiquitin ligases, is an essential mediator of multiple conditions inducing muscle atrophy. While most studies have focused on the role of MuRF1 in protein degradation, the protein may have other roles in regulating skeletal muscle mass and metabolism. We therefore systematically evaluated the effect of MuRF1 on gene expression during denervation and dexamethasone-induced atrophy. We find that the lack of MuRF1 leads to few differences in control animals, but there were several significant differences in specific sets of genes upon denervation- and dexamethasone-induced atrophy. For example, during denervation, MuRF1 knockout mice showed delayed repression of metabolic and structural genes and blunted induction of genes associated with the neuromuscular junction. In the latter case, this pattern correlates with blunted HDAC4 and myogenin upregulation. Lack of MuRF1 caused fewer changes in the dexamethasone-induced atrophy program, but certain genes involved in fat metabolism and intracellular signaling were affected. Our results demonstrate a new role for MuRF1 in influencing gene expression in two important models of muscle atrophy.
Collapse
Affiliation(s)
- J David Furlow
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California; and
| | | | | | | | | | | | | |
Collapse
|
48
|
Castillero E, Alamdari N, Lecker SH, Hasselgren PO. Suppression of atrogin-1 and MuRF1 prevents dexamethasone-induced atrophy of cultured myotubes. Metabolism 2013; 62:1495-502. [PMID: 23866982 DOI: 10.1016/j.metabol.2013.05.018] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 05/28/2013] [Accepted: 05/29/2013] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The mechanistic role of the ubiquitin ligases atrogin-1 and MuRF1 in glucocorticoid-induced muscle wasting is not fully understood. Here, we tested the hypothesis that glucocorticoid-induced muscle atrophy is at least in part linked to atrogin-1 and MuRF1 expression and that the ubiquitin ligases are regulated by compensatory mechanisms. METHODS The expression of atrogin-1 and MuRF1 was suppressed individually or in combination in cultured L6 myotubes by using siRNA technique. Myotubes were treated with dexamethasone followed by determination of mRNA and protein levels for atrogin-1 and MuRF1, protein synthesis and degradation rates, and myotube morphology. RESULTS Suppression of atrogin-1 resulted in increased expression of MuRF1 and vice versa, suggesting that the ubiquitin ligases are regulated by compensatory mechanisms. Simultaneous suppression of atrogin-1 and MuRF1 resulted in myotube hypertrophy, mainly reflecting stimulated protein synthesis, and prevented dexamethasone-induced myotube atrophy, mainly reflecting inhibited protein degradation. CONCLUSIONS The results provide evidence for a link between upregulated atrogin-1 and MuRF1 expression and glucocorticoid-induced muscle atrophy. The study also suggests that atrogin-1 and MuRF1 levels are regulated by compensatory mechanisms and that inhibition of both ubiquitin ligases may be needed to prevent glucocorticoid-induced muscle proteolysis and atrophy.
Collapse
Affiliation(s)
- Estibaliz Castillero
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
49
|
Rudolf R, Bogomolovas J, Strack S, Choi KR, Khan MM, Wagner A, Brohm K, Hanashima A, Gasch A, Labeit D, Labeit S. Regulation of nicotinic acetylcholine receptor turnover by MuRF1 connects muscle activity to endo/lysosomal and atrophy pathways. AGE (DORDRECHT, NETHERLANDS) 2013; 35:1663-1674. [PMID: 22956146 PMCID: PMC3776120 DOI: 10.1007/s11357-012-9468-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 08/20/2012] [Indexed: 06/01/2023]
Abstract
Muscle atrophy is a process of muscle wasting induced under a series of catabolic stress conditions, such as denervation, disuse, cancer cachexia, heart and renal failure, AIDS, and aging. Neuromuscular junctions (NMJs), the synapses between motor neurons and muscle fibers undergo major changes in atrophying muscles, ranging from mild morphological alterations to complete disintegration. In this study, we hypothesized that remodeling of NMJs and muscle atrophy could be linked together. To test this, we examined if a major atrophy-promoting E3 ubiquitin ligase, MuRF1, is involved in the maintenance of NMJs. Immunofluorescence revealed that MuRF1 is highly enriched close to the NMJ. Affinity precipitation and in vivo imaging showed that MuRF1 interacts in endocytic structures with both, acetylcholine receptor, the primary postsynaptic protein of the NMJ, as well as with Bif-1, an autophagy- and endocytosis-regulating factor. In vivo imaging, radio labeling, and weighing approaches demonstrated that metabolic destabilization of acetylcholine receptors and muscle atrophy induced by denervation were significantly rescued in MuRF1-KO animals. Notably, interaction with Bif-1, and the rescue of AChR lifetime and muscle atrophy were specific to MuRF1 but not MuRF2. Our data demonstrate an involvement of MuRF1 in membrane protein-turnover, including the degradation of AChRs at the NMJ under atrophying conditions where MuRF1 also interacts and associates with Bif-1.
Collapse
Affiliation(s)
- Rüdiger Rudolf
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
- Institute of Molecular and Cell Biology, University of Applied Sciences Mannheim, Windeckstrasse 110, 68163 Mannheim, Germany
- Institute of Medical Technology, University of Heidelberg and University of Applied Sciences Mannheim, Paul-Wittsack-Strasse 10, 68163 Mannheim, Germany
| | - Julius Bogomolovas
- Department for Integrative Pathophysiology, Universitätsmedizin Mannheim, Theodor-Kutzer-Ufer, 68167 Mannheim, Germany
| | - Siegfried Strack
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Kyeong-Rok Choi
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Muzamil Majid Khan
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Anika Wagner
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Kathrin Brohm
- Department for Integrative Pathophysiology, Universitätsmedizin Mannheim, Theodor-Kutzer-Ufer, 68167 Mannheim, Germany
| | - Akira Hanashima
- Department for Integrative Pathophysiology, Universitätsmedizin Mannheim, Theodor-Kutzer-Ufer, 68167 Mannheim, Germany
| | - Alexander Gasch
- Department for Integrative Pathophysiology, Universitätsmedizin Mannheim, Theodor-Kutzer-Ufer, 68167 Mannheim, Germany
| | - Dittmar Labeit
- Department for Integrative Pathophysiology, Universitätsmedizin Mannheim, Theodor-Kutzer-Ufer, 68167 Mannheim, Germany
| | - Siegfried Labeit
- Department for Integrative Pathophysiology, Universitätsmedizin Mannheim, Theodor-Kutzer-Ufer, 68167 Mannheim, Germany
| |
Collapse
|
50
|
BMP signaling controls muscle mass. Nat Genet 2013; 45:1309-18. [DOI: 10.1038/ng.2772] [Citation(s) in RCA: 305] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 09/03/2013] [Indexed: 12/14/2022]
|