1
|
Abstract
Myostatin (GDF-8) was discovered 25 years ago as a new transforming growth factor-β family member that acts as a master regulator of skeletal muscle mass. Myostatin is made by skeletal myofibers, circulates in the blood, and acts back on myofibers to limit growth. Myostatin appears to have all of the salient properties of a chalone, which is a term proposed over a half century ago to describe hypothetical circulating, tissue-specific growth inhibitors that control tissue size. The elucidation of the molecular, cellular, and physiological mechanisms underlying myostatin activity suggests that myostatin functions as a negative feedback regulator of muscle mass and raises the question as to whether this type of chalone mechanism is unique to skeletal muscle or whether it also operates in other tissues.
Collapse
Affiliation(s)
- Se-Jin Lee
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, Connecticut, USA.,The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA;
| |
Collapse
|
2
|
Mishra R, Jha R, Mishra B, Kim YS. Maternal immunization against myostatin suppresses post-hatch chicken growth. PLoS One 2022; 17:e0275753. [PMID: 36201511 PMCID: PMC9536644 DOI: 10.1371/journal.pone.0275753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/22/2022] [Indexed: 11/07/2022] Open
Abstract
Myostatin (MSTN) is a negative regulator of skeletal muscle growth, thus it was hypothesized that immunization of hens against MSTN would enhance post-hatch growth and muscle mass via suppression of MSTN activity by anti-MSTN IgY in fertilized eggs. This study investigated the effects of immunization of hens against chicken MSTN (chMSTN) or a MSTN fragment (Myo2) on the growth and muscle mass of offspring. In Experiment 1, hens mixed with roosters were divided into two groups and hens in the Control and chMSTN groups were immunized with 0 and 0.5 mg of chMSTN, respectively. In Experiment 2, hens in the chMSTN group were divided into chMSTN and Myo2 groups while the Control group remained the same. The Control and chMSTN groups were immunized in the same way as Experiment 1. The Myo2 group was immunized against MSTN peptide fragment (Myo2) conjugated to KLH. Eggs collected from each group were incubated, and chicks were reared to examine growth and carcass parameters. ELISA showed the production of IgYs against chMSTN and Myo2 and the presence of these antibodies in egg yolk. IgY from the chMSTN and Myo2 groups showed binding affinity to chMSTN, Myo2, and commercial MSTN in Western blot analysis but did not show MSTN-inhibitory capacity in a reporter gene assay. In Experiment 1, no difference was observed in the body weight and carcass parameters of offspring between the Control and chMSTN groups. In Experiment 2, the body weight of chicks from the Myo2 group was significantly lower than that of the Control or chMSTN groups. The dressing percentage and breast muscle mass of the chMSTN and Myo2 groups were significantly lower than those of the Control group, and the breast muscle mass of Myo2 was significantly lower than that of the chMSTN. In summary, in contrast to our hypothesis, maternal immunization of hens did not increase but decreased the body weight and muscle mass of offspring.
Collapse
Affiliation(s)
- Rajeev Mishra
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Rajesh Jha
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Birendra Mishra
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Yong Soo Kim
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
- * E-mail:
| |
Collapse
|
3
|
Kern-Matschilles S, Gar C, Wanger L, Haschka SJ, Potzel AL, Hesse N, Then C, Seissler J, Lechner A. Association of Serum Myostatin with Body Weight, Visceral Fat Volume, and High Sensitivity C-Reactive Protein But Not With Muscle Mass and Physical Fitness in Premenopausal Women. Exp Clin Endocrinol Diabetes 2021; 130:393-399. [PMID: 34407549 DOI: 10.1055/a-1500-4605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND The myokine myostatin regulates muscle mass and has been linked to insulin resistance and metabolic syndrome. However, data on its role in humans is still limited. We, therefore, investigated the associations of serum myostatin with muscle mass, physical fitness, and components of the metabolic syndrome in a cohort of premenopausal women. METHODS We undertook a cross-sectional analysis of 233 women from the monocenter study PPSDiab, conducted in Munich, Germany. Participants had recently completed a pregnancy with or without gestational diabetes. Our analysis included medical history, anthropometrics, oral glucose tolerance testing, laboratory chemistry, cardiopulmonary exercise testing, and magnetic resonance imaging (n=142) of visceral fat volume, left quadriceps muscle mass, and muscle fat content. Serum myostatin was quantified by a competitive enzyme-linked immunosorbent assay. RESULTS We observed positive correlations of serum myostatin with body mass index (ρ=0.235; p=0.0003), body fat percentage (ρ=0.166; p=0.011), waist circumference (ρ=0.206; p=0.002), intraabdominal fat volume (ρ=0.182; p=0.030) and high-sensitivity C-reactive protein (ρ=0.175; p=0.008). These correlations were reproduced in linear regression analyses with adjustment for age and time after delivery. We saw no correlations with muscle mass, physical fitness, insulin sensitivity, triglycerides, HDL cholesterol, and blood pressure. CONCLUSIONS Our observation of elevated serum myostatin in women with a higher body fat percentage, visceral obesity, and elevated c-reactive protein suggests that this myokine contributes to the altered muscle-adipose tissue crosstalk in metabolic syndrome. Elevated myostatin may advance this pathophysiologic process and could also impair the efficacy of exercise interventions. Further mechanistic studies, therefore, seem warranted.
Collapse
Affiliation(s)
- Stefanie Kern-Matschilles
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| | - Christina Gar
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| | - Lorena Wanger
- Klinik und Poliklinik für Radiologie, LMU Klinikum, München, Germany
| | - Stefanie J Haschka
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| | - Anne L Potzel
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| | - Nina Hesse
- Klinik und Poliklinik für Radiologie, LMU Klinikum, München, Germany
| | - Cornelia Then
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| | - Jochen Seissler
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| | - Andreas Lechner
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| |
Collapse
|
4
|
Vaughan D, Mitchell R, Kretz O, Chambers D, Lalowski M, Amthor H, Ritvos O, Pasternack A, Matsakas A, Vaiyapuri S, Huber TB, Denecke B, Mukherjee A, Widera D, Patel K. A muscle growth-promoting treatment based on the attenuation of activin/myostatin signalling results in long-term testicular abnormalities. Dis Model Mech 2021; 14:dmm.047555. [PMID: 33408083 PMCID: PMC7903914 DOI: 10.1242/dmm.047555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022] Open
Abstract
Activin/myostatin signalling acts to induce skeletal muscle atrophy in adult mammals by inhibiting protein synthesis as well as promoting protein and organelle turnover. Numerous strategies have been successfully developed to attenuate the signalling properties of these molecules, which result in augmenting muscle growth. However, these molecules, in particular activin, play major roles in tissue homeostasis in numerous organs of the mammalian body. We have recently shown that although the attenuation of activin/myostatin results in robust muscle growth, it also has a detrimental impact on the testis. Here, we aimed to discover the long-term consequences of a brief period of exposure to muscle growth-promoting molecules in the testis. We demonstrate that muscle hypertrophy promoted by a soluble activin type IIB ligand trap (sActRIIB) is a short-lived phenomenon. In stark contrast, short-term treatment with sActRIIB results in immediate impact on the testis, which persists after the sessions of the intervention. Gene array analysis identified an expansion in aberrant gene expression over time in the testis, initiated by a brief exposure to muscle growth-promoting molecules. The impact on the testis results in decreased organ size as well as quantitative and qualitative impact on sperm. Finally, we have used a drug-repurposing strategy to exploit the gene expression data to identify a compound - N 6-methyladenosine - that may protect the testis from the impact of the muscle growth-promoting regime. This work indicates the potential long-term harmful effects of strategies aimed at promoting muscle growth by attenuating activin/myostatin signalling. Furthermore, we have identified a molecule that could, in the future, be used to overcome the detrimental impact of sActRIIB treatment on the testis.
Collapse
Affiliation(s)
- Danielle Vaughan
- School of Biological Sciences, University of Reading, Reading NW1 0TU, UK
| | - Robert Mitchell
- School of Biological Sciences, University of Reading, Reading NW1 0TU, UK
| | - Oliver Kretz
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - David Chambers
- Functional Genomics, King's College London, London SE1 1UL, UK
| | - Maciej Lalowski
- Department of Biochemistry and Developmental Biology, HiLIFE, Meilahti Clinical Proteomics Core Facility, University of Helsinki, Helsinki 00014, Finland
| | - Helge Amthor
- Versailles Saint-Quentin-en-Yvelines University, INSERM U1179, LIA BAHN CSM, Montigny-le-Bretonneux 78180, France
| | - Olli Ritvos
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki 00014, Finland
| | - Arja Pasternack
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki 00014, Finland
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, Hull HU6 7RX, UK
| | | | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | | | | | - Darius Widera
- School of Pharmacy, University of Reading, Reading RG6 6UB, UK
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading NW1 0TU, UK
| |
Collapse
|
5
|
Bagheri R, Ashtary-Larky D, Elliott BT, Willoughby DS, Kargarfard M, Alipour M, Lamuchi-Deli N, Kooti W, Asbaghi O, Wong A. The effects of gradual vs. rapid weight loss on serum concentrations of myokines and body composition in overweight and obese females. Arch Physiol Biochem 2021; 129:821-828. [PMID: 33502906 DOI: 10.1080/13813455.2021.1874020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Context: Research has shown the modulations of Follistatin (FST) and Myostatin (MST) following weight loss.Objective: We evaluated the effects of gradual weight loss (GWL) and rapid weight loss (RWL) on serum MST, FST, and body composition in overweight and obese females.Materials and methods: Thirty-six overweight and obese females successfully completed the study interventions: GWL (n = 18) or RWL (n = 18). Serum MST and FST concentrations, as well as anthropometric measurements, were collected at baseline and at the conclusion of each weight loss intervention.Results: MST concentration significantly (p < .05) decreased in the GWL; while FST concentration, body fat percentage and skeletal muscle mass significantly declined in both conditions. The loss in skeletal muscle mass was significantly greater in RWL relative to GWL.Discussion and conclusion: GWL was more effective than RWL in preserving skeletal muscle mass in overweight and obese females. Moreover, GWL leads to declines in MST concentrations.
Collapse
Affiliation(s)
- Reza Bagheri
- Department of Exercise Physiology, University of Isfahan, Isfahan, Iran
| | - Damoon Ashtary-Larky
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bradley T Elliott
- Translational Physiology Research Group, School of Life Sciences, University of Westminster, London, UK
| | - Darryn S Willoughby
- School of Exercise and Sport Science, University of Mary Hardin-Baylor, Belton, TX, USA
| | - Mehdi Kargarfard
- Department of Exercise Physiology, University of Isfahan, Isfahan, Iran
| | - Meysam Alipour
- Alimentary Tract Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nasrin Lamuchi-Deli
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Wesam Kooti
- Lung Diseases & Allergy Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Omid Asbaghi
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Alexei Wong
- Department of Health and Human Performance, Marymount University, Arlington, United States
| |
Collapse
|
6
|
Kramerova I, Marinov M, Owens J, Lee SJ, Becerra D, Spencer MJ. Myostatin inhibition promotes fast fibre hypertrophy but causes loss of AMP-activated protein kinase signalling and poor exercise tolerance in a model of limb-girdle muscular dystrophy R1/2A. J Physiol 2020; 598:3927-3939. [PMID: 33460149 DOI: 10.1113/jp279943] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/08/2020] [Indexed: 05/31/2024] Open
Abstract
KEY POINTS Limb-girdle muscular dystrophy R1 (LGMD R1) is caused by mutations in the CAPN3 gene and is characterized by progressive muscle loss, impaired mitochondrial function and reductions in the slow oxidative gene expression programme. Myostatin is a negative regulator of muscle growth, and its inhibition improves the phenotype in several muscle wasting disorders. The effect of genetic and pharmacological inhibition of myostatin signalling on the disease phenotype in a mouse model of LGMD R1 (CAPN3 knockout mouse-C3KO) was studied. Inhibition of myostatin signalling in C3KO muscles resulted in significant muscle hypertrophy; however, there were no improvements in muscle strength and exacerbation of exercise intolerance concomitant with further reduction of muscle oxidative capacity was observed. Inhibition of myostatin signalling is unlikely to be a valid therapeutic strategy for LGMD R1. ABSTRACT Limb-girdle muscular dystrophy R1 (LGMD R1) is caused by mutations in the CAPN3 gene and is characterized by progressive muscle loss, impaired mitochondrial function and reductions in the slow oxidative gene expression programme. There are currently no therapies available to patients. We sought to determine if induction of muscle growth, through myostatin inhibition, represents a viable therapeutic strategy for this disease. Myostatin is a negative regulator of muscle growth, and its inhibition improves the phenotype in several muscle wasting disorders. However, the effect of myostatin depends on the genetic and pathophysiological context and may not be efficacious in all contexts. We found that genetic inhibition of myostatin through overexpression of follistatin (an endogenous inhibitor of myostatin) in our LGMD R1 model (C3KO) resulted in 1.5- to 2-fold increase of muscle mass for the majority of limb muscles. However, muscle strength was not improved and exercise intolerance was exacerbated. Pharmacological inhibition of myostatin, using an anti-myostatin antibody, resulted in statistically significant increases in muscle mass; however, functional testing did not reveal changes in muscle strength nor endurance in treated C3KO mice. Histochemical and biochemical evaluation of follistatin overexpressing mice revealed a reduction in the percentage of oxidative fibres and decreased activation of AMP-activated protein kinase signalling in transgenics compared to C3KO muscles. Our data suggest that muscle hypertrophy, induced by myostatin inhibition, leads to loss of oxidative capacity, which further compromises metabolically impaired C3KO muscles and thus is unlikely to be a valid strategy for treatment of LGMD R1.
Collapse
Affiliation(s)
- Irina Kramerova
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Masha Marinov
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - Se-Jin Lee
- The Jackson Laboratory and University of Connecticut School of Medicine, Farmington, CT, USA
| | - Diana Becerra
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Melissa J Spencer
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
7
|
Eilers W, Chambers D, Cleasby M, Foster K. Local myostatin inhibition improves skeletal muscle glucose uptake in insulin-resistant high-fat diet-fed mice. Am J Physiol Endocrinol Metab 2020; 319:E163-E174. [PMID: 32459523 DOI: 10.1152/ajpendo.00185.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Myostatin inhibition is thought to improve whole body insulin sensitivity and mitigate the development of insulin resistance in models of obesity. However, although myostatin is known to be a major regulator of skeletal muscle mass, the direct effects of myostatin inhibition in muscle on glucose uptake and the mechanisms that may underlie this are still unclear. We investigated the effect of local myostatin inhibition by adeno-associated virus-mediated overexpression of the myostatin propeptide on insulin-stimulated skeletal muscle glucose disposal in chow-fed or high fat diet-fed mice and evaluated the molecular pathways that might mediate this. We found that myostatin inhibition improved glucose disposal in obese high fat diet-fed mice alongside the induction of muscle hypertrophy but did not have an impact in chow-fed mice. This improvement was not associated with greater glucose transporter or peroxisome proliferator-activated receptor-γ coactivator-1α expression or 5' AMP-activated protein kinase activation as previously suggested. Instead, transcriptomic analysis suggested that the improvement in glucose disposal was associated with significant enrichment in genes involved in fatty acid metabolism and translation of mitochondrial genes. Thus, myostatin inhibition improves muscle insulin-stimulated glucose disposal in obese high fat diet-fed mice independent of muscle hypertrophy, potentially involving previously unidentified pathways.
Collapse
Affiliation(s)
- Wouter Eilers
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - David Chambers
- Wolfson Centre for Age-Related Diseases, King's College, University of London, London, United Kingdom
| | - Mark Cleasby
- Royal Veterinary College, University of London, London, United Kingdom
| | - Keith Foster
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|
8
|
Zhang L, Duan Y, Guo Q, Wang W, Li F. A selectively suppressing amino acid transporter: Sodium-coupled neutral amino acid transporter 2 inhibits cell growth and mammalian target of rapamycin complex 1 pathway in skeletal muscle cells. ACTA ACUST UNITED AC 2020; 6:513-520. [PMID: 33364468 PMCID: PMC7750797 DOI: 10.1016/j.aninu.2020.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 12/18/2022]
Abstract
Sodium-coupled neutral amino acid transporter 2 (SNAT2), also known as solute carrier family 38 member 2 (SLC38A2), is expressed in the skeletal muscle. Our research previously indicated that SNAT2 mRNA expression level in the skeletal muscle was modulated by genotype and dietary protein. The aim of this study was to investigate the key role of the amino acid transporter SNAT2 in muscle cell growth, differentiation, and related signaling pathways via SNAT2 suppression using the inhibitor α-methylaminoisobutyric acid (MeAIB). The results showed that SNAT2 suppression down-regulated both the mRNA and protein expression levels of SNAT2 in C2C12 cells, inhibited cell viability and differentiation of the cell, and regulated the cell distribution in G0/G1 and S phases (P < 0.05). Meanwhile, most of the intercellular amino acid content of the cells after MeAIB co-culturing was significantly lower (P < 0.05). Furthermore, the mRNA expression levels of system L amino acid transporter 1 (LAT1), silent information regulator 1, and peroxisome proliferator-activated receptor-gamma co-activator 1 alpha, as well as the protein expression levels of amino acid transporters LAT1 and vacuolar protein sorting 34, were all down-regulated. The phosphorylated protein expression levels of mammalian target of rapamycin (mTOR), regulatory-associated protein of mTOR, 4E binding protein 1, and ribosomal protein S6 kinase 1 after MeAIB treatment were also significantly down-regulated (P < 0.05), which could contribute to the importance of SNAT2 in amino acid transportation and skeletal muscle cell sensing. In conclusion, SNAT2 suppression inhibited C2C12 cell growth and differentiation, as well as the availability of free amino acids. Although the mTOR complex 1 signaling pathway was found to be involved, its response to different nutrients requires further study.
Collapse
Affiliation(s)
- Lingyu Zhang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan 410125, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan 410125, China
| | - Qiuping Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan 410125, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Wenlong Wang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan 410125, China.,Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University, Changsha, 410018, China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan 410125, China
| |
Collapse
|
9
|
Zhou H, Meng J, Malerba A, Catapano F, Sintusek P, Jarmin S, Feng L, Lu-Nguyen N, Sun L, Mariot V, Dumonceaux J, Morgan JE, Gissen P, Dickson G, Muntoni F. Myostatin inhibition in combination with antisense oligonucleotide therapy improves outcomes in spinal muscular atrophy. J Cachexia Sarcopenia Muscle 2020; 11:768-782. [PMID: 32031328 PMCID: PMC7296258 DOI: 10.1002/jcsm.12542] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 12/10/2019] [Accepted: 12/19/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is caused by genetic defects in the survival motor neuron 1 (SMN1) gene that lead to SMN deficiency. Different SMN-restoring therapies substantially prolong survival and function in transgenic mice of SMA. However, these therapies do not entirely prevent muscle atrophy and restore function completely. To further improve the outcome, we explored the potential of a combinatorial therapy by modulating SMN production and muscle-enhancing approach as a novel therapeutic strategy for SMA. METHODS The experiments were performed in a mouse model of severe SMA. A previously reported 25-mer morpholino antisense oligomer PMO25 was used to restore SMN expression. The adeno-associated virus-mediated expression of myostatin propeptide was used to block the myostatin pathway. Newborn SMA mice were treated with a single subcutaneous injection of 40 μg/g (therapeutic dose) or 10 μg/g (low-dose) PMO25 on its own or together with systemic delivery of a single dose of adeno-associated virus-mediated expression of myostatin propeptide. The multiple effects of myostatin inhibition on survival, skeletal muscle phenotype, motor function, neuromuscular junction maturation, and proprioceptive afferences were evaluated. RESULTS We show that myostatin inhibition acts synergistically with SMN-restoring antisense therapy in SMA mice treated with the higher therapeutic dose PMO25 (40 μg/g), by increasing not only body weight (21% increase in male mice at Day 40), muscle mass (38% increase), and fibre size (35% increase in tibialis anterior muscle in 3 month female SMA mice), but also motor function and physical performance as measured in hanging wire test (two-fold increase in time score) and treadmill exercise test (two-fold increase in running distance). In SMA mice treated with low-dose PMO25 (10 μg/g), the early application of myostatin inhibition prolongs survival (40% increase), improves neuromuscular junction maturation (50% increase) and innervation (30% increase), and increases both the size of sensory neurons in dorsal root ganglia (60% increase) and the preservation of proprioceptive synapses in the spinal cord (30% increase). CONCLUSIONS These data suggest that myostatin inhibition, in addition to the well-known effect on muscle mass, can also positively influence the sensory neural circuits that may enhance motor neurons function. While the availability of the antisense drug Spinraza for SMA and other SMN-enhancing therapies has provided unprecedented improvement in SMA patients, there are still unmet needs in these patients. Our study provides further rationale for considering myostatin inhibitors as a therapeutic intervention in SMA patients, in combination with SMN-restoring drugs.
Collapse
Affiliation(s)
- Haiyan Zhou
- Genetics and Genomic Medicine Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Jinhong Meng
- The Dubowitz Neuromuscular Centre, Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Alberto Malerba
- Centres of Gene and Cell Therapy and Biomedical Sciences, School of Biological Sciences, Royal Holloway, University of London, Egham, UK
| | - Francesco Catapano
- The Dubowitz Neuromuscular Centre, Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Palittiya Sintusek
- The Dubowitz Neuromuscular Centre, Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK.,Department of Paediatrics, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| | - Susan Jarmin
- Centres of Gene and Cell Therapy and Biomedical Sciences, School of Biological Sciences, Royal Holloway, University of London, Egham, UK
| | - Lucy Feng
- The Dubowitz Neuromuscular Centre, Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Ngoc Lu-Nguyen
- Centres of Gene and Cell Therapy and Biomedical Sciences, School of Biological Sciences, Royal Holloway, University of London, Egham, UK
| | - Lianwen Sun
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, China
| | - Virginie Mariot
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Julie Dumonceaux
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Jennifer E Morgan
- The Dubowitz Neuromuscular Centre, Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Paul Gissen
- Genetics and Genomic Medicine Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - George Dickson
- Centres of Gene and Cell Therapy and Biomedical Sciences, School of Biological Sciences, Royal Holloway, University of London, Egham, UK
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| |
Collapse
|
10
|
Vaughan D, Ritvos O, Mitchell R, Kretz O, Lalowski M, Amthor H, Chambers D, Matsakas A, Pasternack A, Collins-Hooper H, Ballesteros R, Huber TB, Denecke B, Widera D, Mukherjee A, Patel K. Inhibition of Activin/Myostatin signalling induces skeletal muscle hypertrophy but impairs mouse testicular development. Eur J Transl Myol 2020; 30:8737. [PMID: 32499882 PMCID: PMC7254437 DOI: 10.4081/ejtm.2019.8737] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 12/05/2019] [Indexed: 01/22/2023] Open
Abstract
Numerous approaches are being developed to promote post-natal muscle growth based on attenuating Myostatin/Activin signalling for clinical uses such as the treatment neuromuscular diseases, cancer cachexia and sarcopenia. However there have been concerns about the effects of inhibiting Activin on tissues other than skeletal muscle. We intraperitoneally injected mice with the Activin ligand trap, sActRIIB, in young, adult and a progeric mouse model. Treatment at any stage in the life of the mouse rapidly increased muscle mass. However at all stages of life the treatment decreased the weights of the testis. Not only were the testis smaller, but they contained fewer sperm compared to untreated mice. We found that the hypertrophic muscle phenotype was lost after the cessation of sActRIIB treatment but abnormal testis phenotype persisted. In summary, attenuation of Myostatin/Activin signalling inhibited testis development. Future use of molecules based on a similar mode of action to promote muscle growth should be carefully profiled for adverse side-effects on the testis. However the effectiveness of sActRIIB as a modulator of Activin function provides a possible therapeutic strategy to alleviate testicular seminoma development.
Collapse
Affiliation(s)
| | - Olli Ritvos
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | | | - Oliver Kretz
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maciej Lalowski
- Department of Biochemistry and Developmental Biology, HiLIFE, Meilahti Clinical Proteomics Core Facility, University of Helsinki, Helsinki, Finland
| | - Helge Amthor
- Versailles Saint-Quentin-en-Yvelines University, INSERM U1179, LIA BAHN CSM, Montigny-le-Bretonneux 78180, France
| | | | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombosis & Metabolic Disease, Hull York Medical School, Hull, UK
| | - Arja Pasternack
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | | | | | - Tobias B Huber
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | - Ketan Patel
- School of Biological Sciences, University of Reading, UK
| |
Collapse
|
11
|
Effects of exosome-mediated delivery of myostatin propeptide on functional recovery of mdx mice. Biomaterials 2020; 236:119826. [DOI: 10.1016/j.biomaterials.2020.119826] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 12/30/2019] [Accepted: 01/25/2020] [Indexed: 01/08/2023]
|
12
|
Tavoian D, Arnold WD, Mort SC, de Lacalle S. Sex differences in body composition but not neuromuscular function following long-term, doxycycline-induced reduction in circulating levels of myostatin in mice. PLoS One 2019; 14:e0225283. [PMID: 31751423 PMCID: PMC6872155 DOI: 10.1371/journal.pone.0225283] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/31/2019] [Indexed: 02/05/2023] Open
Abstract
Age-related declines in muscle function result from changes in muscle structure and contractile properties, as well as from neural adaptations. Blocking myostatin to drive muscle growth is one potential therapeutic approach. While the effects of myostatin depletion on muscle characteristics are well established, we have very little understanding of its effects on the neural system. Here we assess the effects of long-term, post-developmental myostatin reduction on electrophysiological motor unit characteristics and body composition in aging mice. We used male (N = 21) and female (N = 26) mice containing a tetracycline-inducible system to delete the myostatin gene in skeletal muscle. Starting at 12 months of age, half of the mice were administered doxycycline (tetracycline) through their chow for one year. During that time we measured food intake, body composition, and hindlimb electromyographic responses. Doxycycline-induced myostatin reduction had no effect on motor unit properties for either sex, though significant age-dependent declines in motor unit number occurred in all mice. However, treatment with doxycycline induced different changes in body composition between sexes. All female mice increased in total, lean and fat mass, but doxycycline-treated female mice experienced a significantly larger increase in lean mass than controls. All male mice also increased total and lean mass, but administration of doxycycline had no effect. Additionally, doxycycline-treated male mice maintained their fat mass at baseline levels, while the control group experienced a significant increase from baseline and compared to the doxycycline treated group. Our results show that long-term administration of doxycycline results in body composition adaptations that are distinctive between male and female mice, and that the effects of myostatin reduction are most pronounced during the first three months of treatment. We also report that age-related changes in motor unit number are not offset by reduced myostatin levels, despite increased lean mass exhibited by female mice.
Collapse
Affiliation(s)
- Dallin Tavoian
- Program in Translational Biomedical Sciences, 1 Ohio University, Athens, OH, United States of America
| | - W. David Arnold
- Departments of Neurology, PM&R, and Neuroscience, and Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States of America
| | - Sophia C. Mort
- Program in Translational Biomedical Sciences, 1 Ohio University, Athens, OH, United States of America
| | - Sonsoles de Lacalle
- Sonsoles de Lacalle, Department of Biomedical Sciences,1 Ohio University, Athens, OH, United States of America
- * E-mail:
| |
Collapse
|
13
|
Elashry MI, Eldaey A, Glenske K, Matsakas A, Wenisch S, Arnhold S, Patel K. The effect of high-fat diet on the morphological properties of the forelimb musculature in hypertrophic myostatin null mice. J Anat 2019; 235:825-835. [PMID: 31198988 DOI: 10.1111/joa.13025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2019] [Indexed: 12/20/2022] Open
Abstract
Obesity is a worldwide nutritional disorder affecting body performance, including skeletal muscle. Inhibition of myostatin not only increases the muscle mass but also it reduces body fat accumulation. We examined the effect of high-fat diet on the phenotypic properties of forelimb muscles from myostatin null mice. Male wild-type and myostatin null mice were fed on either a normal diet or a high-fat diet (45% fat) for 10 weeks. Musculus triceps brachii Caput longum; M. triceps brachii Caput laterale; M. triceps brachii Caput mediale; M. extensor carpi ulnaris and M. flexor carpi ulnaris were processed for fiber type composition using immunohistochemistry and morphometric analysis. Although the muscle mass revealed no change under a high-fat diet, there were morphometric alterations in the absence of myostatin. We show that high-fat diet reduces the cross-sectional area of the fast (IIB and IIX) fibers in M. triceps brachii Caput longum and M. triceps brachii Caput laterale of both genotypes. In contrast, increases of fast fiber areas were observed in both M. extensor carpi ulnaris of wild-type and M. flexor carpi ulnaris of myostatin null mice. Meanwhile, a high-fat diet increased the area of the fast IIA fibers in wild-type mice; myostatin null mice display a muscle-dependent alteration in the area of the same fiber type. The combined high-fat diet and myostatin deletion shows no effect on the area of slow type I fibers. Although a high-fat diet causes a reduction in the area of the peripheral IIB fibers in both genotypes, only myostatin null mice show an increase in the area of the central IIB fibers. We provide evidence that a high-fat diet induces a muscle-dependent fast to slow myofiber shift in the absence of myostatin. The data suggest that the morphological alterations of muscle fibers under a combined high-fat diet and myostatin deletion reflect a functional adaptation of the muscle to utilize the high energy intake.
Collapse
Affiliation(s)
- Mohamed I Elashry
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, University of Mansoura, Mansoura, Egypt.,Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, Giessen, Germany
| | - Asmaa Eldaey
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, University of Mansoura, Mansoura, Egypt.,Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, Giessen, Germany
| | - Kristina Glenske
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, Giessen, Germany
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, Giessen, Germany
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, Giessen, Germany
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK
| |
Collapse
|
14
|
Kim JH, Kim JH, Sutikno LA, Lee SB, Jin DH, Hong YK, Kim YS, Jin HJ. Identification of the minimum region of flatfish myostatin propeptide (Pep45-65) for myostatin inhibition and its potential to enhance muscle growth and performance in animals. PLoS One 2019; 14:e0215298. [PMID: 30998775 PMCID: PMC6472743 DOI: 10.1371/journal.pone.0215298] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 03/30/2019] [Indexed: 12/31/2022] Open
Abstract
Myostatin (MSTN) negatively regulates skeletal muscle growth, and its activity is inhibited by the binding of MSTN propeptide (MSTNpro), the N-terminal domain of proMSTN that is proteolytically cleaved from the proMSTN. Partial sequences from the N-terminal side of MSTNpro have shown to be sufficient to inhibit MSTN activity. In this study, to determine the minimum size of flatfish MSTNpro for MSTN inhibition, various truncated forms of flatfish MSTNpro with N-terminal maltose binding protein (MBP) fusion were expressed in E. coli and purified. MSTNpro regions consisting of residues 45–68, -69, and -70 with MBP fusion suppressed MSTN activity with a potency comparable to that of full-sequence flatfish MSTNpro in a pGL3-(CAGA)12-luciferase reporter assay. Even though the MSTN-inhibitory potency was about 1,000-fold lower, the flatfish MSTNpro region containing residues 45–65 (MBP-Pro45-65) showed MSTN-inhibitory capacity but not the MBP-Pro45-64, indicating that the region 45–65 is the minimum domain required for MSTN binding and suppression of its activity. To examine the in vivo effect of MBP-fused, truncated flatfish MSTNpro, MBP-Pro45-70-His6 (20 mg/kg body wt) was subcutaneously injected 5 times for 14 days in mice. Body wt gain and bone mass were not affected by the administration. Grip strength and swimming time were significantly enhanced at 7 d after the administration. At 14 d, the effect on grip strength disappeared, and the extent of the effect on swimming time significantly diminished. The presence of antibody against MBP-Pro45-70-His6 was observed at both 7 and 14 d after the administration with the titer value at 14 d being much greater than that at 7 d, suggesting that antibodies against MBP-Pro45-70-His6 neutralized the MSTN-inhibitory effect of MBP-Pro45-70-His6. We, thus, examined the MSTN-inhibitory capacity and in vivo effect of flatfish MSTNpro region 45–65 peptide (Pep45-65-NH2), which was predicted to have no immunogenicity in silico analysis. Pep45-65-NH2 suppressed MSTN activity with a potency similar to that of MBP-Pro45-65 but did not suppress GDF11, or activin A. Pep45-65-NH2 blocked MSTN-induced Smad2 phosphorylation in HepG2 cells. The administration of Pep45-65 (20 mg/kg body wt, 5 times for 2 weeks) increased the body wt gain with a greater gain at 14 d than at 7 d and muscle wt. Grip strength and swimming time were also significantly enhanced by the administration. Antibody titer against Pep45-65 was not detected. In conclusion, current results indicate that MSTN-inhibitory proteins with heterologous fusion partner may not be effective in suppressing MSTN activity in vivo due to an immune response against the proteins. Current results also show that the region of flatfish MSTNpro consisting of 45–65 (Pep45-65) can suppress mouse MSTN activity and increase muscle mass and function without invoking an immune response, implying that Pep45-65 would be a potential agent to enhance skeletal muscle growth and function in animals or to treat muscle atrophy caused by various clinical conditions.
Collapse
Affiliation(s)
- Jeong Hwan Kim
- Department of Marine Molecular Bioscience, Gangneung-Wonju National University, Gangneung-si, Ganwon-do, Korea
| | - Jeong Han Kim
- Department of Marine Molecular Bioscience, Gangneung-Wonju National University, Gangneung-si, Ganwon-do, Korea
| | | | - Sang Beum Lee
- Department of Human Nurtrition, Food and Animal Sciences, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Deuk-Hee Jin
- Department of Marine Molecular Bioscience, Gangneung-Wonju National University, Gangneung-si, Ganwon-do, Korea
| | - Yong-Ki Hong
- Department of Biotechnology, Pukyong National University, Namgu, Busan, Korea
| | - Yong Soo Kim
- Department of Human Nurtrition, Food and Animal Sciences, University of Hawaii, Honolulu, Hawaii, United States of America
- * E-mail: (YK); (HJ)
| | - Hyung-Joo Jin
- Department of Marine Molecular Bioscience, Gangneung-Wonju National University, Gangneung-si, Ganwon-do, Korea
- * E-mail: (YK); (HJ)
| |
Collapse
|
15
|
Recombinant porcine myostatin propeptide generated by the Pichia pastoris elevates myoblast growth and ameliorates high-fat diet-induced glucose intolerance. Res Vet Sci 2019; 124:200-211. [PMID: 30921567 DOI: 10.1016/j.rvsc.2019.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 02/12/2019] [Accepted: 03/12/2019] [Indexed: 01/07/2023]
Abstract
Myostatin (MSTN) was identified as a negative regulator of skeletal muscle growth. MSTN inhibition by myostatin propeptide (MSPP) increased skeletal muscle mass, myofiber growth and muscle force. Thus, this study was designed to produce wild-type porcine MSPP (WT-MSPP) and its mutated form (D75A-MSPP) in yeast Pichia pastoris and to investigate its potential enhancement of myoblast growth and differentiation. In an in vitro study, C2C12 myoblasts were treated with the purified WT-MSPP or D75A-MSPP (10 μg/mL) in either a regular culture medium or in a differentiation medium for 72 h. In an animal trial, post-weaning C57BL/6 mice fed with a high-fat diet (HFD) were administered WT-MSPP or D75A-MSPP for 6 weeks. The results showed that C2C12 myoblasts treated with the purified WT-MSPP or D75A-MSPP could dramatically promote cell proliferation. Both myoD and myogenin were significantly increased (p < .05) after WT-MSPP or D75A-MSPP treatment. D75A-MSPP was particularly more effective than WT-MSPP in promoting myotube formation (p < .05). The post-weaning mice treated with D75A-MSPP significantly increased both body and muscle weights compared with the mock and WT-MSPP groups (p < .05). Furthermore, the mice treatment with D75A-MSPP could prevent increased glucose injection from inducing glucose elevation. Our data indicated that a mutant-type MSPP (D75A-MSPP) was superior to WT-MSPP in effectively enhancing myofiber growth due to the highly resistant to proteolytic cleavage by the bone morphogenetic protein-1/tolloid (BMP-1/TLD) and thus has potential applications for clinical muscle wasting diseases or for increasing muscle mass in meat-producing animals.
Collapse
|
16
|
Scully D, Sfyri P, Verpoorten S, Papadopoulos P, Muñoz‐Turrillas MC, Mitchell R, Aburima A, Patel K, Gutiérrez L, Naseem KM, Matsakas A. Platelet releasate promotes skeletal myogenesis by increasing muscle stem cell commitment to differentiation and accelerates muscle regeneration following acute injury. Acta Physiol (Oxf) 2019; 225:e13207. [PMID: 30339324 DOI: 10.1111/apha.13207] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/27/2018] [Accepted: 10/14/2018] [Indexed: 12/11/2022]
Abstract
AIM The use of platelets as biomaterials has gained intense research interest. However, the mechanisms regarding platelet-mediated skeletal myogenesis remain to be established. The aim of this study was to determine the role of platelet releasate in skeletal myogenesis and muscle stem cell fate in vitro and ex vivo respectively. METHODS We analysed the effect of platelet releasate on proliferation and differentiation of C2C12 myoblasts by means of cell proliferation assays, immunohistochemistry, gene expression and cell bioenergetics. We expanded in vitro findings on single muscle fibres by determining the effect of platelet releasate on murine skeletal muscle stem cells using protein expression profiles for key myogenic regulatory factors. RESULTS TRAP6 and collagen used for releasate preparation had a more pronounced effect on myoblast proliferation vs thrombin and sonicated platelets (P < 0.05). In addition, platelet concentration positively correlated with myoblast proliferation. Platelet releasate increased myoblast and muscle stem cell proliferation in a dose-dependent manner, which was mitigated by VEGFR and PDGFR inhibition. Inhibition of VEGFR and PDGFR ablated MyoD expression on proliferating muscle stem cells, compromising their commitment to differentiation in muscle fibres (P < 0.001). Platelet releasate was detrimental to myoblast fusion and affected differentiation of myoblasts in a temporal manner. Most importantly, we show that platelet releasate promotes skeletal myogenesis through the PDGF/VEGF-Cyclin D1-MyoD-Scrib-Myogenin axis and accelerates skeletal muscle regeneration after acute injury. CONCLUSION This study provides novel mechanistic insights on the role of platelet releasate in skeletal myogenesis and set the physiological basis for exploiting platelets as biomaterials in regenerative medicine.
Collapse
Affiliation(s)
- David Scully
- Molecular Physiology Laboratory, Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School University of Hull Hull UK
| | - Peggy Sfyri
- Molecular Physiology Laboratory, Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School University of Hull Hull UK
| | - Sandrine Verpoorten
- Molecular Physiology Laboratory, Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School University of Hull Hull UK
| | - Petros Papadopoulos
- Department of Hematology, Instituto de Investigación Sanitaria San Carlos (IdISSC) Hospital Clínico San Carlos Madrid Spain
| | - María Carmen Muñoz‐Turrillas
- Centro Comunitario de Sangre y Tejidos de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA) Oviedo Spain
| | - Robert Mitchell
- School of Biological Sciences University of Reading Reading UK
| | - Ahmed Aburima
- Molecular Physiology Laboratory, Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School University of Hull Hull UK
| | - Ketan Patel
- School of Biological Sciences University of Reading Reading UK
| | - Laura Gutiérrez
- Department of Medicine Universidad de Oviedo and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA) Oviedo Spain
| | - Khalid M. Naseem
- Leeds Institute of Cardiovascular and Metabolic Medicine University of Leeds Leeds UK
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School University of Hull Hull UK
| |
Collapse
|
17
|
Past, Present, and Future Perspective of Targeting Myostatin and Related Signaling Pathways to Counteract Muscle Atrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:153-206. [DOI: 10.1007/978-981-13-1435-3_8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
18
|
Barbé C, Bray F, Gueugneau M, Devassine S, Lause P, Tokarski C, Rolando C, Thissen JP. Comparative Proteomic and Transcriptomic Analysis of Follistatin-Induced Skeletal Muscle Hypertrophy. J Proteome Res 2017; 16:3477-3490. [PMID: 28810121 DOI: 10.1021/acs.jproteome.7b00069] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Skeletal muscle, the most abundant body tissue, plays vital roles in locomotion and metabolism. Myostatin is a negative regulator of skeletal muscle mass. In addition to increasing muscle mass, Myostatin inhibition impacts muscle contractility and energy metabolism. To decipher the mechanisms of action of the Myostatin inhibitors, we used proteomic and transcriptomic approaches to investigate the changes induced in skeletal muscles of transgenic mice overexpressing Follistatin, a physiological Myostatin inhibitor. Our proteomic workflow included a fractionation step to identify weakly expressed proteins and a comparison of fast versus slow muscles. Functional annotation of altered proteins supports the phenotypic changes induced by Myostatin inhibition, including modifications in energy metabolism, fiber type, insulin and calcium signaling, as well as membrane repair and regeneration. Less than 10% of the differentially expressed proteins were found to be also regulated at the mRNA level but the Biological Process annotation, and the KEGG pathways analysis of transcriptomic results shows a great concordance with the proteomic data. Thus this study describes the most extensive omics analysis of muscle overexpressing Follistatin, providing molecular-level insights to explain the observed muscle phenotypic changes.
Collapse
Affiliation(s)
- Caroline Barbé
- Pole of Endocrinology, Diabetes and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain , 1200 Brussels, Belgium
| | - Fabrice Bray
- Miniaturisation pour la Synthèse, l'Analyse & la Protéomique (MSAP), CNRS, USR 3290, Université de Lille; Biochimie Structurale & Fonctionnelle des Assemblages Biomoléculaires, CNRS, FR 3688, FRABIO, Université de Lille and Institut Eugène-Michel Chevreul, CNRS, FR 2638, Université de Lille, 59000 Lille, France
| | - Marine Gueugneau
- Pole of Endocrinology, Diabetes and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain , 1200 Brussels, Belgium
| | - Stéphanie Devassine
- Miniaturisation pour la Synthèse, l'Analyse & la Protéomique (MSAP), CNRS, USR 3290, Université de Lille; Biochimie Structurale & Fonctionnelle des Assemblages Biomoléculaires, CNRS, FR 3688, FRABIO, Université de Lille and Institut Eugène-Michel Chevreul, CNRS, FR 2638, Université de Lille, 59000 Lille, France
| | - Pascale Lause
- Pole of Endocrinology, Diabetes and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain , 1200 Brussels, Belgium
| | - Caroline Tokarski
- Miniaturisation pour la Synthèse, l'Analyse & la Protéomique (MSAP), CNRS, USR 3290, Université de Lille; Biochimie Structurale & Fonctionnelle des Assemblages Biomoléculaires, CNRS, FR 3688, FRABIO, Université de Lille and Institut Eugène-Michel Chevreul, CNRS, FR 2638, Université de Lille, 59000 Lille, France
| | - Christian Rolando
- Miniaturisation pour la Synthèse, l'Analyse & la Protéomique (MSAP), CNRS, USR 3290, Université de Lille; Biochimie Structurale & Fonctionnelle des Assemblages Biomoléculaires, CNRS, FR 3688, FRABIO, Université de Lille and Institut Eugène-Michel Chevreul, CNRS, FR 2638, Université de Lille, 59000 Lille, France
| | - Jean-Paul Thissen
- Pole of Endocrinology, Diabetes and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain , 1200 Brussels, Belgium
| |
Collapse
|
19
|
Specific targeting of TGF-β family ligands demonstrates distinct roles in the regulation of muscle mass in health and disease. Proc Natl Acad Sci U S A 2017; 114:E5266-E5275. [PMID: 28607086 DOI: 10.1073/pnas.1620013114] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The transforming growth factor-β (TGF-β) network of ligands and intracellular signaling proteins is a subject of intense interest within the field of skeletal muscle biology. To define the relative contribution of endogenous TGF-β proteins to the negative regulation of muscle mass via their activation of the Smad2/3 signaling axis, we used local injection of adeno-associated viral vectors (AAVs) encoding ligand-specific antagonists into the tibialis anterior (TA) muscles of C57BL/6 mice. Eight weeks after AAV injection, inhibition of activin A and activin B signaling produced moderate (∼20%), but significant, increases in TA mass, indicating that endogenous activins repress muscle growth. Inhibiting myostatin induced a more profound increase in muscle mass (∼45%), demonstrating a more prominent role for this ligand as a negative regulator of adult muscle mass. Remarkably, codelivery of activin and myostatin inhibitors induced a synergistic response, resulting in muscle mass increasing by as much as 150%. Transcription and protein analysis indicated that this substantial hypertrophy was associated with both the complete inhibition of the Smad2/3 pathway and activation of the parallel bone morphogenetic protein (BMP)/Smad1/5 axis (recently identified as a positive regulator of muscle mass). Analyses indicated that hypertrophy was primarily driven by an increase in protein synthesis, but a reduction in ubiquitin-dependent protein degradation pathways was also observed. In models of muscular dystrophy and cancer cachexia, combined inhibition of activins and myostatin increased mass or prevented muscle wasting, respectively, highlighting the potential therapeutic advantages of specifically targeting multiple Smad2/3-activating ligands in skeletal muscle.
Collapse
|
20
|
Elashry MI, Matsakas A, Wenisch S, Arnhold S, Patel K. The effect of caloric restriction on the forelimb skeletal muscle fibers of the hypertrophic myostatin null mice. Acta Histochem 2017. [PMID: 28622884 DOI: 10.1016/j.acthis.2017.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Skeletal muscle mass loss has a broad impact on body performance and physical activity. Muscle wasting occurs due to genetic mutation as in muscular dystrophy, age-related muscle loss (sarcopenia) as well as in chronic wasting disorders as in cancer cachexia. Food restriction reduces muscle mass underpinned by increased muscle protein break down. However the influence of dietary restriction on the morphometry and phenotype of forelimb muscles in a genetically modified myostatin null mice are not fully characterized. The effect of a five week dietary limitation on five anatomically and structurally different forelimb muscles was examined. C57/BL6 wild type (Mstn+/+) and myostatin null (Mstn-/-) mice were either given a standard rodent normal daily diet ad libitum (ND) or 60% food restriction (FR) for a 5 week period. M. triceps brachii Caput laterale (T.lateral), M. triceps brachii Caput longum (T.long), M. triceps brachii Caput mediale (T.medial), M. extensor carpi ulnaris (ECU) and M. flexor carpi ulnaris (FCU) were dissected, weighted and processed for immunohistochemistry. Muscle mass, fibers cross sectional areas (CSA) and myosin heavy chain types IIB, IIX, IIA and type I were analyzed. We provide evidence that caloric restriction results in muscle specific weight reduction with the fast myofibers being more prone to atrophy. We show that slow fibers are less liable to dietary restriction induced muscle atrophy. The effect of dietary restriction was more pronounced in Mstn-/- muscles to implicate the oxidative fibers compared to Mstn+/+. Furthermore, peripherally located myofibers are more susceptible to dietary induced reduction compared to deep fibers. We additionally report that dietary restriction alters the glycolytic phenotype of the Mstn-/- into the oxidative form in a muscle dependent manner. In summary our study shows that calorie restriction alters muscle fiber profile of forelimb muscles of Myostatin null mice.
Collapse
Affiliation(s)
- Mohamed I Elashry
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, University of Mansoura, 35516, Egypt; Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, 35392, Germany.
| | | | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, 35392, Germany
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, 35392, Germany
| | - Ketan Patel
- School of Biological Sciences, Hopkins Building, Whiteknights, University of Reading, RG6 6UB, United Kingdom
| |
Collapse
|
21
|
Lee SB, Park SK, Kim YS. Maltose binding protein-fusion enhances the bioactivity of truncated forms of pig myostatin propeptide produced in E. coli. PLoS One 2017; 12:e0174956. [PMID: 28369115 PMCID: PMC5378391 DOI: 10.1371/journal.pone.0174956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/18/2017] [Indexed: 11/18/2022] Open
Abstract
Myostatin (MSTN) is a potent negative regulator of skeletal muscle growth. MSTN propeptide (MSTNpro) inhibits MSTN binding to its receptor through complex formation with MSTN, implying that MSTNpro can be a useful agent to improve skeletal muscle growth in meat-producing animals. Four different truncated forms of pig MSTNpro containing N-terminal maltose binding protein (MBP) as a fusion partner were expressed in E. coli, and purified by the combination of affinity chromatography and gel filtration. The MSTN-inhibitory capacities of these proteins were examined in an in vitro gene reporter assay. A MBP-fused, truncated MSTNpro containing residues 42-175 (MBP-Pro42-175) exhibited the same MSTN-inhibitory potency as the full sequence MSTNpro. Truncated MSTNpro proteins containing either residues 42-115 (MBP-Pro42-115) or 42-98 (MBP-Pro42-98) also exhibited MSTN-inhibitory capacity even though the potencies were significantly lower than that of full sequence MSTNpro. In pull-down assays, MBP-Pro42-175, MBP-Pro42-115, and MBP-Pro42-98 demonstrated their binding to MSTN. MBP was removed from the truncated MSTNpro proteins by incubation with factor Xa to examine the potential role of MBP on MSTN-inhibitory capacity of those proteins. Removal of MBP from MBP-Pro42-175 and MBP-Pro42-98 resulted in 20-fold decrease in MSTN-inhibitory capacity of Pro42-175 and abolition of MSTN-inhibitory capacity of Pro42-98, indicating that MBP as fusion partner enhanced the MSTN-inhibitory capacity of those truncated MSTNpro proteins. In summary, this study shows that MBP is a very useful fusion partner in enhancing MSTN-inhibitory potency of truncated forms of MSTNpro proteins, and MBP-fused pig MSTNpro consisting of amino acid residues 42-175 is sufficient to maintain the full MSTN-inhibitory capacity.
Collapse
Affiliation(s)
- Sang Beum Lee
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Gangwon-do, South Korea
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, United States of America
| | - Sung Kwon Park
- National Institute of Animal Science, RDA, Suwon, South Korea
| | - Yong Soo Kim
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, United States of America
- * E-mail:
| |
Collapse
|
22
|
Matsakas A, Prosdocimo DA, Mitchell R, Collins-Hooper H, Giallourou N, Swann JR, Potter P, Epting T, Jain MK, Patel K. Investigating mechanisms underpinning the detrimental impact of a high-fat diet in the developing and adult hypermuscular myostatin null mouse. Skelet Muscle 2015; 5:38. [PMID: 26644908 PMCID: PMC4671215 DOI: 10.1186/s13395-015-0063-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 10/23/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Obese adults are prone to develop metabolic and cardiovascular diseases. Furthermore, over-weight expectant mothers give birth to large babies who also have increased likelihood of developing metabolic and cardiovascular diseases. Fundamental advancements to better understand the pathophysiology of obesity are critical in the development of anti-obesity therapies not only for this but also future generations. Skeletal muscle plays a major role in fat metabolism and much work has focused in promoting this activity in order to control the development of obesity. Research has evaluated myostatin inhibition as a strategy to prevent the development of obesity and concluded in some cases that it offers a protective mechanism against a high-fat diet. METHODS Pregnant as well as virgin myostatin null mice and age matched wild type animals were raised on a high fat diet for up to 10 weeks. The effect of the diet was tested on skeletal muscle, liver and fat. Quantitate PCR, Western blotting, immunohistochemistry, in-vivo and ex-vivo muscle characterisation, metabonomic and lipidomic measurements were from the four major cohorts. RESULTS We hypothesised that myostatin inhibition should protect not only the mother but also its developing foetus from the detrimental effects of a high-fat diet. Unexpectedly, we found muscle development was attenuated in the foetus of myostatin null mice raised on a high-fat diet. We therefore re-examined the effect of the high-fat diet on adults and found myostatin null mice were more susceptible to diet-induced obesity through a mechanism involving impairment of inter-organ fat utilization. CONCLUSIONS Loss of myostatin alters fatty acid uptake and oxidation in skeletal muscle and liver. We show that abnormally high metabolic activity of fat in myostatin null mice is decreased by a high-fat diet resulting in excessive adipose deposition and lipotoxicity. Collectively, our genetic loss-of-function studies offer an explanation of the lean phenotype displayed by a host of animals lacking myostatin signalling.
Collapse
Affiliation(s)
- Antonios Matsakas
- Centre for Cardiovascular & Metabolic Research, Hull York Medical School, University of Hull, Hull, UK
| | - Domenick A Prosdocimo
- Case Cardiovascular Research Institute and Harrington Heart & Vascular Institute, Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, USA
| | - Robert Mitchell
- School of Biological Sciences, University of Reading, Reading, RG6 6UB UK
| | | | - Natasa Giallourou
- Department of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Reading, UK
| | - Jonathan R Swann
- Department of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Reading, UK
| | - Paul Potter
- Mammalian Genetics Unit, MRC Harwell, Oxford, UK
| | - Thomas Epting
- Institute for Clinical Chemistry and Laboratory Medicine, Universitat klinikum, Freiburg, Germany
| | - Mukesh K Jain
- Case Cardiovascular Research Institute and Harrington Heart & Vascular Institute, Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, USA
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, RG6 6UB UK.,Freiburg Institute for Advanced Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
23
|
Targeted mutations in myostatin by zinc-finger nucleases result in double-muscled phenotype in Meishan pigs. Sci Rep 2015; 5:14435. [PMID: 26400270 PMCID: PMC4585837 DOI: 10.1038/srep14435] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/28/2015] [Indexed: 12/20/2022] Open
Abstract
Myostatin (MSTN) is a dominant inhibitor of skeletal muscle development and growth. Mutations in MSTN gene can lead to muscle hypertrophy or double-muscled (DM) phenotype in cattle, sheep, dog and human. However, there has not been reported significant muscle phenotypes in pigs in association with MSTN mutations. Pigs are an important source of meat production, as well as serve as a preferred animal model for the studies of human disease. To study the impacts of MSTN mutations on skeletal muscle growth in pigs, we generated MSTN-mutant Meishan pigs with no marker gene via zinc finger nucleases (ZFN) technology. The MSTN-mutant pigs developed and grew normally, had increased muscle mass with decreased fat accumulation compared with wild type pigs, and homozygote MSTN mutant (MSTN−/−) pigs had apparent DM phenotype, and individual muscle mass increased by 100% over their wild-type controls (MSTN+/+) at eight months of age as a result of myofiber hyperplasia. Interestingly, 20% MSTN-mutant pigs had one extra thoracic vertebra. The MSTN-mutant pigs will not only offer a way of fast genetic improvement of lean meat for local fat-type indigenous pig breeds, but also serve as an important large animal model for biomedical studies of musculoskeletal formation, development and diseases.
Collapse
|
24
|
Development of novel activin-targeted therapeutics. Mol Ther 2014; 23:434-44. [PMID: 25399825 DOI: 10.1038/mt.2014.221] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 11/09/2014] [Indexed: 01/07/2023] Open
Abstract
Soluble activin type II receptors (ActRIIA/ActRIIB), via binding to diverse TGF-β proteins, can increase muscle and bone mass, correct anemia or protect against diet-induced obesity. While exciting, these multiple actions of soluble ActRIIA/IIB limit their therapeutic potential and highlight the need for new reagents that target specific ActRIIA/IIB ligands. Here, we modified the activin A and activin B prodomains, regions required for mature growth factor synthesis, to generate specific activin antagonists. Initially, the prodomains were fused to the Fc region of mouse IgG2A antibody and, subsequently, "fastener" residues (Lys(45), Tyr(96), His(97), and Ala(98); activin A numbering) that confer latency to other TGF-β proteins were incorporated. For the activin A prodomain, these modifications generated a reagent that potently (IC(50) 5 nmol/l) and specifically inhibited activin A signaling in vitro, and activin A-induced muscle wasting in vivo. Interestingly, the modified activin B prodomain inhibited both activin A and B signaling in vitro (IC(50) ~2 nmol/l) and in vivo, suggesting it could serve as a general activin antagonist. Importantly, unlike soluble ActRIIA/IIB, the modified prodomains did not inhibit myostatin or GDF-11 activity. To underscore the therapeutic utility of specifically antagonising activin signaling, we demonstrate that the modified activin prodomains promote significant increases in muscle mass.
Collapse
|
25
|
Cleasby ME, Jarmin S, Eilers W, Elashry M, Andersen DK, Dickson G, Foster K. Local overexpression of the myostatin propeptide increases glucose transporter expression and enhances skeletal muscle glucose disposal. Am J Physiol Endocrinol Metab 2014; 306:E814-23. [PMID: 24473441 PMCID: PMC3962614 DOI: 10.1152/ajpendo.00586.2013] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/28/2014] [Indexed: 12/20/2022]
Abstract
Insulin resistance (IR) in skeletal muscle is a prerequisite for type 2 diabetes and is often associated with obesity. IR also develops alongside muscle atrophy in older individuals in sarcopenic obesity. The molecular defects that underpin this syndrome are not well characterized, and there is no licensed treatment. Deletion of the transforming growth factor-β family member myostatin, or sequestration of the active peptide by overexpression of the myostatin propeptide/latency-associated peptide (ProMyo) results in both muscle hypertrophy and reduced obesity and IR. We aimed to establish whether local myostatin inhibition would have a paracrine/autocrine effect to enhance glucose disposal beyond that simply generated by increased muscle mass, and the mechanisms involved. We directly injected adeno-associated virus expressing ProMyo in right tibialis cranialis/extensor digitorum longus muscles of rats and saline in left muscles and compared the effects after 17 days. Both test muscles were increased in size (by 7 and 11%) and showed increased radiolabeled 2-deoxyglucose uptake (26 and 47%) and glycogen storage (28 and 41%) per unit mass during an intraperitoneal glucose tolerance test. This was likely mediated through increased membrane protein levels of GLUT1 (19% higher) and GLUT4 (63% higher). Interestingly, phosphorylation of phosphoinositol 3-kinase signaling intermediates and AMP-activated kinase was slightly decreased, possibly because of reduced expression of insulin-like growth factor-I in these muscles. Thus, myostatin inhibition has direct effects to enhance glucose disposal in muscle beyond that expected of hypertrophy alone, and this approach may offer potential for the therapy of IR syndromes.
Collapse
Affiliation(s)
- M. E. Cleasby
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, United Kingdom
| | - S. Jarmin
- School of Biological Sciences, Royal Holloway, University of London, Egham, Surrey, United Kingdom; and
| | - W. Eilers
- School of Biological Sciences, University of Reading, Reading, Berkshire, United Kingdom
| | - M. Elashry
- School of Biological Sciences, University of Reading, Reading, Berkshire, United Kingdom
| | - D. K. Andersen
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, United Kingdom
| | - G. Dickson
- School of Biological Sciences, Royal Holloway, University of London, Egham, Surrey, United Kingdom; and
| | - K. Foster
- School of Biological Sciences, University of Reading, Reading, Berkshire, United Kingdom
| |
Collapse
|
26
|
Collins-Hooper H, Sartori R, Macharia R, Visanuvimol K, Foster K, Matsakas A, Flasskamp H, Ray S, Dash PR, Sandri M, Patel K. Propeptide-Mediated Inhibition of Myostatin Increases Muscle Mass Through Inhibiting Proteolytic Pathways in Aged Mice. J Gerontol A Biol Sci Med Sci 2014; 69:1049-59. [DOI: 10.1093/gerona/glt170] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
27
|
High-level soluble expression of bioactive porcine myostatin propeptide in E. coli. Appl Microbiol Biotechnol 2013; 97:8517-27. [DOI: 10.1007/s00253-013-5134-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 07/12/2013] [Accepted: 07/15/2013] [Indexed: 01/10/2023]
|
28
|
Szláma G, Trexler M, Patthy L. Latent myostatin has significant activity and this activity is controlled more efficiently by WFIKKN1 than by WFIKKN2. FEBS J 2013; 280:3822-39. [PMID: 23829672 PMCID: PMC3906830 DOI: 10.1111/febs.12377] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 05/30/2013] [Accepted: 06/05/2013] [Indexed: 01/18/2023]
Abstract
Myostatin, a negative regulator of skeletal muscle growth, is produced from myostatin precursor by multiple steps of proteolytic processing. After cleavage by a furin-type protease, the propeptide and growth factor domains remain associated, forming a noncovalent complex, the latent myostatin complex. Mature myostatin is liberated from latent myostatin by bone morphogenetic protein 1/tolloid proteases. Here, we show that, in reporter assays, latent myostatin preparations have significant myostatin activity, as the noncovalent complex dissociates at an appreciable rate, and both mature and semilatent myostatin (a complex in which the dimeric growth factor domain interacts with only one molecule of myostatin propeptide) bind to myostatin receptor. The interaction of myostatin receptor with semilatent myostatin is efficiently blocked by WAP, Kazal, immunoglobulin, Kunitz and NTR domain-containing protein 1 or growth and differentiation factor-associated serum protein 2 (WFIKKN1), a large extracellular multidomain protein that binds both mature myostatin and myostatin propeptide [Kondás et al. (2008) J Biol Chem283, 23677–23684]. Interestingly, the paralogous protein WAP, Kazal, immunoglobulin, Kunitz and NTR domain-containing protein 2 or growth and differentiation factor-associated serum protein 1 (WFIKKN2) was less efficient than WFIKKN1 as an antagonist of the interactions of myostatin receptor with semilatent myostatin. Our studies have shown that this difference is attributable to the fact that only WFIKKN1 has affinity for the propeptide domain, and this interaction increases its potency in suppressing the receptor-binding activity of semilatent myostatin. As the interaction of WFIKKN1 with various forms of myostatin permits tighter control of myostatin activity until myostatin is liberated from latent myostatin by bone morphogenetic protein 1/tolloid proteases, WFIKKN1 may have greater potential as an antimyostatic agent than WFIKKN2.
Collapse
Affiliation(s)
- György Szláma
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | | | | |
Collapse
|
29
|
Monestier O, Brun C, Heu K, Passet B, Malhouroux M, Magnol L, Vilotte JL, Blanquet V. Ubiquitous Gasp1 overexpression in mice leads mainly to a hypermuscular phenotype. BMC Genomics 2012; 13:541. [PMID: 23046573 PMCID: PMC3575399 DOI: 10.1186/1471-2164-13-541] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 10/03/2012] [Indexed: 11/10/2022] Open
Abstract
Background Myostatin, a member of the TGFβ superfamily, is well known as a potent and specific negative regulator of muscle growth. Targeting the myostatin signalling pathway may offer promising therapeutic strategies for the treatment of muscle-wasting disorders. In the last decade, various myostatin-binding proteins have been identified to be able to inhibit myostatin activity. One of these is GASP1 (Growth and Differentiation Factor-Associated Serum Protein-1), a protein containing a follistatin domain as well as multiple domains associated with protease inhibitors. Despite in vitro data, remarkably little is known about in vivo functions of Gasp1. To further address the role of GASP1 during mouse development and in adulthood, we generated a gain-of-function transgenic mouse model that overexpresses Gasp1 under transcriptional control of the human cytomegalovirus immediate-early promoter/enhancer. Results Overexpression of Gasp1 led to an increase in muscle mass observed not before day 15 of postnatal life. The surGasp1 transgenic mice did not display any other gross abnormality. Histological and morphometric analysis of surGasp1 rectus femoris muscles revealed an increase in myofiber size without a corresponding increase in myofiber number. Fiber-type distribution was unaltered. Interestingly, we do not detect a change in total fat mass and lean mass. These results differ from those for myostatin knockout mice, transgenic mice overexpressing the myostatin propeptide or follistatin which exhibit both muscle hypertrophy and hyperplasia, and show minimal fat deposition. Conclusions Altogether, our data give new insight into the in vivo functions of Gasp1. As an extracellular regulatory factor in the myostatin signalling pathway, additional studies on GASP1 and its homolog GASP2 are required to elucidate the crosstalk between the different intrinsic inhibitors of the myostatin.
Collapse
Affiliation(s)
- Olivier Monestier
- INRA, UMR1061 Unité de Génétique Moléculaire Animale, Limoges, 87060, France
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Kim CH, Kim KH, Yoo YM. Melatonin-induced autophagy is associated with degradation of MyoD protein in C2C12 myoblast cells. J Pineal Res 2012; 53:289-97. [PMID: 22582971 DOI: 10.1111/j.1600-079x.2012.00998.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
MyoD is a muscle-specific transcriptional factor that acts as a master switch for skeletal muscle differentiation. This protein regulates myoblast proliferation and myogenic differentiation and is also a short-lived regulatory protein that is degraded by the ubiquitin system. However, the lysosomal pathway of MyoD protein degradation remains unknown. In this study, we sought to determine whether melatonin (1, 2mm)-induced autophagy causes the degradation of MyoD protein in C2C12 myoblast cells. Melatonin induced a significant increase in expression of the microtubule-associated protein 1 light chain 3 (LC3)-II and Beclin-1 proteins in a dose-dependent manner. Melatonin treatment also significantly increased p-ERK, Ras, and p-Akt expressions in a dose-dependent manner. However, Bax expression was high compared with the absence of melatonin treatment, and Bcl-2 expression was high in the 0.1-0.5mm melatonin treatments and low in the 1 and 2mm melatonin treatments. Under the same conditions, cytosolic MyoD protein was significantly decreased in a dose-dependent manner and completely eliminated by 36hr. This decrease in MyoD protein involved ubiquitin-mediated proteasomal activity with proteasome inhibitor MG132 or autophagy-dependent lysosomal degradation with lysosomal inhibitor bafilomycin A1 (Baf-A1). In the same condition, phosphorylation of the mammalian target of rapamycin, p-mTOR, and p-S6K expression with Baf-A1 or Baf-A1-plus melatonin treatment were significantly decreased compared with the levels after treatment with melatonin only. Together, these results suggest that melatonin (1, 2mm)-induced autophagy results in partial lysosomal degradation of MyoD protein in C2C12 myoblast cells.
Collapse
Affiliation(s)
- Chi Hyun Kim
- Department of Biomedical Engineering, College of Health Science, Yonsei University, Wonju, Gangwon-do, Korea
| | | | | |
Collapse
|
31
|
Jackson MF, Luong D, Vang DD, Garikipati DK, Stanton JB, Nelson OL, Rodgers BD. The aging myostatin null phenotype: reduced adiposity, cardiac hypertrophy, enhanced cardiac stress response, and sexual dimorphism. J Endocrinol 2012; 213:263-75. [PMID: 22431133 DOI: 10.1530/joe-11-0455] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The natural aging process results in the physiological decline of multiple tissues and organ systems. Changes commonly occur with middle age and include decreased skeletal muscle mass, bone mineral density, cardiac output, and insulin sensitivity, and increased adiposity, all of which can contribute to the onset of sarcopenia, osteoporosis, heart failure, or type 2 diabetes. Recent studies suggest that myostatin may influence many of these systems. We therefore sought to determine whether they are affected by aging, especially in 'middle-aged' Mstn-/- mice (12-20 months old (m.o.)). Although body weights were similar in wild-type (WT) and Mstn-/- mice, lean fat-free mass and skeletal muscles composed of predominantly type I, II, and mixed fibers were significantly heavier in Mstn-/- mice. These differences were accompanied by lower total adiposity, especially in female mice, white and brown fat pad weights, and adipocyte size. Hearts were heavier in Mstn-/- mice across a large age range (3-24 m.o.) and exhibited signs of dilated cardiomyopathy at rest, which include lower strain measurements compared with WT myocardium. However, Mstn-/- mice responded better to isoproterenol stress tests with greater increases in fractional shortening and ejection fraction-differences that were again more apparent in females and which are consistent with physiological cardiac hypertrophy. Spleens and kidneys were also smaller, although histologically normal, in Mstn-/- mice. These data together suggest that attenuating myostatin could potentially prevent or possibly treat pathological conditions that develop with age. Additional studies are nevertheless needed to definitively assess potential risks to cardiac function.
Collapse
Affiliation(s)
- Melissa F Jackson
- School of Molecular Biosciences, Department of Veterinary Microbiology and Pathology, 124 ASLB, Washington Center for Muscle Biology, Washington State University, Pullman, Washington 99164, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Elashry MI, Collins-Hooper H, Vaiyapuri S, Patel K. Characterisation of connective tissue from the hypertrophic skeletal muscle of myostatin null mice. J Anat 2012; 220:603-11. [PMID: 22463481 DOI: 10.1111/j.1469-7580.2012.01503.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Myostatin is a potent inhibitor of muscle development. Genetic deletion of myostatin in mice results in muscle mass increase, with muscles often weighing three times their normal values. Contracting muscle transfers tension to skeletal elements through an elaborate connective tissue network. Therefore, the connective tissue of skeletal muscle is an integral component of the contractile apparatus. Here we examine the connective tissue architecture in myostatin null muscle. We show that the hypertrophic muscle has decreased connective tissue content compared with wild-type muscle. Secondly, we show that the hypertrophic muscle fails to show the normal increase in muscle connective tissue content during ageing. Therefore, genetic deletion of myostatin results in an increase in contractile elements but a decrease in connective tissue content. We propose a model based on the contractile profile of muscle fibres that reconciles this apparent incompatible tissue composition phenotype.
Collapse
|
33
|
Wang Q, McPherron AC. Myostatin inhibition induces muscle fibre hypertrophy prior to satellite cell activation. J Physiol 2012; 590:2151-65. [PMID: 22393251 DOI: 10.1113/jphysiol.2011.226001] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Muscle fibres are multinucleated post-mitotic cells that can change dramatically in size during adulthood. It has been debated whether muscle fibre hypertrophy requires activation and fusion of muscle stem cells, the satellite cells. Myostatin (MSTN) is a negative regulator of skeletal muscle growth during development and in the adult, and MSTN inhibition is therefore a potential therapy for muscle wasting diseases, some of which are associated with a depletion of satellite cells. Conflicting results have been obtained in previous analyses of the role of MSTN on satellite cell quiescence. Here, we inhibited MSTN in adult mice with a soluble activin receptor type IIB and analysed the incorporation of new nuclei using 5-bromo-2-deoxyuridine (BrdU) labelling by isolating individual myofibres. We found that satellite cells are activated by MSTN inhibition. By varying the dose and time course for MSTN inhibition, however, we found that myofibre hypertrophy precedes the incorporation of new nuclei, and that the overall number of new nuclei is relatively low compared to the number of total myonuclei. These results reconcile some of the previous work obtained by other methods. In contrast with previous reports, we also found that Mstn null mice do not have increased satellite cell numbers during adulthood and are not resistant to sarcopaenia. Our results support a previously proposed model of hypertrophy in which hypertrophy can precede satellite cell activation. Studies of the metabolic and functional effects of postnatal MSTN inhibition are needed to determine the consequences of increasing the cytoplasm/myonuclear ratio after MSTN inhibition.
Collapse
Affiliation(s)
- Qian Wang
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
34
|
Kemaladewi DU, ‘t Hoen PA, ten Dijke P, van Ommen GJ, Hoogaars WM. TGF-β signaling in Duchenne muscular dystrophy. FUTURE NEUROLOGY 2012. [DOI: 10.2217/fnl.12.3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The TGF-β protein family consists of secreted multifunctional cytokines that control diverse processes, such as cell growth and differentiation. Aberrant expression and downstream signaling of these growth factors have been associated with multiple diseases, including muscle wasting disorders, such as Duchenne muscular dystrophy. In this review we discuss recent advances in understanding the role of TGF-β family members during normal skeletal muscle biology/regeneration and their role in muscle pathology, with a special focus on Duchenne muscular dystrophy. In addition, we will highlight progress in the development of potential therapeutics for Duchenne muscular dystrophy based on intervention of TGF-β signaling.
Collapse
Affiliation(s)
- Dwi U Kemaladewi
- Department of Human Genetics, Leiden University Medical Center, Postzone S4-P, PO Box 9600 2300RC Leiden, The Netherlands
- Department of Molecular & Cell Biology and Centre for Biomedical Genetics, Leiden University Medical Center, Postzone S4-P, PO Box 9600 2300RC Leiden, The Netherlands
| | - Peter A ‘t Hoen
- Department of Human Genetics, Leiden University Medical Center, Postzone S4-P, PO Box 9600 2300RC Leiden, The Netherlands
| | - Peter ten Dijke
- Department of Molecular & Cell Biology and Centre for Biomedical Genetics, Leiden University Medical Center, Postzone S4-P, PO Box 9600 2300RC Leiden, The Netherlands
| | - Gert Jan van Ommen
- Department of Human Genetics, Leiden University Medical Center, Postzone S4-P, PO Box 9600 2300RC Leiden, The Netherlands
| | - Willem M Hoogaars
- Department of Human Genetics, Leiden University Medical Center, Postzone S4-P, PO Box 9600 2300RC Leiden, The Netherlands
| |
Collapse
|
35
|
Myostatin regulates preadipocyte differentiation and lipid metabolism of adipocyte via ERK1/2. Cell Biol Int 2012; 35:1141-6. [PMID: 21510842 DOI: 10.1042/cbi20110112] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Myostatin is known as an inhibitor of muscle development, but its role in adipogenesis and lipid metabolism is still unclear, especially the underlying mechanisms. Here, we demonstrated that myostatin inhibited 3T3-L1 preadipocyte differentiation into adipocyte by suppressing C/EBPα (CCAAT/enhancer-binding protein α) and PPARγ (peroxisome-proliferator-activated receptor γ), also activated ERK1/2 (extracellular-signal-regulated kinase 1/2). Furthermore, myostatin enhanced the phosphorylation of HSL (hormone-sensitive lipase) and ACC (acetyl-CoA carboxylase) in fully differentiated adipocytes, as well as ERK1/2. Besides, we noted that myostatin markedly raised the levels of leptin and adiponectin release and mRNA expression during preadipocyte differentiation, but the levels were inhibited by myostatin treatments in fully differentiated adipocytes. These results suggested that myostatin suppressed 3T3-L1 preadipocyte differentiation and regulated lipid metabolism of mature adipocyte, in part, via activation of ERK1/2 signalling pathway.
Collapse
|
36
|
Allen DL, Hittel DS, McPherron AC. Expression and function of myostatin in obesity, diabetes, and exercise adaptation. Med Sci Sports Exerc 2012; 43:1828-35. [PMID: 21364474 DOI: 10.1249/mss.0b013e3182178bb4] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Myostatin is a member of the transforming growth factor-β/bone morphogenetic protein (TGF-β/BMP) superfamily of secreted factors that functions as a potent inhibitor of skeletal muscle growth. Moreover, considerable evidence has accumulated that myostatin also regulates metabolism and that its inhibition can significantly attenuate the progression of obesity and diabetes. Although at least part of these effects on metabolism can be attributable to myostatin's influence over skeletal muscle growth and therefore on the total volume of metabolically active lean body mass, there is mounting evidence that myostatin affects the growth and metabolic state of other tissues, including the adipose and the liver. In addition, recent work has explored the role of myostatin in substrate mobilization, uptake, and/or utilization of muscle independent of its effects on body composition. Finally, the effects of both endurance and resistance exercise on myostatin expression, as well as the potential role of myostatin in the beneficial metabolic adaptations occurring in response to exercise, have also begun to be delineated in greater detail. The purpose of this review was to summarize the work to date on the expression and function of myostatin in obesity, diabetes, and exercise adaptation.
Collapse
Affiliation(s)
- David L Allen
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80309-0354, USA.
| | | | | |
Collapse
|
37
|
Matsakas A, Macharia R, Otto A, Elashry MI, Mouisel E, Romanello V, Sartori R, Amthor H, Sandri M, Narkar V, Patel K. Exercise training attenuates the hypermuscular phenotype and restores skeletal muscle function in the myostatin null mouse. Exp Physiol 2011; 97:125-40. [DOI: 10.1113/expphysiol.2011.063008] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
38
|
Qaisar R, Renaud G, Morine K, Barton ER, Sweeney HL, Larsson L. Is functional hypertrophy and specific force coupled with the addition of myonuclei at the single muscle fiber level? FASEB J 2011; 26:1077-85. [PMID: 22125316 DOI: 10.1096/fj.11-192195] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Muscle force is typically proportional to muscle size, resulting in constant force normalized to muscle fiber cross-sectional area (specific force). Mice overexpressing insulin-like growth factor-1 (IGF-1) exhibit a proportional gain in muscle force and size, but not the myostatin-deficient mice. In an attempt to explore the role of the cytoplasmic volume supported by individual myonuclei [myonuclear domain (MND) size] on functional capacity of skeletal muscle, we have investigated specific force in relation to MND and the content of the molecular motor protein, myosin, at the single muscle fiber level from myostatin-knockout (Mstn(-/-)) and IGF-1-overexpressing (mIgf1(+/+)) mice. We hypothesize that the addition of extra myonuclei is a prerequisite for maintenance of specific force during muscle hypertrophy. A novel algorithm was used to measure individual MNDs in 3 dimensions along the length of single muscle fibers from the fast-twitch extensor digitorum longus and the slow-twitch soleus muscle. A significant effect of the size of individual MNDs in hypertrophic muscle fibers on both specific force and myosin content was observed. This effect was muscle cell type specific and suggested there is a critical volume individual myonuclei can support efficiently. The large MNDs found in fast muscles of Mstn(-/-) mice were correlated with the decrement in specific force and myosin content in Mstn(-/-) muscles. Thus, myostatin inhibition may not be able to maintain the appropriate MND for optimal function.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Department of Neuroscience, Uppsala University, SE-751 85 Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
39
|
Harrison CA, Al-Musawi SL, Walton KL. Prodomains regulate the synthesis, extracellular localisation and activity of TGF-β superfamily ligands. Growth Factors 2011; 29:174-86. [PMID: 21864080 DOI: 10.3109/08977194.2011.608666] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
All transforming growth factor-β (TGF-β) ligands are synthesised as precursor molecules consisting of a signal peptide, an N-terminal prodomain and a C-terminal mature domain. During synthesis, prodomains interact non-covalently with mature domains, maintaining the molecules in a conformation competent for dimerisation. Dimeric precursors are cleaved by proprotein convertases, and TGF-β ligands are secreted from the cell non-covalently associated with their prodomains. Extracellularly, prodomains localise TGF-β ligands within the vicinity of their target cells via interactions with extracellular matrix proteins, including fibrillin and perlecan. For some family members (TGF-β1, TGF-β2, TGF-β3, myostatin, GDF-11 and BMP-10), prodomains bind with high enough affinity to suppress biological activity. The subsequent mechanism of activation of these latent TGF-β ligands varies according to cell type and context, but all activating mechanisms directly target prodomains. Thus, prodomains control many aspects of TGF-β superfamily biology, and alterations in prodomain function are often associated with disease.
Collapse
Affiliation(s)
- Craig A Harrison
- Prince Henry's Institute of Medical Research, Clayton, VIC 3168, Australia.
| | | | | |
Collapse
|
40
|
Makanji Y, Walton KL, Chan KL, Gregorevic P, Robertson DM, Harrison CA. Generation of a specific activin antagonist by modification of the activin A propeptide. Endocrinology 2011; 152:3758-68. [PMID: 21750050 DOI: 10.1210/en.2011-1052] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Elevated activin A levels in inhibin-deficient mice promote the development of gonadal tumors and induce cachexia by reducing muscle, liver, stomach, and fat mass. Because activin A is an important regulator of tissue growth, inhibiting the actions of this TGFβ family ligand may halt or reverse pathology in diseased tissues. In this study, we modified the activin A propeptide to generate a specific activin antagonist. Propeptides mediate the synthesis and secretion of all TGFβ ligands and, for some family members (e.g. TGFβ1), bind the mature growth factor with high enough affinity to confer latency. By linking the C-terminal region of the TGFβ1 propeptide to the N-terminal region of the activin A propeptide, we generated a chimeric molecule [activin/TGFβ1 propeptide (AT propeptide)] with increased affinity for activin A. The AT propeptide was 30-fold more potent than the activin A propeptide at suppressing activin-induced FSH release by LβT2 pituitary gonadotrope cells. Binding of the AT propeptide to activin A shields the type II receptor binding site, thereby reducing Smad2 phosphorylation and downstream signaling. In comparison with the commonly used activin antagonists, follistatin (IC(50) 0.42 nM), soluble activin type II receptor A-Fc (IC(50) 0.47 nM), and soluble activin type II receptor B-Fc (IC(50) 0.91 nM), the AT propeptide (IC(50) 2.6 nM) was slightly less potent. However, it was more specific, inhibiting activin A and activin B (IC(50) 10.26 nM) but not the closely related ligands, myostatin and growth differentiation factor-11. As such, the AT propeptide represents the first specific activin antagonist, and it should be an effective reagent for blocking activin actions in vivo.
Collapse
Affiliation(s)
- Yogeshwar Makanji
- Prince Henry's Institute of Medical Research, 246 Clayton Road, Clayton VIC 3168, Australia
| | | | | | | | | | | |
Collapse
|
41
|
Welle S, Mehta S, Burgess K. Effect of postdevelopmental myostatin depletion on myofibrillar protein metabolism. Am J Physiol Endocrinol Metab 2011; 300:E993-E1001. [PMID: 21406613 PMCID: PMC3118593 DOI: 10.1152/ajpendo.00509.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
It is unclear whether the muscle hypertrophy induced by loss of myostatin signaling in mature muscles is maintained only by increased protein synthesis or whether reduced proteolysis contributes. To address this issue, we depleted myostatin by activating Cre recombinase for 2 wk in mature mice in which Mstn exon 3 was flanked by loxP sequences. The rate of phenylalanine tracer incorporation into myofibrillar proteins was determined 2, 5, and 24 wk after Cre activation ended. At all of these time points, myostatin-deficient mice had increased gastrocnemius and quadriceps muscle mass (≥27%) and increased myofibrillar synthesis rate per gastrocnemius muscle (≥19%) but normal myofibrillar synthesis rates per myofibrillar mass or RNA mass. Mean fractional myofibrillar degradation rates (estimated from the difference between rate of synthesis and rate of change in myofibrillar mass) and muscle concentrations of free 3-methylhistidine (from actin and myosin degradation) were unaffected by myostatin knockout. Overnight food deprivation reduced myofibrillar synthesis and ribosomal protein S6 phosphorylation and increased concentrations of 3-methylhistidine, muscle RING finger-1 mRNA, and atrogin-1 mRNA. Myostatin depletion did not affect these responses to food deprivation. These data indicate that maintenance of the muscle hypertrophy caused by loss of myostatin is mediated by increased protein synthesis per muscle fiber rather than suppression of proteolysis.
Collapse
Affiliation(s)
- Stephen Welle
- Department of Medicine, Division of Endocrinology and Metabolism, University of Rochester, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
42
|
Myostatin: a novel insight into its role in metabolism, signal pathways, and expression regulation. Cell Signal 2011; 23:1441-6. [PMID: 21609762 DOI: 10.1016/j.cellsig.2011.05.003] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 05/09/2011] [Indexed: 11/24/2022]
Abstract
Myostatin, a member of the transforming growth factor-β (TGF-β) superfamily, is a critical autocrine/paracrine inhibitor of skeletal muscle growth. Since the first observed double-muscling phenotype was reported in myostatin-null animals, a functional role of myostatin has been demonstrated in the control of skeletal muscle development. However, beyond the confines of its traditional role in muscle growth inhibition, myostatin has recently been shown to play an important role in metabolism. During the past several years, it has been well established that Smads are canonical mediators of signals for myostatin from the receptors to the nucleus. However, growing evidence supports the notion that Non-Smad signal pathways also participate in myostatin signaling. Myostatin expression is increased in muscle atrophy and metabolic disorders, suggesting that changes in endogenous expression of myostatin may provide therapeutic benefit for these diseases. MicroRNAs (miRNAs) are a class of non-coding RNAs that negatively regulate gene expression and recent evidence has accumulated supporting a role for miRNAs in the regulation of myostatin expression. This review highlights some of these areas in myostatin research: a novel role in metabolism, signal pathways, and miRNA-mediated expression regulation.
Collapse
|
43
|
Burks TN, Cohn RD. Role of TGF-β signaling in inherited and acquired myopathies. Skelet Muscle 2011; 1:19. [PMID: 21798096 PMCID: PMC3156642 DOI: 10.1186/2044-5040-1-19] [Citation(s) in RCA: 177] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 05/04/2011] [Indexed: 01/25/2023] Open
Abstract
The transforming growth factor-beta (TGF-β) superfamily consists of a variety of cytokines expressed in many different cell types including skeletal muscle. Members of this superfamily that are of particular importance in skeletal muscle are TGF-β1, mitogen-activated protein kinases (MAPKs), and myostatin. These signaling molecules play important roles in skeletal muscle homeostasis and in a variety of inherited and acquired neuromuscular disorders. Expression of these molecules is linked to normal processes in skeletal muscle such as growth, differentiation, regeneration, and stress response. However, chronic elevation of TGF-β1, MAPKs, and myostatin is linked to various features of muscle pathology, including impaired regeneration and atrophy. In this review, we focus on the aberrant signaling of TGF-β in various disorders such as Marfan syndrome, muscular dystrophies, sarcopenia, and critical illness myopathy. We also discuss how the inhibition of several members of the TGF-β signaling pathway has been implicated in ameliorating disease phenotypes, opening up novel therapeutic avenues for a large group of neuromuscular disorders.
Collapse
Affiliation(s)
- Tyesha N Burks
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | |
Collapse
|
44
|
Kemaladewi DU, Hoogaars WMH, van Heiningen SH, Terlouw S, de Gorter DJJ, den Dunnen JT, van Ommen GJB, Aartsma-Rus A, ten Dijke P, 't Hoen PAC. Dual exon skipping in myostatin and dystrophin for Duchenne muscular dystrophy. BMC Med Genomics 2011; 4:36. [PMID: 21507246 PMCID: PMC3107769 DOI: 10.1186/1755-8794-4-36] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Accepted: 04/20/2011] [Indexed: 12/26/2022] Open
Abstract
Background Myostatin is a potent muscle growth inhibitor that belongs to the Transforming Growth Factor-β (TGF-β) family. Mutations leading to non functional myostatin have been associated with hypermuscularity in several organisms. By contrast, Duchenne muscular dystrophy (DMD) is characterized by a loss of muscle fibers and impaired regeneration. In this study, we aim to knockdown myostatin by means of exon skipping, a technique which has been successfully applied to reframe the genetic defect of dystrophin gene in DMD patients. Methods We targeted myostatin exon 2 using antisense oligonucleotides (AON) in healthy and DMD-derived myotubes cultures. We assessed the exon skipping level, transcriptional expression of myostatin and its target genes, and combined myostatin and several dystrophin AONs. These AONs were also applied in the mdx mice models via intramuscular injections. Results Myostatin AON induced exon 2 skipping in cell cultures and to a lower extent in the mdx mice. It was accompanied by decrease in myostatin mRNA and enhanced MYOG and MYF5 expression. Furthermore, combination of myostatin and dystrophin AONs induced simultaneous skipping of both genes. Conclusions We conclude that two AONs can be used to target two different genes, MSTN and DMD, in a straightforward manner. Targeting multiple ligands of TGF-beta family will be more promising as adjuvant therapies for DMD.
Collapse
Affiliation(s)
- Dwi U Kemaladewi
- Center for Human and Clinical Genetics, Leiden University Medical Center, Postzone S4-P, PO Box 9600, Leiden, 2300RC, the Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Gentry BA, Ferreira JA, Phillips CL, Brown M. Hindlimb skeletal muscle function in myostatin-deficient mice. Muscle Nerve 2011; 43:49-57. [PMID: 21082689 DOI: 10.1002/mus.21796] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Absence of functional myostatin (MSTN) during fetal development results in adult skeletal muscle hypertrophy and hyperplasia. To more fully characterize MSTN loss in hindlimb muscles, the morphology and contractile function of the soleus, plantaris, gastrocnemius, tibialis anterior, and quadriceps muscles in male and female null (Mstn(-/-)), heterozygous (Mstn(+/-)), and wild-type (Mstn(+/+)) mice were investigated. Muscle weights of Mstn(-/-) mice were greater than those of Mstn(+/+) and Mstn(+/-) mice. Fiber cross-sectional area (CSA) was increased in female Mstn(-/-) soleus and gastrocnemius muscles and in the quadriceps of male Mstn(-/-) mice; peak tetanic force in Mstn(-/-) mice did not parallel the increased muscle weight or CSA. Male Mstn(-/-) muscle exhibited moderate degeneration. Visible pathology in male mice and decreased contractile strength relative to increased muscle weight suggest MSTN loss results in muscle impairment, which is dose-, sex-, and muscle-dependent.
Collapse
Affiliation(s)
- Bettina A Gentry
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | | | | | | |
Collapse
|
46
|
Matsakas A, Otto A, Elashry MI, Brown SC, Patel K. Altered primary and secondary myogenesis in the myostatin-null mouse. Rejuvenation Res 2011; 13:717-27. [PMID: 21204650 DOI: 10.1089/rej.2010.1065] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Skeletal muscle fiber generation occurs principally in two myogenic phases: (1) Primary (embryonic) myogenesis when myoblasts proliferate and fuse to form primary myotubes and (2) secondary (fetal) myogenesis when successive waves of myoblasts fuse along the surface of the primary myotubes, giving rise to a population of smaller and more numerous secondary myotubes. This sequence of events determines fiber number and is completed at or soon after birth in most muscles of the mouse. The adult myostatin null mouse (MSTN(-/-)) displays both an increase in fiber number and size relative to wild type (MSTN(+/+)), suggesting a developmental origin for the hypermuscular phenotype. The focus of the present study was to determine at which point during myogenesis do MSTN(-/-) animals diverge from MSTN(+/+). To achieve this, we focused on the extensor digitorum longus (EDL) muscle and evaluated primary myotube number at embryonic day (E) 13.0 and E14.5 and secondary to primary myotube ratios at E18.5. We show that primary myotube number and size were significantly increased in the MSTN(-/-) mice by E14.5 and the secondary to primary myotube ratio increased at E18.5. This increase in the rate of fiber formation resulted in MSTN(-/-) mice harboring 87% of their final adult fiber number at E18.5, compared to only 73% in MSTN(+/+). An accelerated myogenic program in the MSTN(-/-) mice was further confirmed by our finding of an initial expansion in the myogenic stem cell (identified through Pax7 expression) and myoblast (identified through myogenin expression) cell pools at E14.5 in the EDL muscle of these animals that was, however, followed by a reduction of both populations of cells at E18.5 relative to MSTN(+/+). Overall these data suggest that the genetic loss of myostatin accelerates the developmental myogenic program of primary and secondary skeletal myogenesis.
Collapse
Affiliation(s)
- Antonios Matsakas
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | | | | | | |
Collapse
|
47
|
Abstract
Myostatin is a member of the transforming growth factor β superfamily of secreted growth factors that negatively regulates skeletal muscle size. Mice null for the myostatin gene have a dramatically increased mass of individual muscles, reduced adiposity, increased insulin sensitivity, and resistance to obesity. Myostatin inhibition in adult mice also increases muscle mass which raises the possibility that anti-myostatin therapy could be a useful approach for treating diseases such as obesity or diabetes in addition to muscle wasting diseases. In this review I will describe the present state of our understanding of the role of myostatin and the closely related growth factor growth/differentiation factor 11 on metabolism.
Collapse
Affiliation(s)
- Alexandra C McPherron
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland USA
| |
Collapse
|
48
|
Lee SJ. Extracellular Regulation of Myostatin: A Molecular Rheostat for Muscle Mass. ACTA ACUST UNITED AC 2010; 10:183-194. [PMID: 21423813 DOI: 10.2174/187152210793663748] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Myostatin (MSTN) is a transforming growth factor-ß family member that plays a critical role in regulating skeletal muscle mass. Genetic studies in multiple species have demonstrated that mutations in the Mstn gene lead to dramatic and widespread increases in muscle mass as a result of a combination of increased fiber numbers and increased fiber sizes. MSTN inhibitors have also been shown to cause significant increases in muscle growth when administered to adult mice. As a result, there has been an extensive effort to understand the mechanisms underlying MSTN regulation and activity with the goal of developing the most effective strategies for targeting this signaling pathway for clinical applications. Here, I review the current state of knowledge regarding the regulation of MSTN extracellularly by binding proteins and discuss the implications of these findings both with respect to the fundamental physiological role that MSTN plays in regulating tissue homeostasis and with respect to the development of therapeutic agents to combat muscle loss.
Collapse
Affiliation(s)
- Se-Jin Lee
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, PCTB 803, 725 North Wolfe Street, Baltimore, Maryland 21205, USA
| |
Collapse
|
49
|
MacLea KS, Covi JA, Kim HW, Chao E, Medler S, Chang ES, Mykles DL. Myostatin from the American lobster, Homarus americanus: Cloning and effects of molting on expression in skeletal muscles. Comp Biochem Physiol A Mol Integr Physiol 2010; 157:328-37. [DOI: 10.1016/j.cbpa.2010.07.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 07/27/2010] [Accepted: 07/30/2010] [Indexed: 10/19/2022]
|
50
|
Otto A, Patel K. Signalling and the control of skeletal muscle size. Exp Cell Res 2010; 316:3059-66. [DOI: 10.1016/j.yexcr.2010.04.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Accepted: 04/12/2010] [Indexed: 11/26/2022]
|