1
|
Wetzlich B, Nyakundi BB, Yang J. Therapeutic applications and challenges in myostatin inhibition for enhanced skeletal muscle mass and functions. Mol Cell Biochem 2025; 480:1535-1553. [PMID: 39340593 PMCID: PMC11842502 DOI: 10.1007/s11010-024-05120-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024]
Abstract
Myostatin, a potent negative regulator of skeletal muscle mass, has garnered significant attention as a therapeutic target for muscle dystrophies. Despite extensive research and promising preclinical results, clinical trials targeting myostatin inhibition in muscle dystrophies have failed to yield substantial improvements in muscle function or fitness in patients. This review details the mechanisms behind myostatin's function and the various inhibitors that have been tested preclinically and clinically. It also examines the challenges encountered in clinical translation, including issues with drug specificity, differences in serum myostatin concentrations between animal models and humans, and the necessity of neural input for functional improvements. Additionally, we explore promising avenues of research beyond muscle dystrophies, particularly in the treatment of metabolic syndromes and orthopedic disorders. Insights from these alternative applications suggest that myostatin inhibition may hold the potential for addressing a broader range of pathologies, providing new directions for therapeutic development.
Collapse
Affiliation(s)
- Brock Wetzlich
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Benard B Nyakundi
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Jinzeng Yang
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
2
|
Chen MM, Zhao Y, Yu K, Xu XL, Zhang XS, Zhang JL, Wu SJ, Liu ZM, Yuan YM, Guo XF, Qi SY, Yi G, Wang SQ, Li HX, Wu AW, Liu GS, Deng SL, Han HB, Lv FH, Lian D, Lian ZX. A MSTNDel73C mutation with FGF5 knockout sheep by CRISPR/Cas9 promotes skeletal muscle myofiber hyperplasia. eLife 2024; 12:RP86827. [PMID: 39365728 PMCID: PMC11452178 DOI: 10.7554/elife.86827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2024] Open
Abstract
Mutations in the well-known Myostatin (MSTN) produce a 'double-muscle' phenotype, which makes it commercially invaluable for improving livestock meat production and providing high-quality protein for humans. However, mutations at different loci of the MSTN often produce a variety of different phenotypes. In the current study, we increased the delivery ratio of Cas9 mRNA to sgRNA from the traditional 1:2 to 1:10, which improves the efficiency of the homozygous mutation of biallelic gene. Here, a MSTNDel73C mutation with FGF5 knockout sheep, in which the MSTN and FGF5 dual-gene biallelic homozygous mutations were produced via the deletion of 3-base pairs of AGC in the third exon of MSTN, resulting in cysteine-depleted at amino acid position 73, and the FGF5 double allele mutation led to inactivation of FGF5 gene. The MSTNDel73C mutation with FGF5 knockout sheep highlights a dominant 'double-muscle' phenotype, which can be stably inherited. Both F0 and F1 generation mutants highlight the excellent trait of high-yield meat with a smaller cross-sectional area and higher number of muscle fibers per unit area. Mechanistically, the MSTNDel73C mutation with FGF5 knockout mediated the activation of FOSL1 via the MEK-ERK-FOSL1 axis. The activated FOSL1 promotes skeletal muscle satellite cell proliferation and inhibits myogenic differentiation by inhibiting the expression of MyoD1, and resulting in smaller myotubes. In addition, activated ERK1/2 may inhibit the secondary fusion of myotubes by Ca2+-dependent CaMKII activation pathway, leading to myoblasts fusion to form smaller myotubes.
Collapse
Affiliation(s)
- Ming-Ming Chen
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Yue Zhao
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Kun Yu
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Xue-Ling Xu
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Xiao-Sheng Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Tianjin Academy of Agricultural SciencesTianjinChina
| | - Jin-Long Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Tianjin Academy of Agricultural SciencesTianjinChina
| | - Su-Jun Wu
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Zhi-Mei Liu
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Yi-Ming Yuan
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Xiao-Fei Guo
- Institute of Animal Husbandry and Veterinary Medicine, Tianjin Academy of Agricultural SciencesTianjinChina
| | - Shi-Yu Qi
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Guang Yi
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Shu-Qi Wang
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Huang-Xiang Li
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Ao-Wu Wu
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Guo-Shi Liu
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Shou-Long Deng
- National Center of Technology Innovation for animal model, NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical CollegeBeijingChina
| | - Hong-Bing Han
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Feng-Hua Lv
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| | - Di Lian
- College of Pulmonary and Critical Care Medicine, Chinese PLA General HospitalBeijingChina
| | - Zheng-Xing Lian
- State Key Laboratory of Animal Biotech Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural UniversityBeijingChina
| |
Collapse
|
3
|
Jha NN, Kim JK, Her YR, Monani UR. Muscle: an independent contributor to the neuromuscular spinal muscular atrophy disease phenotype. JCI Insight 2023; 8:e171878. [PMID: 37737261 PMCID: PMC10561723 DOI: 10.1172/jci.insight.171878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023] Open
Abstract
Spinal muscular atrophy (SMA) is a pediatric-onset neuromuscular disorder caused by insufficient survival motor neuron (SMN) protein. SMN restorative therapies are now approved for the treatment of SMA; however, they are not curative, likely due to a combination of imperfect treatment timing, inadequate SMN augmentation, and failure to optimally target relevant organs. Here, we consider the implications of imperfect treatment administration, focusing specifically on outcomes for skeletal muscle. We examine the evidence that muscle plays a contributing role in driving neuromuscular dysfunction in SMA. Next, we discuss how SMN might regulate the health of myofibers and their progenitors. Finally, we speculate on therapeutic outcomes of failing to raise muscle SMN to healthful levels and present strategies to restore function to this tissue to ensure better treatment results.
Collapse
Affiliation(s)
- Narendra N. Jha
- Department of Neurology
- Center for Motor Neuron Biology and Disease, and
| | - Jeong-Ki Kim
- Department of Neurology
- Center for Motor Neuron Biology and Disease, and
| | - Yoon-Ra Her
- Department of Neurology
- Center for Motor Neuron Biology and Disease, and
| | - Umrao R. Monani
- Department of Neurology
- Center for Motor Neuron Biology and Disease, and
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
4
|
Bez Batti Angulski A, Hosny N, Cohen H, Martin AA, Hahn D, Bauer J, Metzger JM. Duchenne muscular dystrophy: disease mechanism and therapeutic strategies. Front Physiol 2023; 14:1183101. [PMID: 37435300 PMCID: PMC10330733 DOI: 10.3389/fphys.2023.1183101] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/24/2023] [Indexed: 07/13/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe, progressive, and ultimately fatal disease of skeletal muscle wasting, respiratory insufficiency, and cardiomyopathy. The identification of the dystrophin gene as central to DMD pathogenesis has led to the understanding of the muscle membrane and the proteins involved in membrane stability as the focal point of the disease. The lessons learned from decades of research in human genetics, biochemistry, and physiology have culminated in establishing the myriad functionalities of dystrophin in striated muscle biology. Here, we review the pathophysiological basis of DMD and discuss recent progress toward the development of therapeutic strategies for DMD that are currently close to or are in human clinical trials. The first section of the review focuses on DMD and the mechanisms contributing to membrane instability, inflammation, and fibrosis. The second section discusses therapeutic strategies currently used to treat DMD. This includes a focus on outlining the strengths and limitations of approaches directed at correcting the genetic defect through dystrophin gene replacement, modification, repair, and/or a range of dystrophin-independent approaches. The final section highlights the different therapeutic strategies for DMD currently in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
5
|
Saraswathy VM, Zhou L, McAdow AR, Burris B, Dogra D, Reischauer S, Mokalled MH. Myostatin is a negative regulator of adult neurogenesis after spinal cord injury in zebrafish. Cell Rep 2022; 41:111705. [PMID: 36417881 PMCID: PMC9742758 DOI: 10.1016/j.celrep.2022.111705] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 05/16/2022] [Accepted: 11/01/2022] [Indexed: 11/23/2022] Open
Abstract
Intrinsic and extrinsic inhibition of neuronal regeneration obstruct spinal cord (SC) repair in mammals. In contrast, adult zebrafish achieve functional recovery after complete SC transection. While studies of innate SC regeneration have focused on axon regrowth as a primary repair mechanism, how local adult neurogenesis affects functional recovery is unknown. Here, we uncover dynamic expression of zebrafish myostatin b (mstnb) in a niche of dorsal SC progenitors after injury. mstnb mutants show impaired functional recovery, normal glial and axonal bridging across the lesion, and an increase in the profiles of newborn neurons. Molecularly, neuron differentiation genes are upregulated, while the neural stem cell maintenance gene fgf1b is downregulated in mstnb mutants. Finally, we show that human fibroblast growth factor 1 (FGF1) treatment rescues the molecular and cellular phenotypes of mstnb mutants. These studies uncover unanticipated neurogenic functions for mstnb and establish the importance of local adult neurogenesis for innate SC repair.
Collapse
Affiliation(s)
- Vishnu Muraleedharan Saraswathy
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lili Zhou
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anthony R McAdow
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brooke Burris
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Deepika Dogra
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany; Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Sven Reischauer
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany; Medical Clinic I, (Cardiology/Angiology) and Campus Kerckhoff, Justus Liebig University, Giessen, 35392 Giessen, Germany; The Cardio-Pulmonary Institute, Frankfurt, Germany
| | - Mayssa H Mokalled
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
6
|
Zheng Y, Zhang Y, Wu L, Riaz H, Li Z, Shi D, Rehman SU, Liu Q, Cui K. Generation of Heritable Prominent Double Muscle Buttock Rabbits via Novel Site Editing of Myostatin Gene Using CRISPR/Cas9 System. Front Vet Sci 2022; 9:842074. [PMID: 35669173 PMCID: PMC9165342 DOI: 10.3389/fvets.2022.842074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/18/2022] [Indexed: 12/25/2022] Open
Abstract
Rabbits have been domesticated for meat, wool, and fur production, and have also been cherished as a companion, artistic inspiration, and an experimental model to study many human diseases. In the present study, the muscle mass negative regulator gene myostatin (MSTN) was knocked out in rabbits at two novel sites in exon3, and the function of these mutations was determined in subsequent generations. The prominent double muscle phenotype with hyperplasia or hypertrophy of muscle fiber was observed in the MSTN-KO rabbits, and a similar phenotype was confirmed in the F1 generation. Moreover, the average weight of 80-day-old MSTN-KO rabbits (2,452 ± 63 g) was higher than that of wild-type rabbits (2,393.2 ± 106.88 g), and also the bodyweight of MSTN-KO rabbits (3,708 ± 43.06g) was significantly higher (P < 0.001) at the age of 180 days than wild-type (WT) rabbits (3,224 ± 48.64g). In MSTN-KO rabbits, fourteen rabbit pups from the F1 generation and thirteen from the F2 generation stably inherited the induced MSTN gene mutations. Totally, 194 pups were produced in the F1 generation of which 49 were MSTN-KO rabbits, while 47 pups were produced in the F2 generation of which 20 were edited rabbits, and the ratio of edited to wild-type rabbits in the F2 generation was approximately 1:1. Thus, we successfully generated a heritable double muscle buttocks rabbits via myostatin mutation with CRISPR/Cas9 system, which could be valuable in rabbit's meat production and also a useful animal model to study the development of muscles among livestock species and improve their important economic traits as well as the human muscle development-related diseases.
Collapse
Affiliation(s)
- Yalin Zheng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| | - Yu Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| | - Liyan Wu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| | - Hasan Riaz
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Zhipeng Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| | - Deshun Shi
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| | - Saif Ur Rehman
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| | - Qingyou Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China.,Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Kuiqing Cui
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| |
Collapse
|
7
|
Jablonka S, Hennlein L, Sendtner M. Therapy development for spinal muscular atrophy: perspectives for muscular dystrophies and neurodegenerative disorders. Neurol Res Pract 2022; 4:2. [PMID: 34983696 PMCID: PMC8725368 DOI: 10.1186/s42466-021-00162-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/21/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Major efforts have been made in the last decade to develop and improve therapies for proximal spinal muscular atrophy (SMA). The introduction of Nusinersen/Spinraza™ as an antisense oligonucleotide therapy, Onasemnogene abeparvovec/Zolgensma™ as an AAV9-based gene therapy and Risdiplam/Evrysdi™ as a small molecule modifier of pre-mRNA splicing have set new standards for interference with neurodegeneration. MAIN BODY Therapies for SMA are designed to interfere with the cellular basis of the disease by modifying pre-mRNA splicing and enhancing expression of the Survival Motor Neuron (SMN) protein, which is only expressed at low levels in this disorder. The corresponding strategies also can be applied to other disease mechanisms caused by loss of function or toxic gain of function mutations. The development of therapies for SMA was based on the use of cell culture systems and mouse models, as well as innovative clinical trials that included readouts that had originally been introduced and optimized in preclinical studies. This is summarized in the first part of this review. The second part discusses current developments and perspectives for amyotrophic lateral sclerosis, muscular dystrophies, Parkinson's and Alzheimer's disease, as well as the obstacles that need to be overcome to introduce RNA-based therapies and gene therapies for these disorders. CONCLUSION RNA-based therapies offer chances for therapy development of complex neurodegenerative disorders such as amyotrophic lateral sclerosis, muscular dystrophies, Parkinson's and Alzheimer's disease. The experiences made with these new drugs for SMA, and also the experiences in AAV gene therapies could help to broaden the spectrum of current approaches to interfere with pathophysiological mechanisms in neurodegeneration.
Collapse
Affiliation(s)
- Sibylle Jablonka
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Versbacher Str. 5, 97078, Wuerzburg, Germany.
| | - Luisa Hennlein
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Versbacher Str. 5, 97078, Wuerzburg, Germany
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Versbacher Str. 5, 97078, Wuerzburg, Germany.
| |
Collapse
|
8
|
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
|
9
|
In Search of a Cure: The Development of Therapeutics to Alter the Progression of Spinal Muscular Atrophy. Brain Sci 2021; 11:brainsci11020194. [PMID: 33562482 PMCID: PMC7915832 DOI: 10.3390/brainsci11020194] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/19/2022] Open
Abstract
Until the recent development of disease-modifying therapeutics, spinal muscular atrophy (SMA) was considered a devastating neuromuscular disease with a poor prognosis for most affected individuals. Symptoms generally present during early childhood and manifest as muscle weakness and progressive paralysis, severely compromising the affected individual’s quality of life, independence, and lifespan. SMA is most commonly caused by the inheritance of homozygously deleted SMN1 alleles with retention of one or more copies of a paralog gene, SMN2, which inversely correlates with disease severity. The recent advent and use of genetically targeted therapies have transformed SMA into a prototype for monogenic disease treatment in the era of genetic medicine. Many SMA-affected individuals receiving these therapies achieve traditionally unobtainable motor milestones and survival rates as medicines drastically alter the natural progression of this disease. This review discusses historical SMA progression and underlying disease mechanisms, highlights advances made in therapeutic research, clinical trials, and FDA-approved medicines, and discusses possible second-generation and complementary medicines as well as optimal temporal intervention windows in order to optimize motor function and improve quality of life for all SMA-affected individuals.
Collapse
|
10
|
Muramatsu H, Kuramochi T, Katada H, Ueyama A, Ruike Y, Ohmine K, Shida-Kawazoe M, Miyano-Nishizawa R, Shimizu Y, Okuda M, Hori Y, Hayashi M, Haraya K, Ban N, Nonaka T, Honda M, Kitamura H, Hattori K, Kitazawa T, Igawa T, Kawabe Y, Nezu J. Novel myostatin-specific antibody enhances muscle strength in muscle disease models. Sci Rep 2021; 11:2160. [PMID: 33495503 PMCID: PMC7835227 DOI: 10.1038/s41598-021-81669-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/08/2021] [Indexed: 11/22/2022] Open
Abstract
Myostatin, a member of the transforming growth factor-β superfamily, is an attractive target for muscle disease therapy because of its role as a negative regulator of muscle growth and strength. Here, we describe a novel antibody therapeutic approach that maximizes the potential of myostatin-targeted therapy. We generated an antibody, GYM329, that specifically binds the latent form of myostatin and inhibits its activation. Additionally, via "sweeping antibody technology", GYM329 reduces or "sweeps" myostatin in the muscle and plasma. Compared with conventional anti-myostatin agents, GYM329 and its surrogate antibody exhibit superior muscle strength-improvement effects in three different mouse disease models. We also demonstrate that the superior efficacy of GYM329 is due to its myostatin specificity and sweeping capability. Furthermore, we show that a GYM329 surrogate increases muscle mass in normal cynomolgus monkeys without any obvious toxicity. Our findings indicate the potential of GYM329 to improve muscle strength in patients with muscular disorders.
Collapse
Affiliation(s)
- Hiroyasu Muramatsu
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Taichi Kuramochi
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #07-11 to 16, Synapse, Singapore, 138623, Singapore
| | - Hitoshi Katada
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Atsunori Ueyama
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Yoshinao Ruike
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Ken Ohmine
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | | | | | - Yuichiro Shimizu
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Momoko Okuda
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #07-11 to 16, Synapse, Singapore, 138623, Singapore
| | - Yuji Hori
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Madoka Hayashi
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Kenta Haraya
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Nobuhiro Ban
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Tatsuya Nonaka
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Masaki Honda
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Hidetomo Kitamura
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Kunihiro Hattori
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Takehisa Kitazawa
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Tomoyuki Igawa
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #07-11 to 16, Synapse, Singapore, 138623, Singapore
| | - Yoshiki Kawabe
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Junichi Nezu
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan.
| |
Collapse
|
11
|
Suh J, Lee YS. Myostatin Inhibitors: Panacea or Predicament for Musculoskeletal Disorders? J Bone Metab 2020; 27:151-165. [PMID: 32911580 PMCID: PMC7571243 DOI: 10.11005/jbm.2020.27.3.151] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/23/2020] [Indexed: 01/19/2023] Open
Abstract
Myostatin, also known as growth differentiation factor 8 (GDF8), is a transforming growth factor-β (TGF-β) family member that functions to limit skeletal muscle growth. Accordingly, loss-of-function mutations in myostatin result in a dramatic increase in muscle mass in humans and various animals, while its overexpression leads to severe muscle atrophy. Myostatin also exerts a significant effect on bone metabolism, as demonstrated by enhanced bone mineral density and bone regeneration in myostatin null mice. The identification of myostatin as a negative regulator of muscle and bone mass has sparked an enormous interest in developing myostatin inhibitors as therapeutic agents for treating a variety of clinical conditions associated with musculoskeletal disorders. As a result, various myostatin-targeting strategies involving antibodies, myostatin propeptides, soluble receptors, and endogenous antagonists have been generated, and many of them have progressed to clinical trials. Importantly, most myostatin inhibitors also repress the activities of other closely related TGF-β family members including GDF11, activins, and bone morphogenetic proteins (BMPs), increasing the potential for unwanted side effects, such as vascular side effects through inhibition of BMP 9/10 and bone weakness induced by follistatin through antagonizing several TGF-β family members. Therefore, a careful distinction between targets that may enhance the efficacy of an agent and those that may cause adverse effects is required with the improvement of the target specificity. In this review, we discuss the current understanding of the endogenous function of myostatin, and provide an overview of clinical trial outcomes from different myostatin inhibitors.
Collapse
Affiliation(s)
- Joonho Suh
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Yun-Sil Lee
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
12
|
A Novel Nanobody Directed against Ovine Myostatin to Enhance Muscle Growth in Mouse. Animals (Basel) 2020; 10:ani10081398. [PMID: 32796682 PMCID: PMC7460164 DOI: 10.3390/ani10081398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/02/2020] [Accepted: 08/04/2020] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Myostatin (MSTN) is a negative regulator of myogenesis, and various strategies have been used to improve livestock production by inhibiting MSTN. In this study, we developed a single-variable domain of the heavy chain antibody-recombinant MSTN nanobody (RMN) against MSTN. The selected RMN was expressed, purified, and assessed for its cytotoxicity, affinity, specificity, and the ability to inhibit MSTN. The results demonstrated that RMN could specifically detect and bind MSTN, further inhibit myostatin activity, as well as enhance muscle growth in mice. Abstract Myostatin (MSTN) is a member of the transforming growth factor beta superfamily and is a negative regulator of myogenesis. It has been shown to function by controlling the proliferation of myoblasts. MSTN inhibition is considered as a promising treatment for promoting animal growth in livestock. Nanobodies, a special antibody discovered in camel, have arisen as an alternative to conventional antibodies and have shown great potential when used as tools in different biotechnology fields, such as diagnostics and therapy. In this study, we examined the effect of MSTN inhibition by RMN on the muscle growth of mice. The results showed that RMN could specifically detect and bind MSTN, as well as inhibit MSTN activity. A significant increase in skeletal muscle mass was observed after intramuscular injection of RMN into mice. Enhanced muscle growth occurred because of myofiber hypertrophy. These results offer a promising approach to enhance muscle growth that warrants further investigation in domestic animals.
Collapse
|
13
|
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: 52] [Impact Index Per Article: 10.4] [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
|
14
|
Kim GD, Lee JH, Song S, Kim SW, Han JS, Shin SP, Park BC, Park TS. Generation of myostatin-knockout chickens mediated by D10A-Cas9 nickase. FASEB J 2020; 34:5688-5696. [PMID: 32100378 DOI: 10.1096/fj.201903035r] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/27/2020] [Accepted: 02/16/2020] [Indexed: 12/20/2022]
Abstract
Many studies have been conducted to improve economically important livestock traits such as feed efficiency and muscle growth. Genome editing technologies represent a major advancement for both basic research and agronomic biotechnology development. The clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 technical platform is a powerful tool used to engineer specific targeted loci. However, the potential occurrence of off-target effects, including the cleavage of unintended targets, limits the practical applications of Cas9-mediated genome editing. In this study, to minimize the off-target effects of this technology, we utilized D10A-Cas9 nickase to generate myostatin-knockout (MSTN KO) chickens via primordial germ cells. D10A-Cas9 nickase (Cas9n)-mediated MSTN KO chickens exhibited significantly larger skeletal muscles in the breast and leg. Degrees of skeletal muscle hypertrophy and hyperplasia induced by myostatin deletion differed by sex and muscle type. The abdominal fat deposition was dramatically lower in MSTN KO chickens than in wild-type chickens. Our results demonstrate that the D10A-Cas9 technical platform can facilitate precise and efficient targeted genome engineering and may broaden the range of applications for genome-edited chickens in practical industrialization and as animal models of human diseases.
Collapse
Affiliation(s)
- Gap-Don Kim
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, South Korea.,Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, South Korea
| | - Jeong Hyo Lee
- Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, South Korea
| | - Sumin Song
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, South Korea
| | - Seo Woo Kim
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, South Korea
| | - Ji Seon Han
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, South Korea
| | - Seung Pyo Shin
- Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, South Korea
| | - Byung-Chul Park
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, South Korea.,Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, South Korea.,Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - Tae Sub Park
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, South Korea.,Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, South Korea
| |
Collapse
|
15
|
Mukund K, Subramaniam S. Skeletal muscle: A review of molecular structure and function, in health and disease. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1462. [PMID: 31407867 PMCID: PMC6916202 DOI: 10.1002/wsbm.1462] [Citation(s) in RCA: 279] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/03/2019] [Accepted: 07/03/2019] [Indexed: 12/11/2022]
Abstract
Decades of research in skeletal muscle physiology have provided multiscale insights into the structural and functional complexity of this important anatomical tissue, designed to accomplish the task of generating contraction, force and movement. Skeletal muscle can be viewed as a biomechanical device with various interacting components including the autonomic nerves for impulse transmission, vasculature for efficient oxygenation, and embedded regulatory and metabolic machinery for maintaining cellular homeostasis. The "omics" revolution has propelled a new era in muscle research, allowing us to discern minute details of molecular cross-talk required for effective coordination between the myriad interacting components for efficient muscle function. The objective of this review is to provide a systems-level, comprehensive mapping the molecular mechanisms underlying skeletal muscle structure and function, in health and disease. We begin this review with a focus on molecular mechanisms underlying muscle tissue development (myogenesis), with an emphasis on satellite cells and muscle regeneration. We next review the molecular structure and mechanisms underlying the many structural components of the muscle: neuromuscular junction, sarcomere, cytoskeleton, extracellular matrix, and vasculature surrounding muscle. We highlight aberrant molecular mechanisms and their possible clinical or pathophysiological relevance. We particularly emphasize the impact of environmental stressors (inflammation and oxidative stress) in contributing to muscle pathophysiology including atrophy, hypertrophy, and fibrosis. This article is categorized under: Physiology > Mammalian Physiology in Health and Disease Developmental Biology > Developmental Processes in Health and Disease Models of Systems Properties and Processes > Cellular Models.
Collapse
Affiliation(s)
- Kavitha Mukund
- Department of BioengineeringUniversity of CaliforniaSan DiegoCalifornia
| | - Shankar Subramaniam
- Department of Bioengineering, Bioinformatics & Systems BiologyUniversity of CaliforniaSan DiegoCalifornia
- Department of Computer Science and EngineeringUniversity of CaliforniaSan DiegoCalifornia
- Department of Cellular and Molecular Medicine and NanoengineeringUniversity of CaliforniaSan DiegoCalifornia
| |
Collapse
|
16
|
Significant body mass increase by oral administration of a cascade of shIL21-MSTN yeast-based DNA vaccine in mice. Biomed Pharmacother 2019; 118:109147. [PMID: 31302418 DOI: 10.1016/j.biopha.2019.109147] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/31/2022] Open
Abstract
Base on the practical of MSTN-specific yeast-based protein vaccine in mice as described previously, this research was designed for developing a better DNA vaccine (a cascade of shIL21-MSTN yeast-based DNA vaccine) than solely MSTN yeast-based DNA vaccine to block the endogenous MSTN in the murine model. We first constructed the target vectors, including CMV-driven MSTN expression vector and a combined shIL21-MSTN vector which containing MSTN expression cassette and shIL21 (short hairpin RNA-IL21) expression cassette. After necessary validation, recombinant yeast vaccines harboring different vectors were well prepared. Subsequently, after 2-month administration, the MSTN-specific immune response was detected with western blots. The commercial ELISA assays indicated that the production of IL21 and IL6 were decreased compared with control groups. More importantly, the MSTN-specific antibody titer was much higher in the shIL21-MSTN group than MSTN group, which was consistent with the western blots result. The most important finding was significant body mass increased after oral administration of these yeast-based DNA vaccines, in which the shIL21-MSTN vaccine is slightly higher than the sole MSTN vaccine in mice. In this study, we confirmed the role of different MSTN-specific yeast-based DNA vaccines on increasing body mass in mice, to provide a good inspiration for livestock breeding through the new type of immunoregulatory method. On the other hand, we also detected the possible modulating role of shIL21 on the dendritic cell-mediated immune function which needs more practical application and deeper exploration.
Collapse
|
17
|
Bai Y, Bi H, Li L, Li J, Yu X, Ren H, Li Y, Ji Y, Li H, Wang H. Effects of myostatin deficiency on PGC-1α and FNDC5 expression in three different murine muscle types. Acta Histochem 2019; 121:323-329. [PMID: 30777303 DOI: 10.1016/j.acthis.2019.02.001] [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: 10/30/2018] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 12/18/2022]
Abstract
Myostatin (MSTN) is a key negative regulator of muscle growth and development. Skeletal, cardiac, and smooth muscles were isolated from MSTN knockout (MSTN-∕-) and control mice to investigate the effect of knocking out MSTN on peroxisome proliferator-activated receptor 1 coactivator (PGC-1α)-III and fibronectin domain 5 (FNDC5) expression. Various molecular biology techniques were used to analyze the changes in PGC-1α-FNDC5 in different muscle types from MSTN-∕- mice. The expression levels of PGC-1α and FNDC5 in the skeletal, cardiac, and smooth muscles of MSTN-∕- mice differed from those in the skeletal, cardiac, and smooth muscles of normal mice. This study revealed that knocking out MSTN resulted in inconsistent PGC-1α and FNDC5 expression in specific muscles. It proved for the first time that MSTN deletion attenuated the expression of PGC-1α and FNDC5 in three different murine muscle types. MSTN deletion may have additional effects on the status ofFNDC5 expression. Further research, however, is needed to confirm this conclusion.
Collapse
|
18
|
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
|
19
|
Lodberg A, van der Eerden BCJ, Boers-Sijmons B, Thomsen JS, Brüel A, van Leeuwen JPTM, Eijken M. A follistatin-based molecule increases muscle and bone mass without affecting the red blood cell count in mice. FASEB J 2019; 33:6001-6010. [PMID: 30759349 DOI: 10.1096/fj.201801969rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Inhibitors of the activin receptor signaling pathway (IASPs) have become candidate therapeutics for sarcopenia and bone remodeling disorders because of their ability to increase muscle and bone mass. However, IASPs utilizing activin type IIA and IIB receptors are also potent stimulators of erythropoiesis, a feature that may restrict their usage to anemic patients because of increased risk of venous thromboembolism. Based on the endogenous TGF-β superfamily antagonist follistatin (FST), a molecule in the IASP class, FSTΔHBS-mFc, was generated and tested in both ovariectomized and naive BALB/c and C57BL/6 mice. In ovariectomized mice, FSTΔHBS-mFc therapy dose-dependently increased cancellous bone mass up to 42% and improved bone microstructural indices. For the highest dosage of FSTΔHBS-mFc (30 mg/kg, 2 times/wk), the increase in cancellous bone mass was similar to that observed with parathyroid hormone therapy (1-34, 80 µg/kg, 5 times/wk). Musculus quadriceps femoris mass dose-dependently increased up to 21% in ovariectomized mice. In both ovariectomized and naive mice, FSTΔHBS-mFc therapy did not influence red blood cell count or hematocrit or hemoglobin levels. If the results are reproduced, a human FSTΔHBS-mFc version could be applicable in patients with musculoskeletal conditions irrespective of hematocrit status.-Lodberg, A., van der Eerden, B. C. J., Boers-Sijmons, B., Thomsen, J. S., Brüel, A., van Leeuwen, J. P. T. M., Eijken, M. A follistatin-based molecule increases muscle and bone mass without affecting the red blood cell count in mice.
Collapse
Affiliation(s)
- Andreas Lodberg
- Department of Pulmonary Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Bianca Boers-Sijmons
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Annemarie Brüel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Marco Eijken
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
20
|
Satkunskiene D, Ratkevicius A, Kamandulis S, Venckunas T. Effects of myostatin on the mechanical properties of muscles during repeated active lengthening in the mouse. Appl Physiol Nutr Metab 2018; 44:381-388. [PMID: 30222937 DOI: 10.1139/apnm-2018-0369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of the present study was to investigate how myostatin dysfunction affects fast and slow muscle stiffness and viscosity during severe repeated loading. Isolated extensor digitorum longus (EDL) and soleus muscles of young adult female mice of the BEH (dysfunctional myostatin) and BEH+/+ (functional myostatin) strains were subjected to 100 contraction-stretching loading cycles during which contractile and mechanical properties were assessed. BEH mice exhibited greater exercise-induced muscle damage, although the effect was muscle- and age-dependent and limited to the early phases of simulated exercise. The relative reduction of the EDL muscle isometric force recorded during the initial 10-30 loading cycles was greater in BEH mice than in BEH+/+ mice and exceeded that of the soleus muscle of either strain. The induced damage was associated with lower muscle stiffness. The effects of myostatin on the mechanical properties of muscles depend on muscle type and maturity.
Collapse
Affiliation(s)
- Danguole Satkunskiene
- Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania.,Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania
| | - Aivaras Ratkevicius
- Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania.,Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania
| | - Sigitas Kamandulis
- Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania.,Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania
| | - Tomas Venckunas
- Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania.,Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania
| |
Collapse
|
21
|
Skeletal muscle mitochondrial bioenergetics and associations with myostatin genotypes in the Thoroughbred horse. PLoS One 2017; 12:e0186247. [PMID: 29190290 PMCID: PMC5708611 DOI: 10.1371/journal.pone.0186247] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/27/2017] [Indexed: 11/19/2022] Open
Abstract
Variation in the myostatin (MSTN) gene has been reported to be associated with race distance, body composition and skeletal muscle fibre composition in the horse. The aim of the present study was to test the hypothesis that MSTN variation influences mitochondrial phenotypes in equine skeletal muscle. Mitochondrial abundance and skeletal muscle fibre types were measured in whole muscle biopsies from the gluteus medius of n = 82 untrained (21 ± 3 months) Thoroughbred horses. Skeletal muscle fibre type proportions were significantly (p < 0.01) different among the three MSTN genotypes and mitochondrial content was significantly (p < 0.01) lower in the combined presence of the C-allele of SNP g.66493737C>T (C) and the SINE insertion 227 bp polymorphism (I). Evaluation of mitochondrial complex activities indicated higher combined mitochondrial complex I+III and II+III activities in the presence of the C-allele / I allele (p ≤ 0.05). The restoration of complex I+III and complex II+III activities following addition of exogenous coenzyme Q1 (ubiquinone1) (CoQ1) in vitro in the TT/NN (homozygous T allele/homozygous no insertion) cohort indicated decreased coenzyme Q in these animals. In addition, decreased gene expression in two coenzyme Q (CoQ) biosynthesis pathway genes (COQ4, p ≤ 0.05; ADCK3, p ≤ 0.01) in the TT/NN horses was observed. This study has identified several mitochondrial phenotypes associated with MSTN genotype in untrained Thoroughbred horses and in addition, our findings suggest that nutritional supplementation with CoQ may aid to restore coenzyme Q activity in TT/NN horses.
Collapse
|
22
|
Follistatin N terminus differentially regulates muscle size and fat in vivo. Exp Mol Med 2017; 49:e377. [PMID: 28912572 PMCID: PMC5628274 DOI: 10.1038/emm.2017.135] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/10/2017] [Accepted: 03/08/2017] [Indexed: 02/07/2023] Open
Abstract
Delivery of follistatin (FST) represents a promising strategy for both muscular dystrophies and diabetes, as FST is a robust antagonist of myostatin and activin, which are critical regulators of skeletal muscle and adipose tissues. FST is a multi-domain protein, and deciphering the function of different domains will facilitate novel designs for FST-based therapy. Our study aims to investigate the role of the N-terminal domain (ND) of FST in regulating muscle and fat mass in vivo. Different FST constructs were created and packaged into the adeno-associated viral vector (AAV). Overexpression of wild-type FST in normal mice greatly increased muscle mass while decreasing fat accumulation, whereas overexpression of an N terminus mutant or N terminus-deleted FST had no effect on muscle mass but moderately decreased fat mass. In contrast, FST-I-I containing the complete N terminus and double domain I without domain II and III had no effect on fat but increased skeletal muscle mass. The effects of different constructs on differentiated C2C12 myotubes were consistent with the in vivo finding. We hypothesized that ND was critical for myostatin blockade, mediating the increase in muscle mass, and was less pivotal for activin binding, which accounts for the decrease in the fat tissue. An in vitro TGF-beta1-responsive reporter assay revealed that FST-I-I and N terminus-mutated or -deleted FST showed differential responses to blockade of activin and myostatin. Our study provided direct in vivo evidence for a role of the ND of FST, shedding light on future potential molecular designs for FST-based gene therapy.
Collapse
|
23
|
Yeh YC, Kinoshita M, Ng TH, Chang YH, Maekawa S, Chiang YA, Aoki T, Wang HC. Using CRISPR/Cas9-mediated gene editing to further explore growth and trade-off effects in myostatin-mutated F4 medaka (Oryzias latipes). Sci Rep 2017; 7:11435. [PMID: 28900124 PMCID: PMC5595883 DOI: 10.1038/s41598-017-09966-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 08/02/2017] [Indexed: 11/21/2022] Open
Abstract
Myostatin (MSTN) suppresses skeletal muscle development and growth in mammals, but its role in fish is less well understood. Here we used CRISPR/Cas9 to mutate the MSTN gene in medaka (Oryzias latipes) and evaluate subsequent growth performance. We produced mutant F0 fish that carried different frameshifts in the OlMSTN coding sequence and confirmed the heritability of the mutant genotypes to the F1 generation. Two F1 fish with the same heterozygous frame-shifted genomic mutations (a 22 bp insertion in one allele; a 32 bp insertion in the other) were then crossbred to produce subsequent generations (F2~F5). Body length and weight of the MSTN-/- F4 medaka were significantly higher than in the wild type fish, and muscle fiber density in the inner and outer compartments of the epaxial muscles was decreased, suggesting that MSTN null mutation induces muscle hypertrophy. From 3~4 weeks post hatching (wph), the expression of three major myogenic related factors (MRFs), MyoD, Myf5 and Myogenin, was also significantly upregulated. Some medaka had a spinal deformity, and we also observed a trade-off between growth and immunity in MSTN-/- F4 medaka. Reproduction was unimpaired in the fast-growth phenotypes.
Collapse
Affiliation(s)
- Ying-Chun Yeh
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Masato Kinoshita
- Division of Applied Bioscience, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Tze Hann Ng
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Yu-Hsuan Chang
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Shun Maekawa
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Yi-An Chiang
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Takashi Aoki
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Han-Ching Wang
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan.
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
24
|
Little RD, Prieto-Potin I, Pérez-Baos S, Villalvilla A, Gratal P, Cicuttini F, Largo R, Herrero-Beaumont G. Compensatory anabolic signaling in the sarcopenia of experimental chronic arthritis. Sci Rep 2017; 7:6311. [PMID: 28740214 PMCID: PMC5524910 DOI: 10.1038/s41598-017-06581-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 06/14/2017] [Indexed: 01/06/2023] Open
Abstract
Inflammatory activity in rheumatoid arthritis may alter the regulation of muscle mass leading to a secondary sarcopenia, commonly termed rheumatoid cachexia (RC). We characterized alterations to muscle structure and various pro-inflammatory, catabolic and regenerative markers in an animal model of RC. Antigen induced arthritis (AiA) was performed in 20 male adult rabbits. AiA animals exhibited significantly less weight gain, a markedly elevated serum C-reactive protein (CRP), lighter muscles with shorter cross-sectional diameter and increased myonuclei when compared to controls. Atrogin-1 and MuRF-1 were up-regulated alongside an increase in IL-1β, active NF-κB and a higher ratio of phosphorylated to inactive p38 MAPK. CCL-2 and TNF levels were reduced and IL-6 was unchanged between groups. We observed decreased pSTAT3, unchanged pSTAT1 and Myf5, but increased Pax7, MyoD and myogenin. AiA rabbits had a reduction in myostatin from gastrocnemii and synovium with a congruent decrease in serum myostatin compared to controls. Chronic arthritis induced an RC-like secondary sarcopenia with increased muscle protein breakdown. Elevated IL-1β may trigger proteolysis via elevated NF-κB and p38 MAPK signaling with a compensatory anabolic response suggested by myonuclear expansion, increased Pax7, MyoD and myogenin, reduced pSTAT3 as well as reduced serum, synovial and muscular myostatin.
Collapse
Affiliation(s)
- Robert D Little
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Alfred Hospital, Melbourne, VIC 3004, Australia
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Iván Prieto-Potin
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
- Red Temática de Investigación Cooperativa de Envejecimiento y Fragilidad (RETICEF)-Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Pérez-Baos
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Amanda Villalvilla
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Paula Gratal
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Flavia Cicuttini
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Raquel Largo
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain.
- Red Temática de Investigación Cooperativa de Envejecimiento y Fragilidad (RETICEF)-Instituto de Salud Carlos III, Madrid, Spain.
| | - Gabriel Herrero-Beaumont
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
- Red Temática de Investigación Cooperativa de Envejecimiento y Fragilidad (RETICEF)-Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
25
|
Kumar R, Singh SP, Mitra A. Short-hairpin Mediated Myostatin Knockdown Resulted in Altered Expression of Myogenic Regulatory Factors with Enhanced Myoblast Proliferation in Fetal Myoblast Cells of Goats. Anim Biotechnol 2017; 29:59-67. [PMID: 28358646 DOI: 10.1080/10495398.2017.1299744] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Myostatin (MSTN) is a well-known negative regulator of skeletal muscle development. Reduced expression due to natural mutations in the coding region and knockout as well as knockdown of MSTN results in an increase in the muscle mass. In the present study, we demonstrated as high as 60 and 52% downregulation (p < 0.01) of MSTN mRNA and protein in the primary fetal myoblast cells of goats using synthetic shRNAs (n = 3), without any interferon response. We, for the first time, evaluated the effect of MSTN knockdown on the expression of MRFs (namely, MyoD, Myf5), follistatin (FST), and IGFs (IGF-1 & IGF-2) in goat myoblast cells. MSTN knockdown caused an upregulation (p < 0.05) of MyoD and downregulation (p < 0.01) of MYf5 and FST expression. Moreover, we report up to ∼four fold (p < 0.001) enhanced proliferation in myoblasts after four days of culture. The anti-MSTN shRNA demonstrated in the present study could be used for the production of transgenic goats to increase the muscle mass.
Collapse
Affiliation(s)
- Rohit Kumar
- a Genome Analysis Laboratory, Animal Genetics Division , ICAR- Indian Veterinary Research Institute , Izatnagar , Bareilly , India
| | - Satyendra Pal Singh
- a Genome Analysis Laboratory, Animal Genetics Division , ICAR- Indian Veterinary Research Institute , Izatnagar , Bareilly , India
| | - Abhijit Mitra
- a Genome Analysis Laboratory, Animal Genetics Division , ICAR- Indian Veterinary Research Institute , Izatnagar , Bareilly , India
| |
Collapse
|
26
|
Nielsen C, Potter RM, Borowy C, Jacinto K, Kumar R, Carlson CG. Postnatal Hyperplasic Effects of ActRIIB Blockade in a Severely Dystrophic Muscle. J Cell Physiol 2017; 232:1774-1793. [PMID: 27859236 DOI: 10.1002/jcp.25694] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 11/16/2016] [Indexed: 01/26/2023]
Abstract
The efficacy of two ActRIIB ligand-trapping agents (RAP-031 and RAP-435) in treating muscular dystrophy was examined by determining their morphological effects on the severely dystrophic triangularis sterni (TS) muscle of the mdx mouse, a model for Duchenne muscular dystrophy. These agents trap all endogenous ligands to the ActRIIB receptor and thereby block myostatin signaling in a highly selective manner. Short-term (1 month) and long-term (3 months) in vivo treatment of 1-month-old mdx mice increased myonuclei and fiber cross section (FCS) density but did not alter individual fiber size. Vehicle-treated mdx mice exhibited age-dependent increases in myonuclei and FCS density, and age-dependent reductions in centronucleation that were each enhanced by treatment with RAP-435. Distributions of FCS area (FCSA) in the mdx TS were 90% identical to those from untreated age-matched nondystrophic mice and were unaltered by the substantial fiber hyperplasia observed with age and RAP-435 treatment. These results were inconsistent with injury-induced fiber regeneration which produces altered FCSA distributions characterized by a distinct class of smaller regenerated fibers. Nondystrophic mice exhibited a constant postnatal density of fiber cross sections and myonuclei, and RAP-435 treatment of nondystrophic mice increased TS mean FCSA but had no effects on myonuclei or FCS density. These results demonstrating a continual postnatal proliferation and fusion of satellite cells and a response to myostatin blockade characteristic of developing prenatal muscle suggest that the lack of dystrophin directly results in unrestrained postnatal satellite cell activation that is not necessarily dependent upon prior fiber degeneration. J. Cell. Physiol. 232: 1774-1793, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cory Nielsen
- Department of Physiology, Midwestern University Glendale, Glendale, Arizona
| | - Ross M Potter
- Department of Physiology, Midwestern University Glendale, Glendale, Arizona
| | - Christopher Borowy
- Department of Physiology, Midwestern University Glendale, Glendale, Arizona
| | - Kimberly Jacinto
- Department of Physiology, Midwestern University Glendale, Glendale, Arizona
| | - Ravi Kumar
- Acceleron Pharma, Inc., Cambridge, Massachusetts
| | - C George Carlson
- Department of Physiology, Midwestern University Glendale, Glendale, Arizona
| |
Collapse
|
27
|
Dushyanth K, Bhattacharya TK, Shukla R, Chatterjee RN, Sitaramamma T, Paswan C, Guru Vishnu P. Gene Expression and Polymorphism of Myostatin Gene and its Association with Growth Traits in Chicken. Anim Biotechnol 2017; 27:269-77. [PMID: 27565871 DOI: 10.1080/10495398.2016.1182541] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Myostatin is a member of TGF-β super family and is directly involved in regulation of body growth through limiting muscular growth. A study was carried out in three chicken lines to identify the polymorphism in the coding region of the myostatin gene through SSCP and DNA sequencing. A total of 12 haplotypes were observed in myostatin coding region of chicken. Significant associations between haplogroups with body weight at day 1, 14, 28, and 42 days, and carcass traits at 42 days were observed across the lines. It is concluded that the coding region of myostatin gene was polymorphic, with varied levels of expression among lines and had significant effects on growth traits. The expression of MSTN gene varied during embryonic and post hatch development stage.
Collapse
Affiliation(s)
- K Dushyanth
- a Poultry Research , Rajendranagar, Hyderabad , India
| | | | - R Shukla
- a Poultry Research , Rajendranagar, Hyderabad , India
| | | | - T Sitaramamma
- a Poultry Research , Rajendranagar, Hyderabad , India
| | - C Paswan
- a Poultry Research , Rajendranagar, Hyderabad , India
| | - P Guru Vishnu
- a Poultry Research , Rajendranagar, Hyderabad , India
| |
Collapse
|
28
|
Reilly BD, Franklin CE. Prevention of muscle wasting and osteoporosis: the value of examining novel animal models. J Exp Biol 2016; 219:2582-95. [DOI: 10.1242/jeb.128348] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
ABSTRACT
Bone mass and skeletal muscle mass are controlled by factors such as genetics, diet and nutrition, growth factors and mechanical stimuli. Whereas increased mechanical loading of the musculoskeletal system stimulates an increase in the mass and strength of skeletal muscle and bone, reduced mechanical loading and disuse rapidly promote a decrease in musculoskeletal mass, strength and ultimately performance (i.e. muscle atrophy and osteoporosis). In stark contrast to artificially immobilised laboratory mammals, animals that experience natural, prolonged bouts of disuse and reduced mechanical loading, such as hibernating mammals and aestivating frogs, consistently exhibit limited or no change in musculoskeletal performance. What factors modulate skeletal muscle and bone mass, and what physiological and molecular mechanisms protect against losses of muscle and bone during dormancy and following arousal? Understanding the events that occur in different organisms that undergo natural periods of prolonged disuse and suffer negligible musculoskeletal deterioration could not only reveal novel regulatory factors but also might lead to new therapeutic options. Here, we review recent work from a diverse array of species that has revealed novel information regarding physiological and molecular mechanisms that dormant animals may use to conserve musculoskeletal mass despite prolonged inactivity. By highlighting some of the differences and similarities in musculoskeletal biology between vertebrates that experience disparate modes of dormancy, it is hoped that this Review will stimulate new insights and ideas for future studies regarding the regulation of atrophy and osteoporosis in both natural and clinical models of muscle and bone disuse.
Collapse
Affiliation(s)
- Beau D. Reilly
- School of Biological Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Craig E. Franklin
- School of Biological Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
29
|
Monestier O, Blanquet V. WFIKKN1 and WFIKKN2: "Companion" proteins regulating TGFB activity. Cytokine Growth Factor Rev 2016; 32:75-84. [PMID: 27325460 DOI: 10.1016/j.cytogfr.2016.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 01/14/2023]
Abstract
The WFIKKN (WAP, Follistatin/kazal, Immunoglobulin, Kunitz and Netrin domain-containing) protein family is composed of two multidomain proteins: WFIKKN1 and WFIKKN2. They were formed by domain shuffling and are likely present in deuterostoms. The WFIKKN (also called GASP) proteins are well known for their function in muscle and skeletal tissues, namely, inhibition of certain members of the transforming growth factor beta (TGFB) superfamily such as myostatin (MSTN) and growth and differentiation factor 11 (GDF11). However, the role of the WFIKKN proteins in other tissues is still poorly understood in spite of evidence suggesting possible action in the inner ear, brain and reproduction. Further, several recent studies based on next generation technologies revealed differential expression of WFIKKN1 and WFIKKN2 in various tissues suggesting that their function is not limited to MSTN and GDF11 inhibition in musculoskeletal tissue. In this review, we summarize current knowledge about the WFIKKN proteins and propose that they are "companion" proteins for various growth factors by providing localized and sustained presentation of TGFB proteins to their respective receptors, thus regulating the balance between the activation of Smad and non-Smad pathways by TGFB.
Collapse
Affiliation(s)
- Olivier Monestier
- INRA, UR1037 Laboratory of Fish Physiology and Genomic, Growth and Flesh Quality Group, Campus de Beaulieu, 35000 Rennes, France.
| | - Véronique Blanquet
- INRA, UMR1061 Unité de Génétique Moléculaire Animale, 87060 Limoges, France; Université de Limoges, 87060 Limoges, France.
| |
Collapse
|
30
|
Gu H, Cao Y, Qiu B, Zhou Z, Deng R, Chen Z, Li R, Li X, Wei Q, Xia X, Yong W. Establishment and phenotypic analysis of an Mstn knockout rat. Biochem Biophys Res Commun 2016; 477:115-122. [PMID: 27289021 DOI: 10.1016/j.bbrc.2016.06.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 06/08/2016] [Indexed: 01/01/2023]
Abstract
Myostatin (Mstn) is an inhibitor of myogenesis, regulating the number and size of skeletal myocytes. In addition to its myogenic regulatory function, Mstn plays important roles in the development of adipose tissues and in metabolism. In the present study, an Mstn knockout rat model was generated using the zinc finger nuclease (ZFN) technique in order to further investigate the function and mechanism of Mstn in metabolism. The knockout possesses a frame shift mutation resulting in an early termination codon and a truncated peptide of 109 amino acids rather than the full 376 amino acids. The absence of detectable mRNA confirmed successful knockout of Mstn. Relative to wild-type (WT) littermates, Knockout (KO) rats exhibited significantly greater body weight, body circumference, and muscle mass. However, no significant differences in grip force was observed, indicating that Mstn deletion results in greater muscle mass but not greater muscle fiber strength. Additionally, KO rats were found to possess less body fat relative to WT littermates, which is consistent with previous studies in mice and cattle. The aforementioned results indicate that Mstn knockout increases muscle mass while decreasing fat content, leading to observed increases in body weight and body circumference. The Mstn knockout rat model provides a novel means to study the role of Mstn in metabolism and Mstn-related muscle hypertrophy.
Collapse
Affiliation(s)
- Hao Gu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Yong Cao
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China; Experimental Medicine Center, The First Affiliated Hospital of Sichuan Medical University, Sichuan 646000, China
| | - Bin Qiu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Zhiqiang Zhou
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Ran Deng
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Zhuang Chen
- Experimental Medicine Center, The First Affiliated Hospital of Sichuan Medical University, Sichuan 646000, China
| | - Rongfeng Li
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot 010021, China
| | - Xueling Li
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot 010021, China
| | - Qiang Wei
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun 130122, China.
| | - Weidong Yong
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
31
|
Dewey CM, Spitler KM, Ponce JM, Hall DD, Grueter CE. Cardiac-Secreted Factors as Peripheral Metabolic Regulators and Potential Disease Biomarkers. J Am Heart Assoc 2016; 5:e003101. [PMID: 27247337 PMCID: PMC4937259 DOI: 10.1161/jaha.115.003101] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Colleen M Dewey
- Division of Cardiovascular Medicine, Department of Internal Medicine and François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Kathryn M Spitler
- Division of Cardiovascular Medicine, Department of Internal Medicine and François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Jessica M Ponce
- Division of Cardiovascular Medicine, Department of Internal Medicine and François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Duane D Hall
- Division of Cardiovascular Medicine, Department of Internal Medicine and François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Chad E Grueter
- Division of Cardiovascular Medicine, Department of Internal Medicine and François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA Fraternal Order of Eagles Diabetes Research Center, Papajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|
32
|
Identification of Deleterious Mutations in Myostatin Gene of Rohu Carp (Labeo rohita) Using Modeling and Molecular Dynamic Simulation Approaches. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7562368. [PMID: 27019850 PMCID: PMC4785247 DOI: 10.1155/2016/7562368] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 01/13/2016] [Accepted: 01/27/2016] [Indexed: 11/18/2022]
Abstract
The myostatin (MSTN) is a known negative growth regulator of skeletal muscle. The mutated myostatin showed a double-muscular phenotype having a positive significance for the farmed animals. Consequently, adequate information is not available in the teleosts, including farmed rohu carp, Labeo rohita. In the absence of experimental evidence, computational algorithms were utilized in predicting the impact of point mutation of rohu myostatin, especially its structural and functional relationships. The four mutations were generated at different positions (p.D76A, p.Q204P, p.C312Y, and p.D313A) of MSTN protein of rohu. The impacts of each mutant were analyzed using SIFT, I-Mutant 2.0, PANTHER, and PROVEAN, wherein two substitutions (p.D76A and p.Q204P) were predicted as deleterious. The comparative structural analysis of each mutant protein with the native was explored using 3D modeling as well as molecular-dynamic simulation techniques. The simulation showed altered dynamic behaviors concerning RMSD and RMSF, for either p.D76A or p.Q204P substitution, when compared with the native counterpart. Interestingly, incorporated two mutations imposed a significant negative impact on protein structure and stability. The present study provided the first-hand information in identifying possible amino acids, where mutations could be incorporated into MSTN gene of rohu carp including other carps for undertaking further in vivo studies.
Collapse
|
33
|
Shin S, Song Y, Ahn J, Kim E, Chen P, Yang S, Suh Y, Lee K. A novel mechanism of myostatin regulation by its alternative splicing variant during myogenesis in avian species. Am J Physiol Cell Physiol 2015; 309:C650-9. [PMID: 26354750 DOI: 10.1152/ajpcell.00187.2015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/02/2015] [Indexed: 11/22/2022]
Abstract
Myostatin (MSTN) is a key negative regulator of muscle growth and development, and an increase of muscle mass is achieved by inhibiting MSTN signaling. In the current study, five alternative splicing isoforms of MSTN mRNAs in avian species were identified in various tissues. Among these five, three truncated forms of myostatin, MSTN-B, -C, and -E created premature stop codons and produced partial MSTN prodomains encoded from exon 1. MSTN-B is the second dominant isoform following full-length MSTN-A, and their expression was dynamically regulated during muscle development of chicken, turkey, and quail in vivo and in vitro. To clarify the function of MSTN-B, two stable cell lines of quail myoblasts (QM7) were generated to overexpress MSTN-A or MSTN-B. Interestingly, MSTN-B promoted both cell proliferation and differentiation similar to the function of the MSTN prodomain to counteract the negative role of MSTN on myogenesis. The coimmunoprecipitation assay revealed that MSTN-B binds to MSTN-A and reduces the generation of mature MSTN. Furthermore, the current study demonstrated that the partial prodomain encoded from exon 1 is critical for binding of MSTN-B to MSTN-A. Altogether, these data imply that alternative splicing isoforms of MSTN could negatively regulate pro-myostatin processing in muscle cells and prevent MSTN-mediated inhibition of myogenesis in avian species.
Collapse
Affiliation(s)
- Sangsu Shin
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio; Department of Animal Biotechnology, Kyungpook National University, Sangju, Gyeongbuk, Republic of Korea; and
| | - Yan Song
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio
| | - Jinsoo Ahn
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio; Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, Ohio
| | - Eunsoo Kim
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio
| | - Paula Chen
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio
| | - Shujin Yang
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio
| | - Yeunsu Suh
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio
| | - Kichoon Lee
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio; Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, Ohio
| |
Collapse
|
34
|
Sharma M, McFarlane C, Kambadur R, Kukreti H, Bonala S, Srinivasan S. Myostatin: expanding horizons. IUBMB Life 2015; 67:589-600. [PMID: 26305594 DOI: 10.1002/iub.1392] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 05/29/2015] [Indexed: 12/13/2022]
Abstract
Myostatin is a secreted growth and differentiation factor that belongs to the TGF-β superfamily. Myostatin is predominantly synthesized and expressed in skeletal muscle and thus exerts a huge impact on muscle growth and function. In keeping with its negative role in myogenesis, myostatin expression is tightly regulated at several levels including epigenetic, transcriptional, post-transcriptional, and post-translational. New revelations regarding myostatin regulation also offer mechanisms that could be exploited for developing myostatin antagonists. Increasingly, it is becoming clearer that besides its conventional role in muscle, myostatin plays a critical role in metabolism. Hence, molecular mechanisms by which myostatin regulates several key metabolic processes need to be further explored.
Collapse
Affiliation(s)
- Mridula Sharma
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
- Department of Cell & Molecular Biology, Brenner Centre for Molecular Medicine, Singapore Institute of Clinical Sciences (SICS), Singapore
| | - Craig McFarlane
- Department of Cell & Molecular Biology, Brenner Centre for Molecular Medicine, Singapore Institute of Clinical Sciences (SICS), Singapore
| | - Ravi Kambadur
- Department of Cell & Molecular Biology, Brenner Centre for Molecular Medicine, Singapore Institute of Clinical Sciences (SICS), Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Himani Kukreti
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
| | - Sabeera Bonala
- Department of Cell & Molecular Biology, Brenner Centre for Molecular Medicine, Singapore Institute of Clinical Sciences (SICS), Singapore
| | - Shruti Srinivasan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
| |
Collapse
|
35
|
Ma D, Jiang S, Gao P, Qian L, Wang Q, Cai C, Xiao G, Yang J, Cui W. Functional verification of a porcine myostatin propeptide mutant. Transgenic Res 2015; 24:837-45. [DOI: 10.1007/s11248-015-9896-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 07/07/2015] [Indexed: 10/23/2022]
|
36
|
AAV2-mediated follistatin overexpression induces ovine primary myoblasts proliferation. BMC Biotechnol 2014; 14:87. [PMID: 25330993 PMCID: PMC4210504 DOI: 10.1186/s12896-014-0087-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 10/06/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Follistatin (FST) has been shown to bind to some TGF-β family members and can function as a potent myostatin (MSTN) antagonist. Recent studies have revealed that over-expression of FST by adeno-associated viruses increases muscle growth in mice, humans and nonhuman primates. In the present study, to determine the effect of FST on ovine primary myoblast (OPM) proliferation, FST was over-expressed using an adeno-associated virus serotype 2 (AAV 2) vector. RESULTS Western blot results showed that AAV induced the expression of FST protein in transduced OPM cells. Real-time quantitative PCR results indicated that over-expression of FST resulted in a dramatic increase in Akt I and CDK2 expression and a decrease in p21 expression. Moreover, cell cycle analysis confirmed that FST down-regulated p21, a CDK inhibitor, and increased the level of CDK2 expression in OPM cells. Hence, follistatin positively regulated the G1 to S progression. Our results showed that FST induced proliferation through a down-regulation of p21, as only the p21 expression level was down-regulated as a result of FST over-expression in myoblasts, whereas no change was observed in the level of p57 expression. CONCLUSIONS These results expanded our understanding of the regulation mechanism of FST in ovine primary myoblasts. Our results provide the first evidence that the AAV viral system can be used for gene transfer in ovine myoblast cells. Moreover, the results showed that an AAV vector can successfully induce the expression of FST in OPM cells in vitro. These findings demonstrated that FST over-expression induces proliferation through a down-regulation of the p21 gene under proliferating conditions.
Collapse
|
37
|
Zhong B, Zhang Y, Yan Y, Wang Z, Ying S, Huang M, Wang F. MicroRNA-mediated myostatin silencing in caprine fetal fibroblasts. PLoS One 2014; 9:e107071. [PMID: 25244645 PMCID: PMC4171098 DOI: 10.1371/journal.pone.0107071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 08/12/2014] [Indexed: 12/13/2022] Open
Abstract
Myostatin functions as a negative regulator of skeletal muscle growth by suppressing proliferation and differentiation of myoblasts. Dysfunction of the myostatin gene, either due to natural mutation or genetic manipulations such as knockout or knockdown, has been reported to increase muscle mass in mammalian species. RNA interference (RNAi) mediated by microRNAs (miRNAs) is a promising method for gene knockdown studies. In the present study, transient and stable silencing of the myostatin gene in caprine fetal fibroblasts (CFF) was evaluated using the two most effective constructs selected from four different miRNA expression constructs screened in 293FT cells. Using these two miRNA constructs, we achieved up to 84% silencing of myostatin mRNA in transiently transfected CFF cells and up to 31% silencing in stably transfected CFF cells. Moreover, off-target effects due to induction of interferon (IFN) response genes, such as interferon beta (IFN-β) and 2′-5′-oligoadenylate synthetase 2 (OAS2), were markedly fewer in stably transfected CFF cells than in transiently transfected cells. Stable expression of anti-myostatin miRNA with minimal induction of interferon shows great promise for increasing muscle mass in transgenic goats.
Collapse
Affiliation(s)
- Bushuai Zhong
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, PR China
- Jiangsu Engineering Technology Research Center of Meat Sheep & Goat Industry, Nanjing Agricultural University, Nanjing, PR China
| | - Yanli Zhang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, PR China
- Jiangsu Engineering Technology Research Center of Meat Sheep & Goat Industry, Nanjing Agricultural University, Nanjing, PR China
| | - Yibo Yan
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, PR China
| | - Ziyu Wang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, PR China
| | - Shijia Ying
- Jiangsu Engineering Technology Research Center of Meat Sheep & Goat Industry, Nanjing Agricultural University, Nanjing, PR China
| | - Mingrui Huang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, PR China
- * E-mail: (MH); (FW)
| | - Feng Wang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, PR China
- Jiangsu Engineering Technology Research Center of Meat Sheep & Goat Industry, Nanjing Agricultural University, Nanjing, PR China
- * E-mail: (MH); (FW)
| |
Collapse
|
38
|
Mori H, Kawai N, Kinouchi N, Hichijo N, Ishida T, Kawakami E, Noji S, Tanaka E. Effectiveness of cationic liposome-mediated local delivery of myostatin-targeting small interfering RNA in vivo. Dev Growth Differ 2014; 56:223-32. [PMID: 24621004 DOI: 10.1111/dgd.12123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/12/2014] [Accepted: 01/12/2014] [Indexed: 11/28/2022]
Abstract
This study evaluated the effectiveness of local administration of cationic liposome-delivered myostatin-targeting siRNA. Myostatin (Mst)-siRNA and scrambled (scr)-siRNA-lipoplexes were injected into the masseter muscles of wild type and dystrophin-deficient mdx mice, which model Duchenne muscular dystrophy. One week after injection, the masseter muscles were dissected for histometric analyses. To evaluate changes in masseter muscle activity, masseter electromyographic (EMG) measurements were performed. One week after local administration of Mst-siRNA-lipoplexes, masseter muscles and myofibrils were significantly larger compared to control masseter muscles treated with scr-siRNA-lipoplexes. Real-time polymerase chain reaction (PCR) analyses revealed significant upregulation of the myogenic regulatory factors MyoD and myogenin and significant downregulation of the adipogenic transcription factors peroxisome proliferator-activated receptor-γ (PPARγ) and CCAAT/enhancer binding protein-α (CEBPα) in masseter muscles treated with Mst-siRNA-lipoplexes. The duty times of masseter muscle activity exceeding 5% showed a slight tendency to increase in both wild type and mdx mice. Therefore, cationic liposome-mediated local administration of Mst-siRNA could increase muscular size and improve muscle activity. Since cationic liposomes delivered siRNA to muscles effectively and are safe and cost-effective, they may represent a therapeutic tool for use in treating muscular diseases.
Collapse
Affiliation(s)
- Hiroyo Mori
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Health Biosciences, The University of Tokushima Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8504, Japan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Li X, Wang SJ, Tan SC, Chew PL, Liu L, Wang L, Wen L, Ma L. The A55T and K153R polymorphisms ofMSTNgene are associated with the strength training-induced muscle hypertrophy among Han Chinese men. J Sports Sci 2014; 32:883-91. [DOI: 10.1080/02640414.2013.865252] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
40
|
Paswan C, Bhattacharya T, Nagaraj C, Chaterjee R, Jayashankar M. SNPs in minimal promoter of myostatin (GDF-8) gene and its association with body weight in broiler chicken. JOURNAL OF APPLIED ANIMAL RESEARCH 2013. [DOI: 10.1080/09712119.2013.846859] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
41
|
Kawakami E, Kawai N, Kinouchi N, Mori H, Ohsawa Y, Ishimaru N, Sunada Y, Noji S, Tanaka E. Local applications of myostatin-siRNA with atelocollagen increase skeletal muscle mass and recovery of muscle function. PLoS One 2013; 8:e64719. [PMID: 23717655 PMCID: PMC3661523 DOI: 10.1371/journal.pone.0064719] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 04/17/2013] [Indexed: 12/25/2022] Open
Abstract
Background Growing evidence suggests that small-interfering RNA (siRNA) can promote gene silencing in mammalian cells without induction of interferon synthesis or nonspecific gene suppression. Recently, a number of highly specific siRNAs targeted against disease-causing or disease-promoting genes have been developed. In this study, we evaluate the effectiveness of atelocollagen (ATCOL)-mediated application of siRNA targeting myostatin (Mst), a negative regulator of skeletal muscle growth, into skeletal muscles of muscular dystrophy model mice. Methods and Findings We injected a nanoparticle complex containing myostatin-siRNA and ATCOL (Mst-siRNA/ATCOL) into the masseter muscles of mutant caveolin-3 transgenic (mCAV-3Tg) mice, an animal model for muscular dystrophy. Scrambled (scr) -siRNA/ATCOL complex was injected into the contralateral muscles as a control. Two weeks after injection, the masseter muscles were dissected for histometric analyses. To investigate changes in masseter muscle activity by local administration of Mst-siRNA/ATCOL complex, mouse masseter electromyography (EMG) was measured throughout the experimental period via telemetry. After local application of the Mst-siRNA/ATCOL complex, masseter muscles were enlarged, while no significant change was observed on the contralateral side. Histological analysis showed that myofibrils of masseter muscles treated with the Mst-siRNA/ATCOL complex were significantly larger than those of the control side. Real-time PCR analysis revealed a significant downregulation of Mst expression in the treated masseters of mCAV-3Tg mice. In addition, expression of myogenic transcription factors was upregulated in the Mst-siRNA-treated masseter muscle, while expression of adipogenic transcription factors was significantly downregulated. EMG results indicate that masseter muscle activity in mCAV-3Tg mice was increased by local administration of the Mst-siRNA/ATCOL complex. Conclusion These data suggest local administration of Mst-siRNA/ATCOL complex could lead to skeletal muscle hypertrophy and recovery of motor disability in mCAV-3Tg mice. Therefore, ATCOL-mediated application of siRNA is a potential tool for therapeutic use in muscular atrophy diseases.
Collapse
Affiliation(s)
- Emi Kawakami
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Nobuhiko Kawai
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Nao Kinouchi
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Hiroyo Mori
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Yutaka Ohsawa
- Department of Neurology, Kawasaki Medical School, Okayama, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Yoshihide Sunada
- Department of Neurology, Kawasaki Medical School, Okayama, Japan
| | - Sumihare Noji
- Department of Life Systems, Institute of Technology and Science, The University of Tokushima, Tokushima, Japan
| | - Eiji Tanaka
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
- * E-mail:
| |
Collapse
|
42
|
Crook M, Grant WN. Dominant negative mutations of Caenorhabditis elegans daf-7 confer a novel developmental phenotype. Dev Dyn 2013; 242:654-64. [PMID: 23526825 DOI: 10.1002/dvdy.23963] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 02/27/2013] [Accepted: 03/05/2013] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND TGF-β signaling pathways are involved in the control of development in every member of the animal kingdom. As such, TGF-β ligands are widely divergent yet retain a set of core conserved features, specifically, a pre-protein cleavage site and several conserved ligand domain residues, the disruption of which produces a dominant negative phenotype. RESULTS We have extended these observations into an invertebrate system by creating a series of loss-of-function Caenorhabditis elegans daf-7 transgenes. When we tested these mutant transgenes in a daf-7/+ background, we saw a molting and excretory canal phenotype. Members of both pathways downstream of daf-4 were required for this phenotype. CONCLUSIONS Our results show that the basic mechanisms of TGF-β function are conserved across the animal kingdom. A subset of our daf-7 mutations also produced an unexpected and novel phenotype. Epistasis experiments demonstrated that both daf-3/-5 and sma-4/-9 were downstream of our mutant daf-7 transgenes, which suggests not only a role for DAF-7 in the control of molting and the development of the excretory system but also that daf-7 and dbl-1 signaling may converge downstream of their shared Type II receptor, daf-4. Our approach may unveil new roles in development for other invertebrate TGF-β ligands.
Collapse
Affiliation(s)
- Matt Crook
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, USA.
| | | |
Collapse
|
43
|
Hu S, Ni W, Sai W, Zi H, Qiao J, Wang P, Sheng J, Chen C. Knockdown of myostatin expression by RNAi enhances muscle growth in transgenic sheep. PLoS One 2013; 8:e58521. [PMID: 23526994 PMCID: PMC3603981 DOI: 10.1371/journal.pone.0058521] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 02/07/2013] [Indexed: 11/18/2022] Open
Abstract
Myostatin (MSTN) has been shown to be a negative regulator of skeletal muscle development and growth. MSTN dysfunction therefore offers a strategy for promoting animal growth performance in livestock production. In this study, we investigated the possibility of using RNAi-based technology to generate transgenic sheep with a double-muscle phenotype. A shRNA expression cassette targeting sheep MSTN was used to generate stable shRNA-expressing fibroblast clones. Transgenic sheep were further produced by somatic cell nuclear transfer (SCNT) technology. Five lambs developed to term and three live lambs were obtained. Integration of shRNA expression cassette in three live lambs was confirmed by PCR. RNase protection assay showed that the shRNAs targeting MSTN were expressed in muscle tissues of three transgenic sheep. MSTN expression was significantly inhibited in muscle tissues of transgenic sheep when compared with control sheep. Moreover, transgenic sheep showed a tendency to faster increase in body weight than control sheep. Histological analysis showed that myofiber diameter of transgenic sheep M17 were bigger than that of control sheep. Our findings demonstrate a promising approach to promoting muscle growth in livestock production.
Collapse
Affiliation(s)
- Shengwei Hu
- College of Animal Science and Technology, Shihezi University, Shihezi, China
- Key Laboratory of Agrobiotechnology, Shihezi University, Shihezi, China
| | - Wei Ni
- College of Animal Science and Technology, Shihezi University, Shihezi, China
- Key Laboratory of Agrobiotechnology, Shihezi University, Shihezi, China
| | - Wujiafu Sai
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Ha Zi
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Jun Qiao
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Pengyang Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Jinliang Sheng
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Chuangfu Chen
- College of Animal Science and Technology, Shihezi University, Shihezi, China
- Key Laboratory of Agrobiotechnology, Shihezi University, Shihezi, China
- * E-mail:
| |
Collapse
|
44
|
Iwasaki S, Miyake M, Hayashi S, Watanabe H, Nagasawa Y, Terada S, Watanabe K, Ohwada S, Kitazawa H, Rose MT, Aso H. Effect of Myostatin on Chemokine Expression in Regenerating Skeletal Muscle Cells. Cells Tissues Organs 2013; 198:66-74. [DOI: 10.1159/000351462] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2013] [Indexed: 11/19/2022] Open
|
45
|
Malerba A, Kang JK, McClorey G, Saleh AF, Popplewell L, Gait MJ, Wood MJ, Dickson G. Dual Myostatin and Dystrophin Exon Skipping by Morpholino Nucleic Acid Oligomers Conjugated to a Cell-penetrating Peptide Is a Promising Therapeutic Strategy for the Treatment of Duchenne Muscular Dystrophy. MOLECULAR THERAPY. NUCLEIC ACIDS 2012; 1:e62. [PMID: 23250360 PMCID: PMC3528303 DOI: 10.1038/mtna.2012.54] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The knockdown of myostatin, a negative regulator of skeletal muscle mass may have
important implications in disease conditions accompanied by muscle mass loss like cancer,
HIV/AIDS, sarcopenia, muscle atrophy, and Duchenne muscular dystrophy (DMD). In DMD
patients, where major muscle loss has occurred due to a lack of dystrophin, the
therapeutic restoration of dystrophin expression alone in older patients may not be
sufficient to restore the functionality of the muscles. We recently demonstrated that
phosphorodiamidate morpholino oligomers (PMOs) can be used to re-direct myostatin splicing
and promote the expression of an out-of-frame transcript so reducing the amount of the
synthesized myostatin protein. Furthermore, the systemic administration of the same PMO
conjugated to an octaguanidine moiety (Vivo-PMO) led to a significant increase in the mass
of soleus muscle of treated mice. Here, we have further optimized the use of Vivo-PMO in
normal mice and also tested the efficacy of the same PMO conjugated to an arginine-rich
cell-penetrating peptide (B-PMO). Similar experiments conducted in mdx dystrophic mice
showed that B-PMO targeting myostatin is able to significantly increase the tibialis
anterior (TA) muscle weight and when coadministered with a B-PMO targeting the dystrophin
exon 23, it does not have a detrimental interaction. This study confirms that myostatin
knockdown by exon skipping is a potential therapeutic strategy to counteract muscle
wasting conditions and dual myostatin and dystrophin skipping has potential as a therapy
for DMD.
Collapse
Affiliation(s)
- Alberto Malerba
- 1] School of Biological Sciences, Royal Holloway, University of London, Surrey, UK [2] Department of Veterinary basic sciences, Royal Veterinary College, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Funkenstein B, Krol E, Esterin E, Kim YS. Structural and functional characterizations of activin type 2B receptor (acvr2b) ortholog from the marine fish, gilthead sea bream, Sparus aurata: evidence for gene duplication of acvr2b in fish. J Mol Endocrinol 2012; 49:175-92. [PMID: 22911153 DOI: 10.1530/jme-12-0075] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myostatin (MSTN), a negative regulator of muscle growth and a member of the transforming growth factor-β superfamily, can bind the two activin type 2 receptors (ACVR2). It has been previously shown that WT mice injected with ACVR2B extracellular domain (ACVR2B-ECD) had higher muscle mass. Likewise, fish larvae immersed in Pichia pastoris culture supernatant, containing goldfish Acvr2b-ECD, showed enhanced larval growth. However, it is not clear whether fish Mstn1 and Mstn2 signal through the same receptor and whether fish express more than one acvr2b gene. In the current study, three cDNAs encoding acvr2b (saacvr2b-1, saacvr2b-2a, and saacvr2b-2b) were cloned from gilthead sea bream. All three contain the short extracellular binding domain, a short transmembrane region, and a conserved catalytic domain of serine/threonine protein kinase. Bioinformatics analysis provided evidence for the existence of two acvr2b genes (acvr2b-1 and acvr2b-2) in several other fish species as well, probably as a result of gene or genome duplication. The two isoforms differ in their amino acid sequences. The direct inhibitory effect of Acvr2b-ECD on Mstn activity was tested in vitro. The saAcvr2b-1-ECD was expressed in the yeast P. pastoris. Evidence is provided for N-glycosylation of Acvr2b-1-ECD. The affinity-purified Acvr2b-1-ECD inhibited recombinant mouse/rat/human mature MSTN activity when determined in vitro using the CAGA-luciferase assay in A204 cells. A lower inhibitory activity was obtained when unprocessed purified, furin-digested, and activated saMstn1 was used. Results of this study demonstrate for the first time the existence of two acvr2b genes in fish. In addition, the study shows that bioactive fish Acvr2b-ECD can be produced from P. pastoris.
Collapse
Affiliation(s)
- Bruria Funkenstein
- Department of Marine Biology and Biotechnology, National Institute of Oceanography, Israel Oceanographic and Limnological Research, Tel-Shikmona, Haifa 31080, Israel.
| | | | | | | |
Collapse
|
47
|
Nadjar-Boger E, Maccatrozzo L, Radaelli G, Funkenstein B. Genomic cloning and promoter functional analysis of myostatin-2 in shi drum, Umbrina cirrosa: conservation of muscle-specific promoter activity. Comp Biochem Physiol B Biochem Mol Biol 2012. [PMID: 23178682 DOI: 10.1016/j.cbpb.2012.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Myostatin (MSTN) is a member of the transforming growth factor-ß superfamily, known as a negative regulator of skeletal muscle development and growth in mammals. In contrast to mammals, fish possess at least two paralogs of MSTN: MSTN-1 and MSTN-2. Here we describe the cloning and sequence analysis of spliced and precursor (unspliced) transcripts as well as the 5' flanking region of MSTN-2 from the marine fish Umbrina cirrosa (ucMSTN-2). In silico analysis revealed numerous putative cis regulatory elements including several E-boxes known as binding sites to myogenic transcription factors. Transient transfection experiments using non-muscle and muscle cell lines showed high transcriptional activity in muscle cells and in differentiated neural cells, in accordance with our previous findings in MSTN-2 promoter from Sparus aurata. Comparative informatics analysis of MSTN-2 from several fish species revealed high conservation of the predicted amino acid sequence as well as the gene structure (exon length) although intron length varied between species. The proximal promoter of MSTN-2 gene was found to be conserved among Perciforms. In conclusion, this study reinforces our conclusion that MSTN-2 promoter is a very strong promoter, especially in muscle cells. In addition, we show that the MSTN-2 gene structure is highly conserved among fishes as is the predicted amino acid sequence of the peptide.
Collapse
Affiliation(s)
- Elisabeth Nadjar-Boger
- Department of Marine Biology & Biotechnology, National Institute of Oceanography, Israel Oceanographic and Limnological Research, Tel-Shikmona, P.O.B 8030, Haifa 31080, Israel
| | | | | | | |
Collapse
|
48
|
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
|
49
|
Iwasaki S, Miyake M, Watanabe H, Kitagawa E, Watanabe K, Ohwada S, Kitazawa H, Rose MT, Aso H. Expression of myostatin in neural cells of the olfactory system. Mol Neurobiol 2012; 47:1-8. [PMID: 22941030 DOI: 10.1007/s12035-012-8342-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 08/22/2012] [Indexed: 10/27/2022]
Abstract
Recent studies show that myostatin mRNA expression is found in some regions of the brain. However, the functional significance of this is currently unknown. We therefore investigated myostatin expression and function in the brain. In this study, we used immunohistochemistry, in situ hybridization, and RT-PCR analysis to reveal that myostatin is expressed in the mitral cells in the olfactory bulb (OB) and in neurons in the olfactory cortex (OC). Using 3D reconstruction, mitral cells positive for myostatin were positioned in the lateral and ventral regions of the OB. In contrast, myostatin-positive mitral cells were detected in mice at 2 weeks of age, but not on days 0 and 7 after birth. Activin receptor IIB, a myostatin receptor, was expressed in the OB, OC, hippocampus, and paraventricular thalamic nucleus. Moreover, c-Fos immunostaining in granule cells in the OB was augmented after intracerebroventricular injection of myostatin. These findings suggest that myostatin is localized in specific cells associated with the olfactory system of the brain and may act as a key inhibitor in cell and/or signal development of the olfactory system.
Collapse
Affiliation(s)
- Shunsuke Iwasaki
- Laboratory of Functional Morphology, Department of Animal Biology, Graduate School of Agricultural Science, Tohoku University, Sendai, 981-8555, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Myostatin (Mstn) is a secreted growth factor that negatively regulates muscle mass and is therefore a potential pharmacological target for the treatment of muscle wasting disorders such as Duchenne muscular dystrophy. Here we describe a novel Mstn blockade approach in which small interfering RNAs (siRNAs) complementary to a promoter-associated transcript induce transcriptional gene silencing (TGS) in two differentiated mouse muscle cell lines. Silencing is sensitive to treatment with the histone deacetylase inhibitor trichostatin A, and the silent state chromatin mark H3K9me2 is enriched at the Mstn promoter following siRNA transfection, suggesting epigenetic remodeling underlies the silencing effect. These observations suggest that long-term epigenetic silencing may be feasible for Mstn and that TGS is a promising novel therapeutic strategy for the treatment of muscle wasting disorders.
Collapse
|