151
|
Dukes A, Davis C, El Refaey M, Upadhyay S, Mork S, Arounleut P, Johnson MH, Hill WD, Isales CM, Hamrick MW. The aromatic amino acid tryptophan stimulates skeletal muscle IGF1/p70s6k/mTor signaling in vivo and the expression of myogenic genes in vitro. Nutrition 2015; 31:1018-24. [PMID: 26059377 DOI: 10.1016/j.nut.2015.02.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 01/22/2015] [Accepted: 02/11/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Nutrition plays a key role in the maintenance of muscle and bone mass, and dietary protein deficiency has in particular been associated with catabolism of both muscle and bone tissue. One mechanism thought to link protein deficiency with loss of muscle mass is deficiency in specific amino acids that play a role in muscle metabolism. The aim of this study was to test the hypothesis that the essential amino acid tryptophan, and its metabolite kynurenine, might directly affect muscle metabolism in the setting of protein deficiency. METHODS Adult mice (12 mo) were fed a normal diet (18% protein), as well as diets with low protein (8%) supplemented with increasing concentrations (50, 100, and 200 uM) of kynurenine (Kyn) or with tryptophan (Trp; 1.5 mM) for 8 weeks. Myoprogenitor cells were also treated with Trp and Kyn in vitro to determine their effects on cell proliferation and expression of myogenic differentiation markers. RESULTS All mice on the low-protein diets weighed less than the group fed normal protein (18%). Lean mass measured by dual-energy X-ray absorptiometry was lowest in mice on the high Kyn diet, whereas percent lean mass was highest in mice receiving Trp supplementation and percent body fat was lowest in mice receiving Trp. Enzyme-linked immunosorbent assays showed significant increases in skeletal muscle insulin-like growth factor-1, leptin, and the myostatin antagonist follistatin with Trp supplementation. mRNA microarray and gene pathway analysis performed on muscle samples demonstrate that mTor/eif4/p70s6k pathway molecules are significantly up-regulated in muscles from mice on Kyn and Trp supplementation. In vitro, neither amino acid affected proliferation of myoprogenitors, but Trp increased the expression of the myogenic markers MyoD, myogenin, and myosin heavy chain. CONCLUSION These findings suggest that dietary amino acids can directly affect molecular signaling in skeletal muscle, further indicating that dietary manipulation with specific amino acids could potentially attenuate muscle loss with dietary protein deficiency.
Collapse
Affiliation(s)
- Amy Dukes
- Georgia Regents University (Formerly Georgia Health Sciences University), Augusta, GA, USA
| | - Colleen Davis
- Georgia Regents University (Formerly Georgia Health Sciences University), Augusta, GA, USA
| | - Mona El Refaey
- Georgia Regents University (Formerly Georgia Health Sciences University), Augusta, GA, USA
| | - Sunil Upadhyay
- Georgia Regents University (Formerly Georgia Health Sciences University), Augusta, GA, USA
| | - Sarah Mork
- Georgia Regents University (Formerly Georgia Health Sciences University), Augusta, GA, USA
| | - Phonepasong Arounleut
- Georgia Regents University (Formerly Georgia Health Sciences University), Augusta, GA, USA
| | - Maribeth H Johnson
- Georgia Regents University (Formerly Georgia Health Sciences University), Augusta, GA, USA
| | - William D Hill
- Georgia Regents University (Formerly Georgia Health Sciences University), Augusta, GA, USA
| | - Carlos M Isales
- Georgia Regents University (Formerly Georgia Health Sciences University), Augusta, GA, USA
| | - Mark W Hamrick
- Georgia Regents University (Formerly Georgia Health Sciences University), Augusta, GA, USA.
| |
Collapse
|
152
|
Wang Q, Guo T, Portas J, McPherron AC. A soluble activin receptor type IIB does not improve blood glucose in streptozotocin-treated mice. Int J Biol Sci 2015; 11:199-208. [PMID: 25561902 PMCID: PMC4279095 DOI: 10.7150/ijbs.10430] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 11/05/2014] [Indexed: 12/18/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM), or insulin dependent DM, is accompanied by decreased muscle mass. The growth factor myostatin (MSTN) is a negative regulator of muscle growth, and a loss of MSTN signaling has been shown to increase muscle mass and prevent the development of obesity, insulin resistance and lipodystrophic diabetes in mice. The effects of MSTN inhibition in a T1DM model on muscle mass and blood glucose are unknown. We asked whether MSTN inhibition would increase muscle mass and decrease hyperglycemia in mice treated with streptozotocin (STZ) to destroy pancreatic beta cells. After diabetes developed, mice were treated with a soluble MSTN/activin receptor fused to Fc (ACVR2B:Fc). ACVR2B:Fc increased body weight and muscle mass compared to vehicle treated mice. Unexpectedly, ACVR2B:Fc reproducibly exacerbated hyperglycemia within approximately one week of administration. ACVR2B:Fc treatment also elevated serum levels of the glucocorticoid corticosterone. These results suggest that although MSTN/activin inhibitors increased muscle mass, they may be counterproductive in improving health in patients with T1DM.
Collapse
Affiliation(s)
- Qian Wang
- 1. Current Addresses: Pathology Department, Stony Brook University Medical Center, Stony Brook, New York, USA
| | - Tingqing Guo
- 2. Novo Nordisk Research Centre China, Changping District, Beijing, China
| | - Jennifer Portas
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
153
|
de Mello F, Streit DP, Sabin N, Gabillard JC. Dynamic expression of tgf-β2, tgf-β3 and inhibin βA during muscle growth resumption and satellite cell differentiation in rainbow trout (Oncorhynchus mykiss). Gen Comp Endocrinol 2015; 210:23-9. [PMID: 25449661 DOI: 10.1016/j.ygcen.2014.10.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/15/2014] [Accepted: 10/17/2014] [Indexed: 02/07/2023]
Abstract
Members of the TGF-β superfamily are involved in numerous cell functions; however, except for myostatin, their roles in the regulation of muscle growth in fish are completely unknown. We measured tgf-β1, tgf-β2, tgf-β3, inhibin βA (inh) and follistatin (fst) gene expression during muscle growth recovery following a fasting period. We observed that tgf-β1a and tgf-β2 expression were quickly down-regulated after refeeding and that tgf-β3 reached its highest level of expression 7days post-refeeding, mirroring myogenin expression. Inh βA1 mRNA levels decreased sharply after refeeding, in contrast to fst b2 expression, which peaked at day 2. No significant modification of expression was observed for tgf-β1a, tgf-β1b, tgf-β1c and tgf-β6 during refeeding. In vitro, tgf-β2 and inh βA1 expression decreased during the differentiation of satellite cells, whereas tgf-β3 expression increased following the same pattern as myogenin. Surprisingly, fst b1 and fst b2 expression decreased during differentiation, whereas no variation was observed in fst a1 and fst a2 expression levels. In vitro analyses also indicated that IGF1 treatment up-regulated tgf-β3, inh βA1 and myogenin expression, and that MSTN treatment increased fst b1 and fst b2 expression. In conclusion, we showed that the expression of tgf-β2, tgf-β3 and inh βA1 is dynamically regulated during muscle growth resumption and satellite cell differentiation, strongly suggesting that these genes have a role in the regulation of muscle growth.
Collapse
Affiliation(s)
- Fernanda de Mello
- Federal University of Rio Grande do Sul, Faculty of Agricultural Sciences, Research Group AQUAM, Aquaculture Sector, Department of Animal Science, Avenue Bento Gonçalves 7712, Agronomia, Porto Alegre 91540-000, RS, Brazil
| | - Danilo Pedro Streit
- Federal University of Rio Grande do Sul, Faculty of Agricultural Sciences, Research Group AQUAM, Aquaculture Sector, Department of Animal Science, Avenue Bento Gonçalves 7712, Agronomia, Porto Alegre 91540-000, RS, Brazil
| | - Nathalie Sabin
- INRA, UR1037 Laboratory of Fish Physiology and Genomic, Growth and Flesh Quality Group, campus de Beaulieu, 35000 Rennes, France
| | - Jean-Charles Gabillard
- INRA, UR1037 Laboratory of Fish Physiology and Genomic, Growth and Flesh Quality Group, campus de Beaulieu, 35000 Rennes, France.
| |
Collapse
|
154
|
Palstra AP, Rovira M, Rizo-Roca D, Torrella JR, Spaink HP, Planas JV. Swimming-induced exercise promotes hypertrophy and vascularization of fast skeletal muscle fibres and activation of myogenic and angiogenic transcriptional programs in adult zebrafish. BMC Genomics 2014; 15:1136. [PMID: 25518849 PMCID: PMC4378002 DOI: 10.1186/1471-2164-15-1136] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 12/11/2014] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND The adult skeletal muscle is a plastic tissue with a remarkable ability to adapt to different levels of activity by altering its excitability, its contractile and metabolic phenotype and its mass. We previously reported on the potential of adult zebrafish as a tractable experimental model for exercise physiology, established its optimal swimming speed and showed that swimming-induced contractile activity potentiated somatic growth. Given that the underlying exercise-induced transcriptional mechanisms regulating muscle mass in vertebrates are not fully understood, here we investigated the cellular and molecular adaptive mechanisms taking place in fast skeletal muscle of adult zebrafish in response to swimming. RESULTS Fish were trained at low swimming speed (0.1 m/s; non-exercised) or at their optimal swimming speed (0.4 m/s; exercised). A significant increase in fibre cross-sectional area (1.290±88 vs. 1.665±106 μm2) and vascularization (298±23 vs. 458±38 capillaries/mm2) was found in exercised over non-exercised fish. Gene expression profiling by microarray analysis evidenced the activation of a series of complex transcriptional networks of extracellular and intracellular signaling molecules and pathways involved in the regulation of muscle mass (e.g. IGF-1/PI3K/mTOR, BMP, MSTN), myogenesis and satellite cell activation (e.g. PAX3, FGF, Notch, Wnt, MEF2, Hh, EphrinB2) and angiogenesis (e.g. VEGF, HIF, Notch, EphrinB2, KLF2), some of which had not been previously associated with exercise-induced contractile activity. CONCLUSIONS The results from the present study show that exercise-induced contractile activity in adult zebrafish promotes a coordinated adaptive response in fast muscle that leads to increased muscle mass by hypertrophy and increased vascularization by angiogenesis. We propose that these phenotypic adaptations are the result of extensive transcriptional changes induced by exercise. Analysis of the transcriptional networks that are activated in response to exercise in the adult zebrafish fast muscle resulted in the identification of key signaling pathways and factors for the regulation of skeletal muscle mass, myogenesis and angiogenesis that have been remarkably conserved during evolution from fish to mammals. These results further support the validity of the adult zebrafish as an exercise model to decipher the complex molecular and cellular mechanisms governing skeletal muscle mass and function in vertebrates.
Collapse
Affiliation(s)
| | | | | | | | | | - Josep V Planas
- Departament de Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
155
|
O'Connell KE, Guo W, Serra C, Beck M, Wachtman L, Hoggatt A, Xia D, Pearson C, Knight H, O'Connell M, Miller AD, Westmoreland SV, Bhasin S. The effects of an ActRIIb receptor Fc fusion protein ligand trap in juvenile simian immunodeficiency virus-infected rhesus macaques. FASEB J 2014; 29:1165-75. [PMID: 25466897 DOI: 10.1096/fj.14-257543] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/05/2014] [Indexed: 01/16/2023]
Abstract
There are no approved therapies for muscle wasting in children infected with human immunodeficiency virus (HIV), which portends poor disease outcomes. To determine whether a soluble ActRIIb receptor Fc fusion protein (ActRIIB.Fc), a ligand trap for TGF-β/activin family members including myostatin, can prevent or restore loss of lean body mass and body weight in simian immunodeficiency virus (SIV)-infected juvenile rhesus macaques (Macaca mulatta). Fourteen pair-housed, juvenile male rhesus macaques were inoculated with SIVmac239 and, 4 wk postinoculation (WPI) treated with intramuscular injections of 10 mg ⋅ kg(-1) ⋅ wk(-1) ActRIIB.Fc or saline placebo. Body weight, lean body mass, SIV titers, and somatometric measurements were assessed monthly for 16 wk. Age-matched SIV-infected rhesus macaques were injected with saline. Intervention groups did not differ at baseline. Gains in lean mass were significantly greater in the ActRIIB.Fc group than in the placebo group (P < 0.001). Administration of ActRIIB.Fc was associated with greater gains in body weight (P = 0.01) and upper arm circumference than placebo. Serum CD4(+) T-lymphocyte counts and SIV copy numbers did not differ between groups. Administration of ActRIIB.Fc was associated with higher muscle expression of myostatin than placebo. ActRIIB.Fc effectively blocked and reversed loss of body weight, lean mass, and fat mass in juvenile SIV-infected rhesus macaques.
Collapse
Affiliation(s)
- Karyn E O'Connell
- *Department of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts, USA; and Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Wen Guo
- *Department of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts, USA; and Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carlo Serra
- *Department of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts, USA; and Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew Beck
- *Department of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts, USA; and Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lynn Wachtman
- *Department of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts, USA; and Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Amber Hoggatt
- *Department of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts, USA; and Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dongling Xia
- *Department of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts, USA; and Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Chris Pearson
- *Department of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts, USA; and Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Heather Knight
- *Department of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts, USA; and Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Micheal O'Connell
- *Department of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts, USA; and Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew D Miller
- *Department of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts, USA; and Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Susan V Westmoreland
- *Department of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts, USA; and Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shalender Bhasin
- *Department of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts, USA; and Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
156
|
Formicola L, Marazzi G, Sassoon DA. The extraocular muscle stem cell niche is resistant to ageing and disease. Front Aging Neurosci 2014; 6:328. [PMID: 25520657 PMCID: PMC4249457 DOI: 10.3389/fnagi.2014.00328] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 11/10/2014] [Indexed: 12/13/2022] Open
Abstract
Specific muscles are spared in many degenerative myopathies. Most notably, the extraocular muscles (EOMs) do not show clinical signs of late stage myopathies including the accumulation of fibrosis and fat. It has been proposed that an altered stem cell niche underlies the resistance of EOMs in these pathologies, however, to date, no reports have provided a detailed characterization of the EOM stem cell niche. PW1/Peg3 is expressed in progenitor cells in all adult tissues including satellite cells and a subset of interstitial non-satellite cell progenitors in muscle. These PW1-positive interstitial cells (PICs) include a fibroadipogenic progenitor population (FAP) that give rise to fat and fibrosis in late stage myopathies. PICs/FAPs are mobilized following injury and FAPs exert a promyogenic role upon myoblasts in vitro but require the presence of a minimal population of satellite cells in vivo. We and others recently described that FAPs express promyogenic factors while satellite cells express antimyogenic factors suggesting that PICs/FAPs act as support niche cells in skeletal muscle through paracrine interactions. We analyzed the EOM stem cell niche in young adult and aged wild-type mice and found that the balance between PICs and satellite cells within the EOM stem cell niche is maintained throughout life. Moreover, in the adult mdx mouse model for Duchenne muscular dystrophy (DMD), the EOM stem cell niche is unperturbed compared to normal mice, in contrast to Tibialis Anterior (TA) muscle, which displays signs of ongoing degeneration/regeneration. Regenerating mdx TA shows increased levels of both PICs and satellite cells, comparable to normal unaffected EOMs. We propose that the increase in PICs that we observe in normal EOMs contributes to preserving the integrity of the myofibers and satellite cells. Our data suggest that molecular cues regulating muscle regeneration are intrinsic properties of EOMs.
Collapse
Affiliation(s)
- Luigi Formicola
- UMRS 1166 INSERM, Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), University of Pierre and Marie Curie Paris VI Paris, France
| | - Giovanna Marazzi
- UMRS 1166 INSERM, Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), University of Pierre and Marie Curie Paris VI Paris, France
| | - David A Sassoon
- UMRS 1166 INSERM, Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), University of Pierre and Marie Curie Paris VI Paris, France
| |
Collapse
|
157
|
Development of novel activin-targeted therapeutics. Mol Ther 2014; 23:434-44. [PMID: 25399825 DOI: 10.1038/mt.2014.221] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 11/09/2014] [Indexed: 01/07/2023] Open
Abstract
Soluble activin type II receptors (ActRIIA/ActRIIB), via binding to diverse TGF-β proteins, can increase muscle and bone mass, correct anemia or protect against diet-induced obesity. While exciting, these multiple actions of soluble ActRIIA/IIB limit their therapeutic potential and highlight the need for new reagents that target specific ActRIIA/IIB ligands. Here, we modified the activin A and activin B prodomains, regions required for mature growth factor synthesis, to generate specific activin antagonists. Initially, the prodomains were fused to the Fc region of mouse IgG2A antibody and, subsequently, "fastener" residues (Lys(45), Tyr(96), His(97), and Ala(98); activin A numbering) that confer latency to other TGF-β proteins were incorporated. For the activin A prodomain, these modifications generated a reagent that potently (IC(50) 5 nmol/l) and specifically inhibited activin A signaling in vitro, and activin A-induced muscle wasting in vivo. Interestingly, the modified activin B prodomain inhibited both activin A and B signaling in vitro (IC(50) ~2 nmol/l) and in vivo, suggesting it could serve as a general activin antagonist. Importantly, unlike soluble ActRIIA/IIB, the modified prodomains did not inhibit myostatin or GDF-11 activity. To underscore the therapeutic utility of specifically antagonising activin signaling, we demonstrate that the modified activin prodomains promote significant increases in muscle mass.
Collapse
|
158
|
de Mello F, Streit DP, Sabin N, Gabillard JC. Identification of TGF-β, inhibin βA and follistatin paralogs in the rainbow trout genome. Comp Biochem Physiol B Biochem Mol Biol 2014; 177-178:46-55. [DOI: 10.1016/j.cbpb.2014.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 07/22/2014] [Accepted: 07/25/2014] [Indexed: 11/15/2022]
|
159
|
Singh R, Braga M, Pervin S. Regulation of brown adipocyte metabolism by myostatin/follistatin signaling. Front Cell Dev Biol 2014; 2:60. [PMID: 25364764 PMCID: PMC4207030 DOI: 10.3389/fcell.2014.00060] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 09/26/2014] [Indexed: 12/11/2022] Open
Abstract
Obesity develops from perturbations of cellular bioenergetics, when energy uptake exceeds energy expenditure, and represents a major risk factor for the development of type 2 diabetes, dyslipidemia, cardiovascular disease, cancer, and other conditions. Brown adipose tissue (BAT) has long been known to dissipate energy as heat and contribute to energy expenditure, but its presence and physiological role in adult human physiology has been questioned for years. Recent demonstrations of metabolically active brown fat depots in adult humans have revolutionized current therapeutic approaches for obesity-related diseases. The balance between white adipose tissue (WAT) and BAT affects the systemic energy balance and is widely believed to be the key determinant in the development of obesity and related metabolic diseases. Members of the transforming growth factor-beta (TGF-β) superfamily play an important role in regulating overall energy homeostasis by modulation of brown adipocyte characteristics. Inactivation of TGF-β/Smad3/myostatin (Mst) signaling promotes browning of white adipocytes, increases mitochondrial biogenesis and protects mice from diet-induced obesity, suggesting the need for development of a novel class of TGF-β/Mst antagonists for the treatment of obesity and related metabolic diseases. We recently described an important role of follistatin (Fst), a soluble glycoprotein that is known to bind and antagonize Mst actions, during brown fat differentiation and the regulation of cellular metabolism. Here we highlight various investigations performed using different in vitro and in vivo models to support the contention that targeting TGF-β/Mst signaling enhances brown adipocyte functions and regulates energy balance, reducing insulin resistance, and curbing the development of obesity and diabetes.
Collapse
Affiliation(s)
- Rajan Singh
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science Los Angeles, CA, USA ; Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA Los Angeles, CA, USA
| | - Melissa Braga
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science Los Angeles, CA, USA ; Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA Los Angeles, CA, USA
| | - Shehla Pervin
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science Los Angeles, CA, USA ; Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA Los Angeles, CA, USA
| |
Collapse
|
160
|
Sartori R, Gregorevic P, Sandri M. TGFβ and BMP signaling in skeletal muscle: potential significance for muscle-related disease. Trends Endocrinol Metab 2014; 25:464-71. [PMID: 25042839 DOI: 10.1016/j.tem.2014.06.002] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 06/09/2014] [Accepted: 06/10/2014] [Indexed: 01/07/2023]
Abstract
The transforming growth factor beta (TGFβ) superfamily comprises a large number of secreted proteins that regulate various fundamental biological processes underlying embryonic development and the postnatal regulation of many cell types and organs. Sequence similarities define two ligand subfamilies: the TGFβ/activin subfamily and the bone morphogenetic protein (BMP) subfamily. The discovery that myostatin, a member of the TGFβ/activin subfamily, negatively controls muscle mass attracted attention to this pathway. However, recent findings of a positive role for BMP-mediated signaling in muscle have challenged the model of how the TGFβ network regulates skeletal muscle phenotype. This review illustrates how this complex network integrates crosstalk among members of the TGFβ superfamily and downstream signaling elements to regulate muscle in health and disease.
Collapse
Affiliation(s)
- Roberta Sartori
- Dulbecco Telethon Institute, Venetian Institute of Molecular Medicine, 35129 Padova, Italy; Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy
| | - Paul Gregorevic
- Division of Cell Signaling and Metabolism, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia
| | - Marco Sandri
- Dulbecco Telethon Institute, Venetian Institute of Molecular Medicine, 35129 Padova, Italy; Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy; Telethon Institute of Genetics and Medicine (TIGEM), 80131 Napoli, Italy.
| |
Collapse
|
161
|
Li X, Wang J, Liu H, Wang H, Sun L, Yang C, Li L, Zheng Y. Construction of a eukaryotic expression vector for pEGFP-FST and its biological activity in duck myoblasts. ELECTRON J BIOTECHN 2014. [DOI: 10.1016/j.ejbt.2014.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
162
|
Wei C, Li L, Su H, Xu L, Lu J, Zhang L, Liu W, Ren H, Du L. Identification of the crucial molecular events during the large-scale myoblast fusion in sheep. Physiol Genomics 2014; 46:429-40. [DOI: 10.1152/physiolgenomics.00184.2013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
It is well known that in sheep most myofibers are formed before birth; however, the crucial myogenic stage and the cellular and molecular mechanisms underpinning phenotypic variation of fetal muscle development remain to be ascertained. We used histological, microarray, and quantitative real-time PCR (qPCR) methods to examine the developmental characteristics of fetal muscle at 70, 85, 100, 120, and 135 days of gestation in sheep. We show that day 100 is an important checkpoint for change in muscle transcriptome and histomorphology in fetal sheep and that the period of 85–100 days is the vital developmental stage for large-scale myoblast fusion. Furthermore, we identified the cis-regulatory motifs for E2F1 or MEF2A in a list of decreasingly or increasingly expressed genes between 85 and 100 days, respectively. Further analysis demonstrated that the mRNA and phosphorylated protein levels of E2F1 and MEF2A significantly declined with myogenic progression in vivo and in vitro. qRT-PCR analysis indicated that PI3K and FST, as targets of E2F1, may be involved in myoblast differentiation and fusion and that downregulation of MEF2A contributes to transition of myofiber types by differential regulation of the target genes involved at the stage of 85–100 days. We clarify for the first time the timing of myofiber proliferation and development during gestation in sheep, which would be beneficial to meat sheep production. Our findings present a repertoire of gene expression in muscle during large-scale myoblast fusion at transcriptome-wide level, which contributes to elucidate the regulatory network of myogenic differentiation.
Collapse
Affiliation(s)
- Caihong Wei
- National Center for Molecular Genetics and Breeding of Animal, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Li Li
- National Center for Molecular Genetics and Breeding of Animal, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, Sichuan, China; and
| | - Hongwei Su
- College of Animal Science and Technology, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Lingyang Xu
- National Center for Molecular Genetics and Breeding of Animal, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jian Lu
- National Center for Molecular Genetics and Breeding of Animal, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Li Zhang
- National Center for Molecular Genetics and Breeding of Animal, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Wenzhong Liu
- College of Animal Science and Technology, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Hangxing Ren
- National Center for Molecular Genetics and Breeding of Animal, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, China
| | - Lixin Du
- National Center for Molecular Genetics and Breeding of Animal, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
163
|
Dschietzig TB. Myostatin — From the Mighty Mouse to cardiovascular disease and cachexia. Clin Chim Acta 2014; 433:216-24. [DOI: 10.1016/j.cca.2014.03.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 03/19/2014] [Accepted: 03/19/2014] [Indexed: 02/02/2023]
|
164
|
Waning DL, Guise TA. Molecular mechanisms of bone metastasis and associated muscle weakness. Clin Cancer Res 2014; 20:3071-7. [PMID: 24677373 DOI: 10.1158/1078-0432.ccr-13-1590] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Bone is a preferred site for breast cancer metastasis and leads to pathologic bone loss due to increased osteoclast-induced bone resorption. The homing of tumor cells to the bone depends on the support of the bone microenvironment in which the tumor cells prime the premetastatic niche. The colonization and growth of tumor cells then depend on adaptations in the invading tumor cells to take advantage of normal physiologic responses by mimicking bone marrow cells. This concerted effort by tumor cells leads to uncoupled bone remodeling in which the balance of osteoclast-driven bone resorption and osteoblast-driven bone deposition is lost. Breast cancer bone metastases often lead to osteolytic lesions due to hyperactive bone resorption. Release of growth factors from bone matrix during resorption then feeds a "vicious cycle" of bone destruction leading to many skeletal-related events. In addition to activity in bone, some of the factors released during bone resorption are also known to be involved in skeletal muscle regeneration and contraction. In this review, we discuss the mechanisms that lead to osteolytic breast cancer bone metastases and the potential for cancer-induced bone-muscle cross-talk leading to skeletal muscle weakness.
Collapse
Affiliation(s)
- David L Waning
- Authors' Affiliation: Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Theresa A Guise
- Authors' Affiliation: Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis, Indiana
| |
Collapse
|
165
|
Uezumi A, Ikemoto-Uezumi M, Tsuchida K. Roles of nonmyogenic mesenchymal progenitors in pathogenesis and regeneration of skeletal muscle. Front Physiol 2014; 5:68. [PMID: 24605102 PMCID: PMC3932482 DOI: 10.3389/fphys.2014.00068] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 02/04/2014] [Indexed: 12/25/2022] Open
Abstract
Adult skeletal muscle possesses a remarkable regenerative ability that is dependent on satellite cells. However, skeletal muscle is replaced by fatty and fibrous connective tissue in several pathological conditions. Fatty and fibrous connective tissue becomes a major cause of muscle weakness and leads to further impairment of muscle function. Because the occurrence of fatty and fibrous connective tissue is usually associated with severe destruction of muscle, the idea that dysregulation of the fate switch in satellite cells may underlie this pathological change has emerged. However, recent studies identified nonmyogenic mesenchymal progenitors in skeletal muscle and revealed that fatty and fibrous connective tissue originates from these progenitors. Later, these progenitors were also demonstrated to be the major contributor to heterotopic ossification in skeletal muscle. Because nonmyogenic mesenchymal progenitors represent a distinct cell population from satellite cells, targeting these progenitors could be an ideal therapeutic strategy that specifically prevents pathological changes of skeletal muscle, while preserving satellite cell-dependent regeneration. In addition to their roles in pathogenesis of skeletal muscle, nonmyogenic mesenchymal progenitors may play a vital role in muscle regeneration by regulating satellite cell behavior. Conversely, muscle cells appear to regulate behavior of nonmyogenic mesenchymal progenitors. Thus, these cells regulate each other reciprocally and a proper balance between them is a key determinant of muscle integrity. Furthermore, nonmyogenic mesenchymal progenitors have been shown to maintain muscle mass in a steady homeostatic condition. Understanding the nature of nonmyogenic mesenchymal progenitors will provide valuable insight into the pathophysiology of skeletal muscle. In this review, we focus on nonmyogenic mesenchymal progenitors and discuss their roles in muscle pathogenesis, regeneration, and homeostasis.
Collapse
Affiliation(s)
- Akiyoshi Uezumi
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University Aichi, Japan
| | - Madoka Ikemoto-Uezumi
- Department of Regenerative Medicine, National Center for Geriatrics and Gerontology, National Institute for Longevity Sciences Aichi, Japan
| | - Kunihiro Tsuchida
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University Aichi, Japan
| |
Collapse
|
166
|
Liu H, Li X, Sun L, Wang H, Zhang R, Yang C, Li L, Wang J, He H, Krumm C. Effects of the regulation of follistatin mRNA expression by IGF-1 in duck (Anas platyrhynchos) skeletal muscle. Growth Horm IGF Res 2014; 24:35-41. [PMID: 24429073 DOI: 10.1016/j.ghir.2013.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 11/01/2013] [Accepted: 12/19/2013] [Indexed: 10/25/2022]
Abstract
The IGF-1 and TGF-β pathways have been shown to be involved in regulating muscle development. Many mediators that are associated with the regulation of muscle development have been found to participate in the cross-talk between these two pathways. To research the relationships between IGF-1 and the follistatin-mediated TGF-β pathways in duck skeletal muscle development, a series of studies were conducted. The results showed that follistatin had similar expression patterns to IGF-1 during duck embryonic muscle development. The in ovo feeding of IGF-1 to duck eggs was shown to increase follistatin expression in the duck skeletal muscle. Thus, IGF-1 may induce the mRNA expression of follistatin. These results suggest that follistatin may be a key regulator of multiple signaling cascades responding to the cross-talk between the IGF-1 and TGF-β pathways.
Collapse
Affiliation(s)
- Hehe Liu
- Institute of Animal Breeding and Genetics, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Xinxin Li
- Institute of Animal Breeding and Genetics, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Lingli Sun
- Institute of Animal Breeding and Genetics, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Haohan Wang
- Institute of Animal Breeding and Genetics, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Rongping Zhang
- Institute of Animal Breeding and Genetics, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Chao Yang
- Institute of Animal Breeding and Genetics, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Liang Li
- Institute of Animal Breeding and Genetics, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Jiwen Wang
- Institute of Animal Breeding and Genetics, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China.
| | - Hua He
- Institute of Animal Breeding and Genetics, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Christopher Krumm
- Institute of Animal Breeding and Genetics, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| |
Collapse
|
167
|
Braga M, Reddy ST, Vergnes L, Pervin S, Grijalva V, Stout D, David J, Li X, Tomasian V, Reid CB, Norris KC, Devaskar SU, Reue K, Singh R. Follistatin promotes adipocyte differentiation, browning, and energy metabolism. J Lipid Res 2014; 55:375-84. [PMID: 24443561 PMCID: PMC3934723 DOI: 10.1194/jlr.m039719] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Follistatin (Fst) functions to bind and neutralize the activity of members of the transforming growth factor-β superfamily. Fst has a well-established role in skeletal muscle, but we detected significant Fst expression levels in interscapular brown and subcutaneous white adipose tissue, and further investigated its role in adipocyte biology. Fst expression was induced during adipogenic differentiation of mouse brown preadipocytes and mouse embryonic fibroblasts (MEFs) as well as in cold-induced brown adipose tissue from mice. In differentiated MEFs from Fst KO mice, the induction of brown adipocyte proteins including uncoupling protein 1, PR domain containing 16, and PPAR gamma coactivator-1α was attenuated, but could be rescued by treatment with recombinant FST. Furthermore, Fst enhanced thermogenic gene expression in differentiated mouse brown adipocytes and MEF cultures from both WT and Fst KO groups, suggesting that Fst produced by adipocytes may act in a paracrine manner. Our microarray gene expression profiling of WT and Fst KO MEFs during adipogenic differentiation identified several genes implicated in lipid and energy metabolism that were significantly downregulated in Fst KO MEFs. Furthermore, Fst treatment significantly increases cellular respiration in Fst-deficient cells. Our results implicate a novel role of Fst in the induction of brown adipocyte character and regulation of energy metabolism.
Collapse
Affiliation(s)
- Melissa Braga
- Division of Endocrinology and Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059; and
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Regulation of the follistatin gene by RSPO-LGR4 signaling via activation of the WNT/β-catenin pathway in skeletal myogenesis. Mol Cell Biol 2013; 34:752-64. [PMID: 24344199 DOI: 10.1128/mcb.01285-13] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
WNT signaling plays multiple roles in skeletal myogenesis during gestation and postnatal stages. The R-spondin (RSPO) family of secreted proteins and their cognate receptors, members of leucine-rich repeat-containing G protein-coupled receptor (LGR) family, have emerged as new regulatory components of the WNT signaling pathway. We previously showed that RSPO2 promoted myogenic differentiation via activation of WNT/β-catenin signaling in mouse myoblast C2C12 cells in vitro. However, the molecular mechanism by which RSPO2 regulates myogenic differentiation is unknown. Herein, we show that depletion of the LGR4 receptor severely disrupts myogenic differentiation and significantly diminishes the response to RSPO2 in C2C12 cells, showing a requirement of LGR4 in RSPO signaling during myogenic differentiation. We identify the transforming growth factor β (TGF-β) antagonist follistatin (Fst) as a key mediator of RSPO-LGR4 signaling in myogenic differentiation. We further demonstrate that Fst is a direct target of the WNT/β-catenin pathway. Activation and inactivation of β-catenin induced and inhibited Fst expression, respectively, in both C2C12 cells and mouse embryos. Specific TCF/LEF1 binding sites within the promoter and intron 1 region of the Fst gene were required for RSPO2 and WNT/β-catenin-induced Fst expression. This study uncovers a molecular cross talk between WNT/β-catenin and TGF-β signaling pivotal in myogenic differentiation.
Collapse
|
169
|
An antibody blocking activin type II receptors induces strong skeletal muscle hypertrophy and protects from atrophy. Mol Cell Biol 2013; 34:606-18. [PMID: 24298022 DOI: 10.1128/mcb.01307-13] [Citation(s) in RCA: 227] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The myostatin/activin type II receptor (ActRII) pathway has been identified to be critical in regulating skeletal muscle size. Several other ligands, including GDF11 and the activins, signal through this pathway, suggesting that the ActRII receptors are major regulatory nodes in the regulation of muscle mass. We have developed a novel, human anti-ActRII antibody (bimagrumab, or BYM338) to prevent binding of ligands to the receptors and thus inhibit downstream signaling. BYM338 enhances differentiation of primary human skeletal myoblasts and counteracts the inhibition of differentiation induced by myostatin or activin A. BYM338 prevents myostatin- or activin A-induced atrophy through inhibition of Smad2/3 phosphorylation, thus sparing the myosin heavy chain from degradation. BYM338 dramatically increases skeletal muscle mass in mice, beyond sole inhibition of myostatin, detected by comparing the antibody with a myostatin inhibitor. A mouse version of the antibody induces enhanced muscle hypertrophy in myostatin mutant mice, further confirming a beneficial effect on muscle growth beyond myostatin inhibition alone through blockade of ActRII ligands. BYM338 protects muscles from glucocorticoid-induced atrophy and weakness via prevention of muscle and tetanic force losses. These data highlight the compelling therapeutic potential of BYM338 for the treatment of skeletal muscle atrophy and weakness in multiple settings.
Collapse
|
170
|
Abstract
In an intriguing new study, Loffredo et al report that joining the circulation of old mice with that of young mice reduces age-related cardiac hypertrophy. They also found that the growth factor growth/differentiation factor 11 is a circulating negative regulator of cardiac hypertrophy which suggests that raising growth/differentiation factor 11 levels may be useful to treat cardiac hypertrophy associated with aging.
Collapse
Affiliation(s)
- Alexandra C McPherron
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
171
|
Vamvini MT, Aronis KN, Panagiotou G, Huh JY, Chamberland JP, Brinkoetter MT, Petrou M, Christophi CA, Kales SN, Christiani DC, Mantzoros CS. Irisin mRNA and circulating levels in relation to other myokines in healthy and morbidly obese humans. Eur J Endocrinol 2013; 169:829-34. [PMID: 24062354 PMCID: PMC3857961 DOI: 10.1530/eje-13-0276] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Skeletal muscle is considered to be an endocrine organ that secretes a number of myokines including follistatin (FST), myostatin (MSTN), activin A, and the newly identified irisin. Irisin's biology and function exhibit similarities with the functions of the FST-MSTN-activin A axis. It remains unknown whether there is any interplay among these molecules. The aim of this study is to examine potential associations of irisin with the FST, MSTN, and activin A axis. METHODS Two observational studies were performed to evaluate the associations of irisin with the other three peptides. Study A included 150 healthy males aged 18.48±0.16 years with BMI 23.18±3.75 kg/m(2). Fasting serum samples were used to measure the levels of the molecules of interest. Study B included 14 morbidly obese individuals, candidates for bariatric surgery, aged 53.14±8.93 years with BMI 50.18±10.63 kg/m(2). Blood samples were obtained after an overnight fast. Eight out of the 14 participants consented to an optional thigh biopsy during their bariatric surgery. Using the above blood and tissue samples, we measured circulating levels and muscle mRNA of irisin, FST, MSTN, and activin A. RESULTS We report that FNDC5 mRNA in muscle is positively correlated with FST mRNA expression in morbidly obese subjects (ρ=0.93, P<0.001). We also found that circulating irisin is positively correlated with FST circulating levels among lean subjects (ρ=0.17, P=0.05) while this association was suggestive among the obese (ρ=0.56, P=0.07). CONCLUSION The newly identified myokine irisin may be positively associated with FST at both the mRNA and circulating protein level.
Collapse
Affiliation(s)
- Maria T. Vamvini
- Division of Endocrinology, Diabetes & Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Konstantinos N. Aronis
- Division of Endocrinology, Diabetes & Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Department of Internal Medicine, Boston Medical Center, Boston University, Boston, MA
- Section of Endocrinology, Boston VA Healthcare System, Boston, MA
| | - Grigorios Panagiotou
- Division of Endocrinology, Diabetes & Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Joo Young Huh
- Division of Endocrinology, Diabetes & Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - John P. Chamberland
- Division of Endocrinology, Diabetes & Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Mary T. Brinkoetter
- Division of Endocrinology, Diabetes & Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | | | - Costas A. Christophi
- Cyprus International Institute for Environmental and Public Health in association with Harvard School of Public Health, Cyprus University of Technology, Limassol, Cyprus
| | - Stefanos N. Kales
- Department of Environmental Health, Harvard School of Public Health, Boston, MA, USA
| | - David C. Christiani
- Department of Environmental Health, Harvard School of Public Health, Boston, MA, USA
| | - Christos S. Mantzoros
- Division of Endocrinology, Diabetes & Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Section of Endocrinology, Boston VA Healthcare System, Boston, MA
| |
Collapse
|
172
|
Abstract
Myostatin, a member of the transforming growth factor beta (TGF-β) superfamily, was first described in 1997. Since then, myostatin has gained growing attention because of the discovery that myostatin inhibition leads to muscle mass accrual. Myostatin not only plays a key role in muscle homeostasis, but also affects fat and bone. This review will focus on the impact of myostatin and its inhibition on muscle mass/function, adipose tissue and bone density/geometry in humans. Although existing data are sparse, myostatin inhibition leads to increased lean mass and 1 study found a decrease in fat mass and increase in bone formation. In addition, myostatin levels are increased in sarcopenia, cachexia and bed rest whereas they are increased after resistance training, suggesting physiological regulatory of myostatin. Increased myostatin levels have also been found in obesity and levels decrease after weight loss from caloric restriction. Knowledge on the relationship of myostatin with bone is largely based on animal data where elevated myostatin levels lead to decreased BMD and myostatin inhibition improved BMD. In summary, myostatin appears to be a key factor in the integrated physiology of muscle, fat, and bone. It is unclear whether myostatin directly affects fat and bone, or indirectly via muscle. Whether via direct or indirect effects, myostatin inhibition appears to increase muscle and bone mass and decrease fat tissue-a combination that truly appears to be a holy grail. However, at this time, human data for both efficacy and safety are extremely limited. Moreover, whether increased muscle mass also leads to improved function remains to be determined. Ultimately potential beneficial effects of myostatin inhibition will need to be determined based on hard outcomes such as falls and fractures.
Collapse
Affiliation(s)
- B Buehring
- Division of Geriatrics and Gerontology, University of Wisconsin Osteoporosis Clinical Research Program, UW Madison, 2870 University Ave, Suite 100, Madison, WI, 53705, USA,
| | | |
Collapse
|
173
|
Winbanks CE, Chen JL, Qian H, Liu Y, Bernardo BC, Beyer C, Watt KI, Thomson RE, Connor T, Turner BJ, McMullen JR, Larsson L, McGee SL, Harrison CA, Gregorevic P. The bone morphogenetic protein axis is a positive regulator of skeletal muscle mass. ACTA ACUST UNITED AC 2013; 203:345-57. [PMID: 24145169 PMCID: PMC3812980 DOI: 10.1083/jcb.201211134] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The BMP signaling pathway promotes muscle growth and inhibits muscle wasting via SMAD1/5-dependent signaling. Although the canonical transforming growth factor β signaling pathway represses skeletal muscle growth and promotes muscle wasting, a role in muscle for the parallel bone morphogenetic protein (BMP) signaling pathway has not been defined. We report, for the first time, that the BMP pathway is a positive regulator of muscle mass. Increasing the expression of BMP7 or the activity of BMP receptors in muscles induced hypertrophy that was dependent on Smad1/5-mediated activation of mTOR signaling. In agreement, we observed that BMP signaling is augmented in models of muscle growth. Importantly, stimulation of BMP signaling is essential for conservation of muscle mass after disruption of the neuromuscular junction. Inhibiting the phosphorylation of Smad1/5 exacerbated denervation-induced muscle atrophy via an HDAC4-myogenin–dependent process, whereas increased BMP–Smad1/5 activity protected muscles from denervation-induced wasting. Our studies highlight a novel role for the BMP signaling pathway in promoting muscle growth and inhibiting muscle wasting, which may have significant implications for the development of therapeutics for neuromuscular disorders.
Collapse
Affiliation(s)
- Catherine E Winbanks
- Division of Cell Signaling and Metabolism, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Abstract
Myostatin (MSTN) and growth and differentiation factor-11 (GDF-11) are highly related TGF-β family members that have distinct biological functions. MSTN is expressed primarily in skeletal muscle and acts to limit muscle growth. GDF-11 is expressed more widely and plays multiple roles, including regulating axial skeletal patterning during development. Several MSTN and GDF-11 binding proteins have been identified, including GDF-associated serum protein-1 (GASP-1) and GASP-2, which are capable of inhibiting the activities of these ligands. Here, we show that GASP-1 and GASP-2 act by blocking the initial signaling event (namely, the binding of the ligand to the type II receptor). Moreover, we show that mice lacking Gasp1 and Gasp2 have phenotypes consistent with overactivity of MSTN and GDF-11. Specifically, we show that Gasp2(-/-) mice have posteriorly directed transformations of the axial skeleton, which contrast with the anteriorly directed transformations seen in Gdf11(-/-) mice. We also show that both Gasp1(-/-) and Gasp2(-/-) mice have reductions in muscle weights, a shift in fiber type from fast glycolytic type IIb fibers to fast oxidative type IIa fibers, and impaired muscle regeneration ability, which are the reverse of what are seen in Mstn(-/-) mice. All of these findings suggest that both GASP-1 and GASP-2 are important modulators of GDF-11 and MSTN activity in vivo.
Collapse
|
175
|
Goodman CA, McNally RM, Hoffmann FM, Hornberger TA. Smad3 induces atrogin-1, inhibits mTOR and protein synthesis, and promotes muscle atrophy in vivo. Mol Endocrinol 2013; 27:1946-57. [PMID: 24002653 DOI: 10.1210/me.2013-1194] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Myostatin, a member of the TGF superfamily, is sufficient to induce skeletal muscle atrophy. Myostatin-induced atrophy is associated with increases in E3-ligase atrogin-1 expression and protein degradation and decreases in Akt/mechanistic target of rapamycin (mTOR) signaling and protein synthesis. Myostatin signaling activates the transcription factor Smad3 (Small Mothers Against Decapentaplegic), which has been shown to be necessary for myostatin-induced atrogin-1 expression and atrophy; however, it is not known whether Smad3 is sufficient to induce these events or whether Smad3 simply plays a permissive role. Thus, the aim of this study was to address these questions with an in vivo model. To accomplish this goal, in vivo transfection of plasmid DNA was used to create transient transgenic mouse skeletal muscles, and our results show for the first time that Smad3 expression is sufficient to stimulate atrogin-1 promoter activity, inhibit Akt/mTOR signaling and protein synthesis, and induce muscle fiber atrophy. Moreover, we propose that Akt/mTOR signaling is inhibited by a Smad3-induced decrease in microRNA-29 (miR-29) expression and a subsequent increase in the translation of phosphatase and tensin homolog (PTEN) mRNA. Smad3 is also sufficient to inhibit peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α) promoter activity and to increase FoxO (Forkhead Box Protein, Subclass O)-mediated signaling and the promoter activity of plasminogen activator inhibitor 1 (PAI-1). Combined, this study provides the first evidence that Smad3 is sufficient to regulate many of the events associated with myostatin-induced atrophy and therefore suggests that Smad3 signaling may be a viable target for therapies aimed at preventing myostatin-induced muscle atrophy.
Collapse
Affiliation(s)
- Craig A Goodman
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706.
| | | | | | | |
Collapse
|
176
|
Liu H, Zhang R, Li X, Sun L, Wang H, Yang C, Li L, Wang J, Xu F. Influence of recombinant duck follistatin protein on embryonic muscle development and gene expressions. J Anim Physiol Anim Nutr (Berl) 2013; 98:522-9. [PMID: 23957442 DOI: 10.1111/jpn.12104] [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] [Received: 01/15/2013] [Accepted: 06/13/2013] [Indexed: 01/30/2023]
Abstract
Follistatin (FST) acts as a positive regulator of muscle development by inhibiting the activities and expression of myostatin. The recombinant duck FST protein was injected into hatching eggs and was also added to the medium of duck myoblast to study its role on duck embryonic muscle development and gene expressions. Duck embryo weight increased 3.49% (p > 0.05) in FST treatment group as compared with control group, but minor effects were found on leg or breast muscle weights of ducklings at 2 days post-hatching (p > 0.05). Relative expression of Pax7 was upregulated in both leg and breast muscle tissues (p < 0.05), while MyoD was only upregulated in leg muscle (p < 0.05), and Myf5 was only upregulated in breast muscle (p < 0.05). Relative expression of myostatin was downregulated in both muscle tissues researched (p < 0.05). In vitro studies also showed some maker genes relevant to protein synthesis and degradation, cells' proliferation and differentiation had significant changes in myoblasts after treated with FST. These results suggested that in ovo feeding of recombinant FST protein to duck hatching eggs had an effect on duck embryo development but have less roles on the duck embryonic muscle development.
Collapse
Affiliation(s)
- H Liu
- Institute of Animal Breeding & Genetic, Sichuan Agricultural University, Ya'an, Sichuan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
177
|
Damatto R, Martinez P, Lima A, Cezar M, Campos D, Oliveira Junior S, Guizoni D, Bonomo C, Nakatani B, Dal Pai Silva M, Carvalho R, Okoshi K, Okoshi M. Heart failure-induced skeletal myopathy in spontaneously hypertensive rats. Int J Cardiol 2013; 167:698-703. [DOI: 10.1016/j.ijcard.2012.03.063] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 03/02/2012] [Accepted: 03/03/2012] [Indexed: 12/31/2022]
|
178
|
Terada S, Ota S, Kobayashi M, Kobayashi T, Mifune Y, Takayama K, Witt M, Vadalà G, Oyster N, Otsuka T, Fu FH, Huard J. Use of an antifibrotic agent improves the effect of platelet-rich plasma on muscle healing after injury. J Bone Joint Surg Am 2013; 95:980-8. [PMID: 23780535 DOI: 10.2106/jbjs.l.00266] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Muscle contusions are a common type of muscle injury and are frequently encountered in athletes and military personnel. Although these injuries are capable of healing in most instances, incomplete functional recovery often occurs because of the development of fibrosis in the muscle. We hypothesized that a combination of platelet-rich plasma (PRP) injection and oral administration of losartan (an antifibrotic agent) could enhance muscle healing by stimulating muscle regeneration and angiogenesis and by preventing fibrosis in contusion-injured skeletal muscle. METHODS Contusion injuries were created in the tibialis anterior muscles of mice. Two treatments were tested, alone and in combination: 20 μL of PRP injected into the contusion site one day after injury, and 10 mg/kg/day of losartan administered beginning three days after injury and continuing until the end point of the experiment. Muscle regeneration and fibrosis development were evaluated by histological analysis, and functional recovery was measured by physiological testing. RESULTS Muscle regeneration and muscle function were significantly promoted in the combined PRP + losartan treatment group compared with the other groups. Combined PRP + losartan treatment significantly decreased the expression of phosphorylated Smad2/3 and the development of fibrosis compared with PRP treatment alone, and it increased vascular endothelial growth factor (VEGF) expression and the number of CD31-positive structures compared with losartan treatment alone. Follistatin, a positive regulator of muscle growth, was expressed at a higher level in the PRP + losartan group compared with the other groups. CONCLUSIONS PRP + losartan combinatorial therapy improved overall skeletal muscle healing after muscle contusion injury by enhancing angiogenesis and follistatin expression and by reducing the expression of phosphorylated Smad2/3 and the development of fibrosis. These results suggest that blocking the expression of transforming growth factor (TGF)-β1 with losartan improves the effect of PRP therapy on muscle healing after a contusion injury. CLINICAL RELEVANCE These findings could contribute to the development of biological treatments that aid in the healing of skeletal muscle after injury.
Collapse
Affiliation(s)
- Satoshi Terada
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Bridgeside Point II, Suite 206, 450 Technology Drive, Pittsburgh, PA 15219, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Roberts EW, Deonarine A, Jones JO, Denton AE, Feig C, Lyons SK, Espeli M, Kraman M, McKenna B, Wells RJ, Zhao Q, Caballero OL, Larder R, Coll AP, O’Rahilly S, Brindle KM, Teichmann SA, Tuveson DA, Fearon DT. Depletion of stromal cells expressing fibroblast activation protein-α from skeletal muscle and bone marrow results in cachexia and anemia. J Exp Med 2013; 210:1137-51. [PMID: 23712428 PMCID: PMC3674708 DOI: 10.1084/jem.20122344] [Citation(s) in RCA: 334] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 04/29/2013] [Indexed: 12/15/2022] Open
Abstract
Fibroblast activation protein-α (FAP) identifies stromal cells of mesenchymal origin in human cancers and chronic inflammatory lesions. In mouse models of cancer, they have been shown to be immune suppressive, but studies of their occurrence and function in normal tissues have been limited. With a transgenic mouse line permitting the bioluminescent imaging of FAP(+) cells, we find that they reside in most tissues of the adult mouse. FAP(+) cells from three sites, skeletal muscle, adipose tissue, and pancreas, have highly similar transcriptomes, suggesting a shared lineage. FAP(+) cells of skeletal muscle are the major local source of follistatin, and in bone marrow they express Cxcl12 and KitL. Experimental ablation of these cells causes loss of muscle mass and a reduction of B-lymphopoiesis and erythropoiesis, revealing their essential functions in maintaining normal muscle mass and hematopoiesis, respectively. Remarkably, these cells are altered at these sites in transplantable and spontaneous mouse models of cancer-induced cachexia and anemia. Thus, the FAP(+) stromal cell may have roles in two adverse consequences of cancer: their acquisition by tumors may cause failure of immunosurveillance, and their alteration in normal tissues contributes to the paraneoplastic syndromes of cachexia and anemia.
Collapse
Affiliation(s)
- Edward W. Roberts
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Andrew Deonarine
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - James O. Jones
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Alice E. Denton
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Christine Feig
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Scott K. Lyons
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Marion Espeli
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Matthew Kraman
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Brendan McKenna
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Richard J.B. Wells
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Qi Zhao
- Ludwig Collaborative Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Otavia L. Caballero
- Ludwig Collaborative Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Rachel Larder
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Anthony P. Coll
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Stephen O’Rahilly
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Kevin M. Brindle
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Sarah A. Teichmann
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - David A. Tuveson
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Douglas T. Fearon
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| |
Collapse
|
180
|
Zhong SS, Jiang XY, Sun CF, Zou SM. Identification of a second follistatin gene in grass carp (Ctenopharyngodon idellus) and its regulatory function in myogenesis during embryogenesis. Gen Comp Endocrinol 2013; 185:19-27. [PMID: 23396016 DOI: 10.1016/j.ygcen.2013.01.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 01/15/2013] [Accepted: 01/20/2013] [Indexed: 11/27/2022]
Abstract
Follistatin can antagonize the function of myostatin as a competitive binding protein and promote muscle growth in vivo. Here, we report the isolation and characterization of a second follistatin gene fst2 in grass carp (Ctenopharyngodon idellus). The grass carp fst2 cDNA was 1,376 bp in length, with an open reading frame (ORF) encoding 350 amino acid residues. A relatively low sequence identity of 78% was found between grass carp Fst2 and its paralog Fst1. Sequence and phylogenetic analyses suggest that the grass carp fst2 originated from fish-specific gene duplication. In adult fish, fst2 mRNA expression was observed in most tissues but was strongly expressed in the eyes, muscles, skin and ovary. Grass carp fst2 mRNA could be detected as early as 16 h post-fertilization (hpf), while fst1 mRNA was detected throughout embryogenesis. Using in situ hybridization, fst2 transcripts were detected in the anterior somites at 24 hpf and in the brain and posterior somites at 36 hpf. Meanwhile, fst1 mRNA was transcribed mainly in the optic vesicle and at the cephalic mesoderm at 12 hpf, in the eyes, cephalic mesoderm and at the lateral edge of most somites at 24 hpf, and mainly in the brain at 36 hpf. Furthermore, overexpression of fst2 mRNA markedly affected the formation of the embryonic midline and somite structures. Based on comparisons with fst1, our findings suggest that fst2 retained the ancestral functions of regulating muscle development and growth during embryogenesis in grass carp.
Collapse
Affiliation(s)
- Sha-Sha Zhong
- Key Laboratory of Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Huchenghuan Road 999, Shanghai 201306, China
| | | | | | | |
Collapse
|
181
|
Abstract
PURPOSE OF REVIEW There are a variety of pathophysiologic conditions that are known to induce skeletal muscle atrophy. However, muscle wasting can occur through multiple distinct signaling pathways with differential sensitivity between selective skeletal muscle fiber subtypes. This review summarizes some of the underlying molecular mechanisms responsible for fiber-specific muscle mass regulation. RECENT FINDINGS Peroxisome proliferator-activated receptor gamma coactivator 1-alpha protects slow-twitch oxidative fibers from denervation/immobilization (disuse)-induced muscle atrophies. Nutrient-related muscle atrophies, such as those induced by cancer cachexia, sepsis, chronic heart failure, or diabetes, are largely restricted to fast-twitch glycolytic fibers, of which the underlying mechanism is usually related to abnormality of protein degradation, including proteasomal and lysosomal pathways. In contrast, nuclear factor kappaB activation apparently serves a dual function by inducing both fast-twitch fiber atrophy and slow-twitch fiber degeneration. SUMMARY Fast-twitch glycolytic fibers are more vulnerable than slow-twitch oxidative fibers under a variety of atrophic conditions related to signaling transduction of Forkhead box O family, autophagy inhibition, transforming growth factor beta family, and nuclear factor-kappaB. The resistance of oxidative fibers may result from the protection of peroxisome proliferator-activated receptor gamma coactivator 1-alpha.
Collapse
Affiliation(s)
- Yichen Wang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Diabetes Research and Training Center, Bronx, New York, USA
| | - Jeffrey E. Pessin
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Diabetes Research and Training Center, Bronx, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, Diabetes Research and Training Center, Bronx, New York, USA
| |
Collapse
|
182
|
Merrick BA, Phadke DP, Auerbach SS, Mav D, Stiegelmeyer SM, Shah RR, Tice RR. RNA-Seq profiling reveals novel hepatic gene expression pattern in aflatoxin B1 treated rats. PLoS One 2013; 8:e61768. [PMID: 23630614 PMCID: PMC3632591 DOI: 10.1371/journal.pone.0061768] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/13/2013] [Indexed: 01/16/2023] Open
Abstract
Deep sequencing was used to investigate the subchronic effects of 1 ppm aflatoxin B1 (AFB1), a potent hepatocarcinogen, on the male rat liver transcriptome prior to onset of histopathological lesions or tumors. We hypothesized RNA-Seq would reveal more differentially expressed genes (DEG) than microarray analysis, including low copy and novel transcripts related to AFB1’s carcinogenic activity compared to feed controls (CTRL). Paired-end reads were mapped to the rat genome (Rn4) with TopHat and further analyzed by DESeq and Cufflinks-Cuffdiff pipelines to identify differentially expressed transcripts, new exons and unannotated transcripts. PCA and cluster analysis of DEGs showed clear separation between AFB1 and CTRL treatments and concordance among group replicates. qPCR of eight high and medium DEGs and three low DEGs showed good comparability among RNA-Seq and microarray transcripts. DESeq analysis identified 1,026 differentially expressed transcripts at greater than two-fold change (p<0.005) compared to 626 transcripts by microarray due to base pair resolution of transcripts by RNA-Seq, probe placement within transcripts or an absence of probes to detect novel transcripts, splice variants and exons. Pathway analysis among DEGs revealed signaling of Ahr, Nrf2, GSH, xenobiotic, cell cycle, extracellular matrix, and cell differentiation networks consistent with pathways leading to AFB1 carcinogenesis, including almost 200 upregulated transcripts controlled by E2f1-related pathways related to kinetochore structure, mitotic spindle assembly and tissue remodeling. We report 49 novel, differentially-expressed transcripts including confirmation by PCR-cloning of two unique, unannotated, hepatic AFB1-responsive transcripts (HAfT’s) on chromosomes 1.q55 and 15.q11, overexpressed by 10 to 25-fold. Several potentially novel exons were found and exon refinements were made including AFB1 exon-specific induction of homologous family members, Ugt1a6 and Ugt1a7c. We find the rat transcriptome contains many previously unidentified, AFB1-responsive exons and transcripts supporting RNA-Seq’s capabilities to provide new insights into AFB1-mediated gene expression leading to hepatocellular carcinoma.
Collapse
MESH Headings
- Aflatoxin B1/toxicity
- Animals
- Carcinogens/toxicity
- Carcinoma, Hepatocellular/chemically induced
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Cell Transformation, Neoplastic/chemically induced
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- E2F1 Transcription Factor/physiology
- Exons
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Regulatory Networks
- Glucuronosyltransferase/genetics
- Glucuronosyltransferase/metabolism
- High-Throughput Nucleotide Sequencing
- Liver/drug effects
- Liver/metabolism
- Liver Neoplasms, Experimental/chemically induced
- Liver Neoplasms, Experimental/genetics
- Liver Neoplasms, Experimental/metabolism
- Male
- Oligonucleotide Array Sequence Analysis
- Precancerous Conditions/chemically induced
- Precancerous Conditions/metabolism
- Principal Component Analysis
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Sequence Analysis, RNA
- Transcriptome
Collapse
Affiliation(s)
- B Alex Merrick
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America.
| | | | | | | | | | | | | |
Collapse
|
183
|
Mangner N, Matsuo Y, Schuler G, Adams V. Cachexia in chronic heart failure: endocrine determinants and treatment perspectives. Endocrine 2013; 43:253-65. [PMID: 22903414 DOI: 10.1007/s12020-012-9767-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 07/24/2012] [Indexed: 12/11/2022]
Abstract
It is well documented in the current literature that chronic heart failure is often associated with cachexia, defined as involuntary weight loss of 5 % in 12 month or less. Clinical studies unraveled that the presence of cachexia decreases significantly mean survival of the patient. At the molecular level mainly myofibrillar proteins are degraded, although a reduced protein synthesis may also contribute to the loss of muscle mass. Endocrine factors clearly regulate muscle mass and function by influencing the normally precisely controlled balance between protein breakdown and protein synthesis The aim of the present article is to review the knowledge in the field with respect to the role of endocrine factors for the regulation of cachexia in patients with CHF and deduce treatment perspectives.
Collapse
Affiliation(s)
- Norman Mangner
- Heart Center Leipzig, University Leipzig, Strümpellstrasse 39, 04289, Leipzig, Germany
| | | | | | | |
Collapse
|
184
|
Sun L, Lu K, Liu H, Wang H, Li X, Yang C, Li L, Wang J. The effects of endoplasmic reticulum stress response on duck decorin stimulate myotube hypertrophy in myoblasts. Mol Cell Biochem 2013; 377:151-61. [DOI: 10.1007/s11010-013-1581-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 01/30/2013] [Indexed: 12/24/2022]
|
185
|
Abstract
Sarcopenia is one of the leading causes of disability in the elderly. Despite the growing prevalence of sarcopenia, the molecular mechanisms that control aging-related changes in muscle mass are not fully understood. The ubiquitin proteasome system is one of the major pathways that regulate muscle protein degradation, and this system plays a central role in controlling muscle size. Atrogin-1 and MuRF-1 are two E3 ubiquitin ligases that are important regulators of ubiquitin-mediated protein degradation in skeletal muscle. In this review, we will discuss: (i) aging-related changes to skeletal muscle structure and function; (ii) the regulation of protein synthesis and protein degradation by IGF-1, TGF-β, and myostatin, with emphasis on the control of atrogin-1 and MuRF-1 expression; and (iii) the potential for modulating atrogin-1 and MuRF-1 expression to treat or prevent sarcopenia.
Collapse
Affiliation(s)
- Jonathan P Gumucio
- Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 2017, Ann Arbor, MI, 48109-2200, USA
| | | |
Collapse
|
186
|
Affiliation(s)
- Masayuki Shimano
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, 715 Albany Street, Boston, MA 02118, USA
| | | | | |
Collapse
|
187
|
Kärst S, Strucken EM, Schmitt AO, Weyrich A, de Villena FPM, Yang H, Brockmann GA. Effect of the myostatin locus on muscle mass and intramuscular fat content in a cross between mouse lines selected for hypermuscularity. BMC Genomics 2013; 14:16. [PMID: 23324137 PMCID: PMC3626839 DOI: 10.1186/1471-2164-14-16] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 12/19/2012] [Indexed: 12/07/2022] Open
Abstract
Background This study is aimed at the analysis of genetic and physiological effects of myostatin on economically relevant meat quality traits in a genetic background of high muscularity. For this purpose, we generated G3 populations of reciprocal crosses between the two hypermuscular mouse lines BMMI866, which carries a myostatin mutation and is lean, and BMMI806, which has high intramuscular and body fat content. To assess the relationship between muscle mass, body composition and muscle quality traits, we also analysed intramuscular fat content (IMF), water holding capacity (WHC), and additional physiological parameters in M. quadriceps and M. longissimus in 308 G3-animals. Results We found that individuals with larger muscles have significantly lower total body fat (r = −0.28) and IMF (r = −0.64), and in females, a lower WHC (r = −0.35). In males, higher muscle mass was also significantly correlated with higher glycogen contents (r = 0.2) and lower carcass pH-values 24 hours after dissection (r = −0.19). Linkage analyses confirmed the influence of the myostatin mutation on higher lean mass (1.35 g), reduced body fat content (−1.15%), and lower IMF in M. longissimus (−0.13%) and M. quadriceps (−0.07%). No effect was found for WHC. A large proportion of variation of intramuscular fat content of the M. longissimus at the myostatin locus could be explained by sex (23%) and direction-of-cross effects (26%). The effects were higher in males (+0.41%). An additional locus with negative over-dominance effects on total fat mass (−0.55 g) was identified on chromosome 16 at 94 Mb (86–94 Mb) which concurs with fat related QTL in syntenic regions on SSC13 in pigs and BTA1 in cattle. Conclusion The data shows QTL effects on mouse muscle that are similar to those previously observed in livestock, supporting the mouse model. New information from the mouse model helps to describe variation in meat quantity and quality, and thus contribute to research in livestock.
Collapse
Affiliation(s)
- Stefan Kärst
- Department for Crop and Animal Sciences, Breeding Biology and Molecular Genetics, Humboldt-Universität zu Berlin, Invalidenstrasse 42, 10115, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
188
|
Palstra AP, Beltran S, Burgerhout E, Brittijn SA, Magnoni LJ, Henkel CV, Jansen HJ, van den Thillart GEEJM, Spaink HP, Planas JV. Deep RNA sequencing of the skeletal muscle transcriptome in swimming fish. PLoS One 2013; 8:e53171. [PMID: 23308156 PMCID: PMC3540090 DOI: 10.1371/journal.pone.0053171] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 11/26/2012] [Indexed: 11/20/2022] Open
Abstract
Deep RNA sequencing (RNA-seq) was performed to provide an in-depth view of the transcriptome of red and white skeletal muscle of exercised and non-exercised rainbow trout (Oncorhynchus mykiss) with the specific objective to identify expressed genes and quantify the transcriptomic effects of swimming-induced exercise. Pubertal autumn-spawning seawater-raised female rainbow trout were rested (n = 10) or swum (n = 10) for 1176 km at 0.75 body-lengths per second in a 6,000-L swim-flume under reproductive conditions for 40 days. Red and white muscle RNA of exercised and non-exercised fish (4 lanes) was sequenced and resulted in 15–17 million reads per lane that, after de novo assembly, yielded 149,159 red and 118,572 white muscle contigs. Most contigs were annotated using an iterative homology search strategy against salmonid ESTs, the zebrafish Danio rerio genome and general Metazoan genes. When selecting for large contigs (>500 nucleotides), a number of novel rainbow trout gene sequences were identified in this study: 1,085 and 1,228 novel gene sequences for red and white muscle, respectively, which included a number of important molecules for skeletal muscle function. Transcriptomic analysis revealed that sustained swimming increased transcriptional activity in skeletal muscle and specifically an up-regulation of genes involved in muscle growth and developmental processes in white muscle. The unique collection of transcripts will contribute to our understanding of red and white muscle physiology, specifically during the long-term reproductive migration of salmonids.
Collapse
Affiliation(s)
- Arjan P Palstra
- Departament de Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
189
|
Phelps MP, Jaffe IM, Bradley TM. Muscle growth in teleost fish is regulated by factors utilizing the activin II B receptor. J Exp Biol 2013; 216:3742-50. [DOI: 10.1242/jeb.086660] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Summary
The activin type IIB receptor (Acvr2b) is the cell surface receptor for multiple transforming growth factor β (TGF-β) superfamily ligands, several of which regulate muscle growth in mammals. To investigate the role of the Acvr2b signaling pathway in the growth and development of skeletal muscle in teleost fish, transgenic rainbow trout (RBT; Oncorhynchus mykiss, Walbaum) expressing a truncated form of the acvr2b-2a (acvr2bΔ) in muscle tissue were produced. High levels of acvr2bΔ expression were detected in the majority of P1 transgenic fish. Transgenic P1 trout developed enhanced, localized musculature in both the epaxial and hypaxial regions (dubbed "six pack"). The F1 transgenic offspring did not exhibit localized muscle growth, but rather developed a uniform body morphology with greater girth, condition factor, and increased muscle fiber hypertrophy. There was a high degree of variation in the weight of both P1 and F1 transgenic fish with several fish of each generation exhibiting enhanced growth compared to other transgenic and control siblings. The "six pack" phenotype observed in P1 transgenic RBT overexpressing a acvr2bΔ and the presence of F1 individuals with altered muscle morphology provides compelling evidence for the importance of TGF-β signaling molecules in regulating muscle growth in teleost fish.
Collapse
|
190
|
Laser capture microdissection of metachromatically stained skeletal muscle allows quantification of fiber type specific gene expression. Mol Cell Biochem 2012. [PMID: 23196635 DOI: 10.1007/s11010-012-1538-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Skeletal muscle contains various myofiber types closely associated with satellite stem cells, vasculature, and neurons, thus making it difficult to perform genetic or proteomic expression analysis with sufficient cellular specificity to resolve differences at the individual cell or myofiber type level. Here, we describe the combination of a simple histochemical method capable of simultaneously identifying Type I, IIA, IIB, and IIC myofibers followed by laser capture micro-dissection (LCM) to compare the expression profiles of individual fiber types, myonuclear domains, and satellite cells in frozen muscle sections of control and atrophied muscle. Quantitative RT-PCR (qPCR) was used to verify the integrity of the cell-specific RNAs harvested after histologic staining, while qPCR for specific genes of interest was used to quantify atrophy-associated changes in mRNA. Our data demonstrate that the differential myofiber atrophy previously described by histologic means is related to differential expression of atrophy-related genes, such as MuRF1 and MAFbx (a.k.a. Atrogin-1), within different myofiber type populations. This spatially resolved molecular pathology (SRMP) technique allowed quantitation of atrophy-related gene products within individual fiber types that could not be resolved by expression analysis of the whole muscle. The present study demonstrates the importance of fiber type specific expression profiling in understanding skeletal muscle biology especially during muscle atrophy and provides a practical method of performing such research.
Collapse
|
191
|
Bowser M, Herberg S, Arounleut P, Shi X, Fulzele S, Hill WD, Isales CM, Hamrick MW. Effects of the activin A-myostatin-follistatin system on aging bone and muscle progenitor cells. Exp Gerontol 2012. [PMID: 23178301 DOI: 10.1016/j.exger.2012.11.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The activin A-myostatin-follistatin system is thought to play an important role in the regulation of muscle and bone mass throughout growth, development, and aging; however, the effects of these ligands on progenitor cell proliferation and differentiation in muscle and bone are not well understood. In addition, age-associated changes in the relative expression of these factors in musculoskeletal tissues have not been described. We therefore examined changes in protein levels of activin A, follistatin, and myostatin (GDF-8) in both muscle and bone with age in C57BL6 mice using ELISA. We then investigated the effects of activin A, myostatin and follistatin on the proliferation and differentiation of primary myoblasts and mouse bone marrow stromal cells (BMSCs) in vitro. Myostatin levels and the myostatin:follistatin ratio increased with age in the primarily slow-twitch mouse soleus muscle, whereas the pattern was reversed with age in the fast-twitch extensor digitorum longus muscle. Myostatin levels and the myostatin:follistatin ratio increased significantly (+75%) in mouse bone marrow with age, as did activin A levels (+17%). Follistatin increased the proliferation of primary myoblasts from both young and aged mice, whereas myostatin increased proliferation of younger myoblasts but decreased proliferation of older myoblasts. Myostatin reduced proliferation of both young and aged BMSCs in a dose-dependent fashion, and activin A increased mineralization in both young and aged BMSCs. Together these data suggest that aging in mice is accompanied by changes in the expression of activin A and myostatin, as well as changes in the response of bone and muscle progenitor cells to these factors. Myostatin appears to play a particularly important role in the impaired proliferative capacity of muscle and bone progenitor cells from aged mice.
Collapse
Affiliation(s)
- Matthew Bowser
- Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | |
Collapse
|
192
|
Mid-gestational gene expression profile in placenta and link to pregnancy complications. PLoS One 2012; 7:e49248. [PMID: 23145134 PMCID: PMC3492272 DOI: 10.1371/journal.pone.0049248] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Accepted: 10/04/2012] [Indexed: 12/25/2022] Open
Abstract
Despite the importance of placenta in mediating rapid physiological changes in pregnancy, data on temporal dynamics of placental gene expression are limited. We completed the first transcriptome profiling of human placental gene expression dynamics (GeneChips, Affymetrix®; ∼47,000 transcripts) from early to mid-gestation (n = 10; gestational weeks 5–18) and report 154 genes with significant transcriptional changes (ANOVA, FDR P<0.1). TaqMan RT-qPCR analysis (n = 43; gestational weeks 5–41) confirmed a significant (ANOVA and t-test, FDR P<0.05) mid-gestational peak of placental gene expression for BMP5, CCNG2, CDH11, FST, GATM, GPR183, ITGBL1, PLAGL1, SLC16A10 and STC1, followed by sharp decrease in mRNA levels at term (t-test, FDR P<0.05). We hypothesized that normal course of late pregnancy may be affected when genes characteristic to mid-gestation placenta remain highly expressed until term, and analyzed their expression in term placentas from normal and complicated pregnancies [preeclampsia (PE), n = 12; gestational diabetes mellitus (GDM), n = 12; small- and large-for-gestational-age newborns (SGA, LGA), n = 12+12]. STC1 (stanniocalcin 1) exhibited increased mRNA levels in all studied complications, with the most significant effect in PE- and SGA-groups (t-test, FDR P<0.05). In post-partum maternal plasma, the highest STC1 hormone levels (ELISA, n = 129) were found in women who had developed PE and delivered a SGA newborn (median 731 vs 418 pg/ml in controls; ANCOVA, P = 0.00048). Significantly higher expression (t-test, FDR P<0.05) of CCNG2 and LYPD6 accompanied with enhanced immunostaining of the protein was detected in placental sections of PE and GDM cases (n = 15). Our study demonstrates the importance of temporal dynamics of placental transcriptional regulation across three trimesters of gestation. Interestingly, many genes with high expression in mid-gestation placenta have also been implicated in adult complex disease, promoting the discussion on the role of placenta in developmental programming. The discovery of elevated maternal plasma STC1 in pregnancy complications warrants further investigations of its potential as a biomarker.
Collapse
|
193
|
Fan H, Zhang R, Tesfaye D, Tholen E, Looft C, Hölker M, Schellander K, Cinar MU. Sulforaphane causes a major epigenetic repression of myostatin in porcine satellite cells. Epigenetics 2012; 7:1379-90. [PMID: 23092945 DOI: 10.4161/epi.22609] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Satellite cells function as skeletal muscle stem cells to support postnatal muscle growth and regeneration following injury or disease. There is great promise for the improvement of muscle performance in livestock and for the therapy of muscle pathologies in humans by the targeting of myostatin (MSTN) in this cell population. Human diet contains many histone deacetylase (HDAC) inhibitors, such as the bioactive component sulforaphane (SFN), whose epigenetic effects on MSTN gene in satellite cells are unknown. Therefore, we aimed to investigate the epigenetic influences of SFN on the MSTN gene in satellite cells. The present work provides the first evidence, which is distinct from the effects of trichostatin A (TSA), that SFN supplementation in vitro not only acts as a HDAC inhibitor but also as a DNA methyltransferase (DNMT) inhibitor in porcine satellite cells. Compared with TSA and 5-aza-2'-deoxycytidine (5-aza-dC), SFN treatment significantly represses MSTN expression, accompanied by strongly attenuated expression of negative feedback inhibitors of the MSTN signaling pathway. miRNAs targeting MSTN are not implicated in posttranscriptional regulation of MSTN. Nevertheless, a weakly enriched myoblast determination (MyoD) protein associated with diminished histone acetylation in the MyoD binding site located in the MSTN promoter region may contribute to the transcriptional repression of MSTN by SFN. These findings reveal a new mode of epigenetic repression of MSTN by the bioactive compound SFN. This novel pharmacological, biological activity of SFN in satellite cells may thus allow for the development of novel approaches to weaken the MSTN signaling pathway, both for therapies of human skeletal muscle disorders and for livestock production improvement.
Collapse
Affiliation(s)
- Huitao Fan
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, Bonn, Germany
| | | | | | | | | | | | | | | |
Collapse
|
194
|
Malik V, Rodino-Klapac LR, Mendell JR. Emerging drugs for Duchenne muscular dystrophy. Expert Opin Emerg Drugs 2012; 17:261-77. [PMID: 22632414 DOI: 10.1517/14728214.2012.691965] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is the most common, severe childhood form of muscular dystrophy. Treatment is limited to glucocorticoids that have the benefit of prolonging ambulation by approximately 2 years and preventing scoliosis. Finding a more satisfactory treatment should focus on maintaining long-term efficacy with a minimal side effect profile. AREAS COVERED Authors discuss different therapeutic strategies that have been used in pre-clinical and clinical settings. EXPERT OPINION Multiple treatment approaches have emerged. Most attractive are molecular-based therapies that can express the missing dystrophin protein (exon skipping or mutation suppression) or a surrogate gene product (utrophin). Other approaches include increasing the strength of muscles (myostatin inhibitors), reducing muscle fibrosis and decreasing oxidative stress. Additional targets include inhibiting NF-κB to reduce inflammation or promoting skeletal muscle blood flow and muscle contractility using phosphodiesterase inhibitors or nitric oxide (NO) donors. The potential for each of these treatment strategies to enter clinical trials is a central theme of discussion. The review emphasizes that the goal of treatment should be to find a product at least as good as glucocorticoids with a lower side effect profile or with a significant glucocorticoid sparing effect.
Collapse
Affiliation(s)
- Vinod Malik
- The Ohio State University, Research Institute, Nationwide Children's Hospital and, Department of Pediatrics, Columbus, OH 43205, USA
| | | | | |
Collapse
|
195
|
Epistatic interactions between Tgfb1 and genetic loci, Tgfbm2 and Tgfbm3, determine susceptibility to an asthmatic stimulus. Proc Natl Acad Sci U S A 2012; 109:18042-7. [PMID: 23064636 DOI: 10.1073/pnas.1205374109] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
TGFβ activation and signaling have been extensively studied in experimental models of allergen-induced asthma as potential therapeutic targets during chronic or acute phases of the disease. Outcomes of experimental manipulation of TGFβ activity have been variable, in part due to use of different model systems. Using an ovalbumin (OVA)-induced mouse model of asthma, we here show that innate variation within TGFβ1 genetic modifier loci, Tgfbm2 and Tgfbm3, alters disease susceptibility. Specifically, Tgfbm2(129) and Tgfbm3(C57) synergize to reverse accentuated airway hyperresponsiveness (AHR) caused by low TGFβ1 levels in Tgfb1(+/-) mice of the NIH/OlaHsd strain. Moreover, epistatic interaction between Tgfbm2(129) and Tgfbm3(C57) uncouples the inflammatory response to ovalbumin from those of airway remodeling and airway hyperresponsiveness, illustrating independent genetic control of these responses. We conclude that differential inheritance of genetic variants of Tgfbm genes alters biological responses to reduced TGFβ1 signaling in an experimental asthma model. TGFβ antagonists for treatment of lung diseases might therefore give diverse outcomes, dependent on genetic variation.
Collapse
|
196
|
Olguín HC, Pisconti A. Marking the tempo for myogenesis: Pax7 and the regulation of muscle stem cell fate decisions. J Cell Mol Med 2012; 16:1013-25. [PMID: 21615681 PMCID: PMC4365881 DOI: 10.1111/j.1582-4934.2011.01348.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Post-natal growth and regeneration of skeletal muscle is highly dependent on a population of resident myogenic precursors known as satellite cells. Transcription factors from the Pax gene family, Pax3 and Pax7, are critical for satellite cell biogenesis, survival and potentially self-renewal; however, the underlying molecular mechanisms remain unsolved. This is particularly true in the case of Pax7, which appears to regulate myogenesis at multiple levels. Accordingly, recent data have highlighted the importance of a functional relationship between Pax7 and the MyoD family of muscle regulatory transcription factors during normal muscle formation and disease. Here we will critically review key findings suggesting that Pax7 may play a dual role by promoting resident muscle progenitors to commit to the skeletal muscle lineage while preventing terminal differentiation, thus keeping muscle progenitors poised to differentiate upon environmental cues. In addition, potential regulatory mechanisms for the control of Pax7 activity will be proposed.
Collapse
Affiliation(s)
- Hugo C Olguín
- Departamento Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | | |
Collapse
|
197
|
Yamada AK, Verlengia R, Bueno Junior CR. Myostatin: genetic variants, therapy and gene doping. BRAZ J PHARM SCI 2012. [DOI: 10.1590/s1984-82502012000300003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Since its discovery, myostatin (MSTN) has been at the forefront of muscle therapy research because intrinsic mutations or inhibition of this protein, by either pharmacological or genetic means, result in muscle hypertrophy and hyperplasia. In addition to muscle growth, MSTN inhibition potentially disturbs connective tissue, leads to strength modulation, facilitates myoblast transplantation, promotes tissue regeneration, induces adipose tissue thermogenesis and increases muscle oxidative phenotype. It is also known that current advances in gene therapy have an impact on sports because of the illicit use of such methods. However, the adverse effects of these methods, their impact on athletic performance in humans and the means of detecting gene doping are as yet unknown. The aim of the present review is to discuss biosynthesis, genetic variants, pharmacological/genetic manipulation, doping and athletic performance in relation to the MSTN pathway. As will be concluded from the manuscript, MSTN emerges as a promising molecule for combating muscle wasting diseases and for triggering wide-ranging discussion in view of its possible use in gene doping.
Collapse
|
198
|
Fanzani A, Conraads VM, Penna F, Martinet W. Molecular and cellular mechanisms of skeletal muscle atrophy: an update. J Cachexia Sarcopenia Muscle 2012; 3:163-79. [PMID: 22673968 PMCID: PMC3424188 DOI: 10.1007/s13539-012-0074-6] [Citation(s) in RCA: 247] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 05/13/2012] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle atrophy is defined as a decrease in muscle mass and it occurs when protein degradation exceeds protein synthesis. Potential triggers of muscle wasting are long-term immobilization, malnutrition, severe burns, aging as well as various serious and often chronic diseases, such as chronic heart failure, obstructive lung disease, renal failure, AIDS, sepsis, immune disorders, cancer, and dystrophies. Interestingly, a cooperation between several pathophysiological factors, including inappropriately adapted anabolic (e.g., growth hormone, insulin-like growth factor 1) and catabolic proteins (e.g., tumor necrosis factor alpha, myostatin), may tip the balance towards muscle-specific protein degradation through activation of the proteasomal and autophagic systems or the apoptotic pathway. Based on the current literature, we present an overview of the molecular and cellular mechanisms that contribute to muscle wasting. We also focus on the multifacetted therapeutic approach that is currently employed to prevent the development of muscle wasting and to counteract its progression. This approach includes adequate nutritional support, implementation of exercise training, and possible pharmacological compounds.
Collapse
Affiliation(s)
- Alessandro Fanzani
- Department of Biomedical Sciences and Biotechnologies and Interuniversitary Institute of Myology (IIM), University of Brescia, viale Europa 11, 25123, Brescia, Italy,
| | | | | | | |
Collapse
|
199
|
Role of satellite cells versus myofibers in muscle hypertrophy induced by inhibition of the myostatin/activin signaling pathway. Proc Natl Acad Sci U S A 2012; 109:E2353-60. [PMID: 22869749 DOI: 10.1073/pnas.1206410109] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Myostatin and activin A are structurally related secreted proteins that act to limit skeletal muscle growth. The cellular targets for myostatin and activin A in muscle and the role of satellite cells in mediating muscle hypertrophy induced by inhibition of this signaling pathway have not been fully elucidated. Here we show that myostatin/activin A inhibition can cause muscle hypertrophy in mice lacking either syndecan4 or Pax7, both of which are important for satellite cell function and development. Moreover, we show that muscle hypertrophy after pharmacological blockade of this pathway occurs without significant satellite cell proliferation and fusion to myofibers and without an increase in the number of myonuclei per myofiber. Finally, we show that genetic ablation of Acvr2b, which encodes a high-affinity receptor for myostatin and activin A specifically in myofibers is sufficient to induce muscle hypertrophy. All of these findings are consistent with satellite cells playing little or no role in myostatin/activin A signaling in vivo and render support that inhibition of this signaling pathway can be an effective therapeutic approach for increasing muscle growth even in disease settings characterized by satellite cell dysfunction.
Collapse
|
200
|
Ohsawa Y, Okada T, Nishimatsu SI, Ishizaki M, Suga T, Fujino M, Murakami T, Uchino M, Tsuchida K, Noji S, Hinohara A, Shimizu T, Shimizu K, Sunada Y. An inhibitor of transforming growth factor beta type I receptor ameliorates muscle atrophy in a mouse model of caveolin 3-deficient muscular dystrophy. J Transl Med 2012; 92:1100-14. [PMID: 22584670 DOI: 10.1038/labinvest.2012.78] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Skeletal muscle expressing Pro104Leu mutant caveolin 3 (CAV3(P104L)) in mouse becomes atrophied and serves as a model of autosomal dominant limb-girdle muscular dystrophy 1C. We previously found that caveolin 3-deficient muscles showed activated intramuscular transforming growth factor beta (TGF-β) signals. However, the cellular mechanism by which loss of caveolin 3 leads to muscle atrophy is unknown. Recently, several small-molecule inhibitors of TGF-β type I receptor (TβRI) kinase have been developed as molecular-targeting drugs for cancer therapy by suppressing intracellular TGF-β1, -β2, and -β3 signaling. Here, we show that a TβRI kinase inhibitor, Ki26894, restores impaired myoblast differentiation in vitro caused by activin, myostatin, and TGF-β1, as well as CAV3(P104L). Oral administration of Ki26894 increased muscle mass and strength in vivo in wild-type mice, and improved muscle atrophy and weakness in the CAV3(P104L) mice. The inhibitor restored the number of satellite cells, the resident stem cells of adult skeletal muscle, with suppression of the increased phosphorylation of Smad2, an effector, and the upregulation of p21 (also known as Cdkn1a), a target gene of the TGF-β family members in muscle. These data indicate that both TGF-β-dependent reduction in satellite cells and impairment of myoblast differentiation contribute to the cellular mechanism underlying caveolin 3-deficient muscle atrophy. TβRI kinase inhibitors could antagonize the activation of intramuscular anti-myogenic TGF-β signals, thereby providing a novel therapeutic rationale for the alternative use of this type of anticancer drug in reversing muscle atrophy in various clinical settings.
Collapse
Affiliation(s)
- Yutaka Ohsawa
- Department of Neurology, Kawasaki Medical School, Okayama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|