51
|
Ji L, Bishayee K, Sadra A, Choi S, Choi W, Moon S, Jho EH, Huh SO. Defective neuronal migration and inhibition of bipolar to multipolar transition of migrating neural cells by Mesoderm-Specific Transcript, Mest, in the developing mouse neocortex. Neuroscience 2017; 355:126-140. [PMID: 28501506 DOI: 10.1016/j.neuroscience.2017.05.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 05/02/2017] [Accepted: 05/02/2017] [Indexed: 12/22/2022]
Abstract
Brain developmental disorders such as lissencephaly can result from faulty neuronal migration and differentiation during the formation of the mammalian neocortex. The cerebral cortex is a modular structure, where developmentally, newborn neurons are generated as a neuro-epithelial sheet and subsequently differentiate, migrate and organize into their final positions in the cerebral cortical plate via a process involving both tangential and radial migration. The specific role of Mest, an imprinted gene, in neuronal migration has not been previously studied. In this work, we reduced expression of Mest with in utero electroporation of neuronal progenitors in the developing embryonic mouse neocortex. Reduction of Mest levels by shRNA significantly reduced the number of neurons migrating to the cortical plate. Also, Mest-knockdown disrupted the transition of bipolar neurons into multipolar neurons migrating out of the sub-ventricular zone region. The migrating neurons also adopted a more tangential migration pattern upon knockdown of the Mest message, losing their potential to attach to radial glia cells, required for radial migration. The differentiation and migration properties of neurons via Wnt-Akt signaling were affected by Mest changes. In addition, miR-335, encoded in a Mest gene intron, was identified as being responsible for blocking the default tangential migration of the neurons. Our results suggest that Mest and its intron product, miR-335, play important roles in neuronal migration with Mest regulating the morphological transition of primary neurons required in the formation of the mammalian neocortex.
Collapse
Affiliation(s)
- Liting Ji
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon, Gangwon-do, South Korea
| | - Kausik Bishayee
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon, Gangwon-do, South Korea
| | - Ali Sadra
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon, Gangwon-do, South Korea
| | - Seunghyuk Choi
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon, Gangwon-do, South Korea
| | - Wooyul Choi
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon, Gangwon-do, South Korea
| | - Sungho Moon
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Eek-Hoon Jho
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Sung-Oh Huh
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon, Gangwon-do, South Korea.
| |
Collapse
|
52
|
Ye DJ, Kwon YJ, Shin S, Baek HS, Shin DW, Chun YJ. Induction of Integrin Signaling by Steroid Sulfatase in Human Cervical Cancer Cells. Biomol Ther (Seoul) 2017; 25:321-328. [PMID: 27956712 PMCID: PMC5424643 DOI: 10.4062/biomolther.2016.155] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 09/02/2016] [Accepted: 10/04/2016] [Indexed: 12/19/2022] Open
Abstract
Steroid sulfatase (STS) is an enzyme responsible for the hydrolysis of aryl and alkyl sulfates. STS plays a pivotal role in the regulation of estrogens and androgens that promote the growth of hormone-dependent tumors, such as those of breast or prostate cancer. However, the molecular function of STS in tumor growth is still not clear. To elucidate the role of STS in cancer cell proliferation, we investigated whether STS is able to regulate the integrin signaling pathway. We found that overexpression of STS in HeLa cells increases the protein and mRNA levels of integrin β1 and fibronectin, a ligand of integrin α5β1. Dehydroepiandrosterone (DHEA), one of the main metabolites of STS, also increases mRNA and protein expression of integrin β1 and fibronectin. Further, STS expression and DHEA treatment enhanced phosphorylation of focal adhesion kinase (FAK) at the Tyr 925 residue. Moreover, increased phosphorylation of ERK at Thr 202 and Tyr 204 residues by STS indicates that STS activates the MAPK/ERK pathway. In conclusion, these results suggest that STS expression and DHEA treatment may enhance MAPK/ERK signaling through up-regulation of integrin β1 and activation of FAK.
Collapse
Affiliation(s)
- Dong-Jin Ye
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yeo-Jung Kwon
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sangyun Shin
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hyoung-Seok Baek
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Dong-Won Shin
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Young-Jin Chun
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
53
|
Wang H, Shi L, Liang T, Wang B, Wu W, Su G, Wei J, Li P, Huang R. MiR-696 Regulates C2C12 Cell Proliferation and Differentiation by Targeting CNTFRα. Int J Biol Sci 2017; 13:413-425. [PMID: 28529450 PMCID: PMC5436562 DOI: 10.7150/ijbs.17508] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/06/2017] [Indexed: 12/28/2022] Open
Abstract
Micro-696 (miR-696) has been previously known as an exercise related miRNA, which has a profound role in fatty acid oxidation and mitochondrial biogenesis of skeletal muscle. However, its role in skeletal myoblast proliferation and differentiation is still unclear. In this study, we found that miR-696 expressed highly in skeletal muscle and reduced during C2C12 myoblasts differentiation. MiR-696 overexpression repressed C2C12 myoblast proliferation and myofiber formation, while knockdown of endogenous miR-696 expression showed opposite results. During myogenesis, we observed an inversed expression pattern between miR-696 and CNTFRα in vitro, and demonstrated that miR-696 could specifically target CNTFRα and repress the expression of CNTFRα. Additionally, we further found that knockdown of CNTFRα suppressed the proliferation and differentiation of C2C12 cells. Taking all things together, we propose a novel insight that miR-696 down-regulates C2C12 cell myogenesis by inhibiting CNTFRα expression.
Collapse
Affiliation(s)
- Han Wang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Lei Shi
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Tingting Liang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - BinBin Wang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - WangJun Wu
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Guosheng Su
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
| | - Julong Wei
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Pinghua Li
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ruihua Huang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
54
|
Kocsis T, Trencsenyi G, Szabo K, Baan JA, Muller G, Mendler L, Garai I, Reinauer H, Deak F, Dux L, Keller-Pinter A. Myostatin propeptide mutation of the hypermuscular Compact mice decreases the formation of myostatin and improves insulin sensitivity. Am J Physiol Endocrinol Metab 2017; 312:E150-E160. [PMID: 27965203 DOI: 10.1152/ajpendo.00216.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 11/29/2016] [Accepted: 11/30/2016] [Indexed: 12/30/2022]
Abstract
The TGFβ family member myostatin (growth/differentiation factor-8) is a negative regulator of skeletal muscle growth. The hypermuscular Compact mice carry the 12-bp Mstn(Cmpt-dl1Abc) deletion in the sequence encoding the propeptide region of the precursor promyostatin, and additional modifier genes of the Compact genetic background contribute to determine the full expression of the phenotype. In this study, by using mice strains carrying mutant or wild-type myostatin alleles with the Compact genetic background and nonmutant myostatin with the wild-type background, we studied separately the effect of the Mstn(Cmpt-dl1Abc) mutation or the Compact genetic background on morphology, metabolism, and signaling. We show that both the Compact myostatin mutation and Compact genetic background account for determination of skeletal muscle size. Despite the increased musculature of Compacts, the absolute size of heart and kidney is not influenced by myostatin mutation; however, the Compact genetic background increases them. Both Compact myostatin and genetic background exhibit systemic metabolic effects. The Compact mutation decreases adiposity and improves whole body glucose uptake, insulin sensitivity, and 18FDG uptake of skeletal muscle and white adipose tissue, whereas the Compact genetic background has the opposite effect. Importantly, the mutation does not prevent the formation of mature myostatin; however, a decrease in myostatin level was observed, leading to altered activation of Smad2, Smad1/5/8, and Akt, and an increased level of p-AS160, a Rab-GTPase-activating protein responsible for GLUT4 translocation. Based on our analysis, the Compact genetic background strengthens the effect of myostatin mutation on muscle mass, but those can compensate for each other when systemic metabolic effects are compared.
Collapse
Affiliation(s)
- Tamas Kocsis
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | | | - Kitti Szabo
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Julia Aliz Baan
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Geza Muller
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Luca Mendler
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | | | | | - Ferenc Deak
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Laszlo Dux
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Aniko Keller-Pinter
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary;
| |
Collapse
|
55
|
Sakuma K, Aoi W, Yamaguchi A. Molecular mechanism of sarcopenia and cachexia: recent research advances. Pflugers Arch 2017; 469:573-591. [PMID: 28101649 DOI: 10.1007/s00424-016-1933-3] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 12/28/2016] [Accepted: 12/29/2016] [Indexed: 12/17/2022]
Abstract
Skeletal muscle provides a fundamental basis for human function, enabling locomotion and respiration. Muscle loss occurs as a consequence of several chronic diseases (cachexia) and normal aging (sarcopenia). Although many negative regulators (atrogin-1, muscle ring finger-1, nuclear factor-kappaB (NF-κB), myostatin, etc.) have been proposed to enhance protein degradation during both sarcopenia and cachexia, the adaptation of these mediators markedly differs within both conditions. Sarcopenia and cachectic muscles have been demonstrated to be abundant in myostatin-linked molecules. The ubiquitin-proteasome system (UPS) is activated during rapid atrophy model (cancer cachexia), but few mediators of the UPS change during sarcopenia. NF-κB signaling is activated in cachectic, but not in sarcopenic, muscle. Recent studies have indicated the age-related defect of autophagy signaling in skeletal muscle, whereas autophagic activation occurs in cachectic muscle. This review provides recent research advances dealing with molecular mediators modulating muscle mass in both sarcopenia and cachexia.
Collapse
Affiliation(s)
- Kunihiro Sakuma
- Institute for Liberal Arts, Environment and Society, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro-ku, Tokyo, 152-8550, Japan.
| | - Wataru Aoi
- Laboratory of Health Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-cho Shimogamo, Sakyo-ku, Kyoto, 606-8522, Japan
| | - Akihiko Yamaguchi
- Department of Physical Therapy, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| |
Collapse
|
56
|
Walker RG, Poggioli T, Katsimpardi L, Buchanan SM, Oh J, Wattrus S, Heidecker B, Fong YW, Rubin LL, Ganz P, Thompson TB, Wagers AJ, Lee RT. Biochemistry and Biology of GDF11 and Myostatin: Similarities, Differences, and Questions for Future Investigation. Circ Res 2016; 118:1125-41; discussion 1142. [PMID: 27034275 DOI: 10.1161/circresaha.116.308391] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 03/07/2016] [Indexed: 02/06/2023]
Abstract
Growth differentiation factor 11 (GDF11) and myostatin (or GDF8) are closely related members of the transforming growth factor β superfamily and are often perceived to serve similar or overlapping roles. Yet, despite commonalities in protein sequence, receptor utilization and signaling, accumulating evidence suggests that these 2 ligands can have distinct functions in many situations. GDF11 is essential for mammalian development and has been suggested to regulate aging of multiple tissues, whereas myostatin is a well-described negative regulator of postnatal skeletal and cardiac muscle mass and modulates metabolic processes. In this review, we discuss the biochemical regulation of GDF11 and myostatin and their functions in the heart, skeletal muscle, and brain. We also highlight recent clinical findings with respect to a potential role for GDF11 and/or myostatin in humans with heart disease. Finally, we address key outstanding questions related to GDF11 and myostatin dynamics and signaling during development, growth, and aging.
Collapse
Affiliation(s)
- Ryan G Walker
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Tommaso Poggioli
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Lida Katsimpardi
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Sean M Buchanan
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Juhyun Oh
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Sam Wattrus
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Bettina Heidecker
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Yick W Fong
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Lee L Rubin
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Peter Ganz
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Thomas B Thompson
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Amy J Wagers
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.).
| | - Richard T Lee
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.).
| |
Collapse
|
57
|
Ou Y, Ho WS. Crocodile blood extract induces the apoptosis of lung cancer cells through PTEN activity. Oncol Rep 2016; 36:1457-66. [DOI: 10.3892/or.2016.4914] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/17/2016] [Indexed: 11/05/2022] Open
|
58
|
Monestier O, Blanquet V. WFIKKN1 and WFIKKN2: "Companion" proteins regulating TGFB activity. Cytokine Growth Factor Rev 2016; 32:75-84. [PMID: 27325460 DOI: 10.1016/j.cytogfr.2016.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 01/14/2023]
Abstract
The WFIKKN (WAP, Follistatin/kazal, Immunoglobulin, Kunitz and Netrin domain-containing) protein family is composed of two multidomain proteins: WFIKKN1 and WFIKKN2. They were formed by domain shuffling and are likely present in deuterostoms. The WFIKKN (also called GASP) proteins are well known for their function in muscle and skeletal tissues, namely, inhibition of certain members of the transforming growth factor beta (TGFB) superfamily such as myostatin (MSTN) and growth and differentiation factor 11 (GDF11). However, the role of the WFIKKN proteins in other tissues is still poorly understood in spite of evidence suggesting possible action in the inner ear, brain and reproduction. Further, several recent studies based on next generation technologies revealed differential expression of WFIKKN1 and WFIKKN2 in various tissues suggesting that their function is not limited to MSTN and GDF11 inhibition in musculoskeletal tissue. In this review, we summarize current knowledge about the WFIKKN proteins and propose that they are "companion" proteins for various growth factors by providing localized and sustained presentation of TGFB proteins to their respective receptors, thus regulating the balance between the activation of Smad and non-Smad pathways by TGFB.
Collapse
Affiliation(s)
- Olivier Monestier
- INRA, UR1037 Laboratory of Fish Physiology and Genomic, Growth and Flesh Quality Group, Campus de Beaulieu, 35000 Rennes, France.
| | - Véronique Blanquet
- INRA, UMR1061 Unité de Génétique Moléculaire Animale, 87060 Limoges, France; Université de Limoges, 87060 Limoges, France.
| |
Collapse
|
59
|
Mo C, Zhao R, Vallejo J, Igwe O, Bonewald L, Wetmore L, Brotto M. Prostaglandin E2 promotes proliferation of skeletal muscle myoblasts via EP4 receptor activation. Cell Cycle 2016; 14:1507-16. [PMID: 25785867 PMCID: PMC4615122 DOI: 10.1080/15384101.2015.1026520] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
We recently demonstrated that conditioned media (CM) from osteocytes enhances myogenic differentiation of myoblasts, suggesting that signaling from bone may be important for skeletal muscle myogenesis. The effect of CM was closely mimicked by prostaglandin E2 (PGE2), a bioactive lipid mediator in various physiological or pathological conditions. PGE2 is secreted at high levels by osteocytes and such secretion is further enhanced under loading conditions. Although four types of receptors, EP1 to EP4, mediate PGE2 signaling, it is unknown whether these receptors play a role in myogenesis. Therefore, in this study, the expression of EPs in mouse primary myoblasts was characterized, followed by examination of their roles in myoblast proliferation by treating myoblasts with PGE2 or specific agonists. All four PGE2 receptor mRNAs were detectable by quantitative real-time PCR (qPCR), but only PGE2 and EP4 agonist CAY 10598 significantly enhance myoblast proliferation. EP1/EP3 agonist 17-phenyl trinor PGE2 (17-PT PGE2) and EP2 agonist butaprost did not have any significant effects. Moreover, treatment with EP4 antagonist L161,982 dose-dependently inhibited myoblast proliferation. These results were confirmed by cell cycle analysis and the gene expression of cell cycle regulators. Concomitant with the inhibition of myoblast proliferation, treatment with L161,982 significantly increased intracellular reactive oxygen species (ROS) levels. Cotreatment with antioxidant N-acetyl cysteine (NAC) or sodium ascorbate (SA) successfully reversed the inhibition of myoblast proliferation and ROS overproduction caused by L161,982. Therefore, PGE2 signaling via the EP4 receptor regulates myogenesis by promoting myoblast proliferation and blocking this receptor results in increased ROS production in myoblasts.
Collapse
Key Words
- CDK, cyclin dependent kinase
- CM, conditioned media
- EP4
- Keap1/Nrf2, Kelch-like ECH-associated protein 1/NF-E2-related factor 2
- NAC, N-acetyl cysteine
- PGC-1α, proliferator-activated receptor gamma coactivator 1-α
- PGD2, prostaglandin D2
- PGE2, prostaglandin E2
- PGF2α, prostaglandin F2α; PGI2, prostaglandin I2
- Prostaglandin E2
- RB, retinoblastoma protein
- ROS, reactive oxygen species
- SA, sodium ascorbate
- SOD1, superoxide dismutase 1
- bone-muscle crosstalk
- myogenesis
- proliferation
- qPCR, quantitative real-time PCR
- reactive oxygen species
Collapse
Affiliation(s)
- Chenglin Mo
- a Muscle Biology Research Group-MUBIG ; School of Nursing and Health Studies; University of Missouri-Kansas City ; Kansas City , MO USA
| | | | | | | | | | | | | |
Collapse
|
60
|
Fan J, Kou X, Jia S, Yang X, Yang Y, Chen N. Autophagy as a Potential Target for Sarcopenia. J Cell Physiol 2015; 231:1450-9. [DOI: 10.1002/jcp.25260] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 11/17/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Jingjing Fan
- College of Health Science; Hubei Provincial Collaborative Innovation Center for Exercise and Health Promotion; Wuhan Sports University; Wuhan China
- Hubei Exercise Training and Monitoring Key Laboratory; Wuhan Sports University; Wuhan China
| | - Xianjuan Kou
- College of Health Science; Hubei Provincial Collaborative Innovation Center for Exercise and Health Promotion; Wuhan Sports University; Wuhan China
- Hubei Exercise Training and Monitoring Key Laboratory; Wuhan Sports University; Wuhan China
| | - Shaohui Jia
- College of Health Science; Hubei Provincial Collaborative Innovation Center for Exercise and Health Promotion; Wuhan Sports University; Wuhan China
- Hubei Exercise Training and Monitoring Key Laboratory; Wuhan Sports University; Wuhan China
| | - Xiaoqi Yang
- Graduate School; Wuhan Sports University; Wuhan China
| | - Yi Yang
- Hubei Exercise Training and Monitoring Key Laboratory; Wuhan Sports University; Wuhan China
| | - Ning Chen
- College of Health Science; Hubei Provincial Collaborative Innovation Center for Exercise and Health Promotion; Wuhan Sports University; Wuhan China
- Hubei Exercise Training and Monitoring Key Laboratory; Wuhan Sports University; Wuhan China
| |
Collapse
|
61
|
Santra M, Chopp M, Santra S, Nallani A, Vyas S, Zhang ZG, Morris DC. Thymosin beta 4 up-regulates miR-200a expression and induces differentiation and survival of rat brain progenitor cells. J Neurochem 2015; 136:118-32. [PMID: 26466330 DOI: 10.1111/jnc.13394] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/27/2015] [Accepted: 10/05/2015] [Indexed: 12/24/2022]
Abstract
Thymosin beta 4 (Tβ4), a secreted 43 amino acid peptide, promotes oligodendrogenesis, and improves neurological outcome in rat models of neurologic injury. We demonstrated that exogenous Tβ4 treatment up-regulated the expression of the miR-200a in vitro in rat brain progenitor cells and in vivo in the peri-infarct area of rats subjected to middle cerebral artery occlusion (MCAO). The up-regulation of miR-200a down-regulated the expression of the following targets in vitro and in vivo models: (i) growth factor receptor-bound protein 2 (Grb2), an adaptor protein involved in epidermal growth factor receptor (EGFR)/Grb2/Ras/MEK/ERK1/c-Jun signaling pathway, which negatively regulates the expression of myelin basic protein (MBP), a marker of mature oligodendrocyte; (ii) ERRFI-1/Mig-6, an endogenous potent kinase inhibitor of EGFR, which resulted in activation/phosphorylation of EGFR; (iii) friend of GATA 2, and phosphatase and tensin homolog deleted in chromosome 10 (PTEN), which are potent inhibitors of the phosphatidylinositol-3-kinase (PI3K)/AKT signaling pathway, and resulted in marked activation of AKT; and (iv) transcription factor, p53, which induces pro-apoptotic genes, and possibly reduced apoptosis of the progenitor cells subjected to oxygen glucose deprivation (OGD). Anti-miR-200a transfection reversed all the effects of Tβ4 treatment in vitro. Thus, Tβ4 up-regulated MBP synthesis, and inhibited OGD-induced apoptosis in a novel miR-200a dependent EGFR signaling pathway. Our findings of miR-200a-mediated protection of progenitor cells may provide a new therapeutic importance for the treatment of neurologic injury. Tβ4-induced micro-RNA-200a (miR-200a) regulates EGFR signaling pathways for MBP synthesis and apoptosis: up-regulation of miR-200a after Tβ4 treatment, increases MBP synthesis after targeting Grb2 and thereby inactivating c-Jun from inhibition of MBP synthesis; and also inhibits OGD-mediated apoptosis after targeting EGFR inhibitor (Mig-6), PI3K inhibitors (FOG2 and Pten) and an inducer (p53) of pro-apoptotic genes, for AKT activation and down-regulation of p53. These findings may contribute the therapeutic benefits for stroke and other neuronal diseases associated with demyelination disorders.
Collapse
Affiliation(s)
- Manoranjan Santra
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA.,Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Sutapa Santra
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Ankita Nallani
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Shivam Vyas
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Daniel C Morris
- Department of Emergency Medicine, Henry Ford Health System, Detroit, Michigan, USA
| |
Collapse
|
62
|
Jing X, Cheng W, Wang S, Li P, He L. Resveratrol induces cell cycle arrest in human gastric cancer MGC803 cells via the PTEN-regulated PI3K/Akt signaling pathway. Oncol Rep 2015; 35:472-8. [PMID: 26530632 DOI: 10.3892/or.2015.4384] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 10/01/2015] [Indexed: 12/22/2022] Open
Abstract
Resveratrol is a polyphenolic compound that is extracted from Polygonum cuspidatum and is used in traditional Chinese medicine. Previous data have shown that resveratrol inhibits the growth of human gastric cancer. MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide] and trypan blue assays showed that resveratrol significantly decreased the survival rate of MGC803 cells in a concentration- and time-dependent manner. Our flow cytometric analysis showed that resveratrol treatment arrested the cells at the G0/G1 phase of the cell cycle. Furthermore, western blotting demonstrated that resveratrol decreased the protein expression of phospho-glycogen synthase kinase 3β (p-GSK3β), cyclin D1, phospho-phosphatase and tensin homologue (p-PTEN), phospho-phosphatidylinositol 3'-OH kinase (p-PI3K), and phospho-protein kinase B (p-PKB/Akt). We also found that resveratrol inhibited the progression of the cell cycle in MGC803 cells by repressing p-PI3K and p-Akt expression. Meanwhile, resveratrol did not decrease the phosphorylation level of Akt when the PTEN gene expression was knocked down by an siRNA in the MGC803 cells. Taken together, these results suggest that resveratrol induced cell cycle arrest in human gastric cancer MGC803 cells by regulating the PTEN/PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Xiaoping Jing
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, P.R. China
| | - Weiwei Cheng
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, P.R. China
| | - Shiying Wang
- Department of General Surgery, Putuo Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Pin Li
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, P.R. China
| | - Li He
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, P.R. China
| |
Collapse
|
63
|
Sasi Kumar K, Ramadhas A, Nayak S, Kaniyappan S, Dayma K, Radha V. C3G (RapGEF1), a regulator of actin dynamics promotes survival and myogenic differentiation of mouse mesenchymal cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2629-39. [DOI: 10.1016/j.bbamcr.2015.06.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/17/2015] [Accepted: 06/27/2015] [Indexed: 12/11/2022]
|
64
|
Liu SH, Yang RS, Yen YP, Chiu CY, Tsai KS, Lan KC. Low-Concentration Arsenic Trioxide Inhibits Skeletal Myoblast Cell Proliferation via a Reactive Oxygen Species-Independent Pathway. PLoS One 2015; 10:e0137907. [PMID: 26359868 PMCID: PMC4567280 DOI: 10.1371/journal.pone.0137907] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 08/22/2015] [Indexed: 12/25/2022] Open
Abstract
Myoblast proliferation and differentiation are essential for skeletal muscle regeneration. Myoblast proliferation is a critical step in the growth and maintenance of skeletal muscle. The precise action of inorganic arsenic on myoblast growth has not been investigated. Here, we investigated the in vitro effect of inorganic arsenic trioxide (As2O3) on the growth of C2C12 myoblasts. As2O3 decreased myoblast growth at submicromolar concentrations (0.25–1 μM) after 72 h of treatment. Submicromolar concentrations of As2O3 did not induce the myoblast apoptosis. Low-concentration As2O3 (0.5 and 1 μM) significantly suppressed the myoblast cell proliferative activity, which was accompanied by a small proportion of bromodeoxyuridine (BrdU) incorporation and decreased proliferating cell nuclear antigen (PCNA) protein expression. As2O3 (0.5 and 1 μM) increased the intracellular arsenic content but did not affect the reactive oxygen species (ROS) levels in the myoblasts. Cell cycle analysis indicated that low-concentrations of As2O3 inhibited cell proliferation via cell cycle arrest in the G1 and G2/M phases. As2O3 also decreased the protein expressions of cyclin D1, cyclin E, cyclin B1, cyclin-dependent kinase (CDK) 2, and CDK4, but did not affect the protein expressions of p21 and p27. Furthermore, As2O3 inhibited the phosphorylation of Akt. Insulin-like growth factor-1 significantly reversed the inhibitory effect of As2O3 on Akt phosphorylation and cell proliferation in the myoblasts. These results suggest that submicromolar concentrations of As2O3 alter cell cycle progression and reduce myoblast proliferation, at least in part, through a ROS-independent Akt inhibition pathway.
Collapse
Affiliation(s)
- Shing Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Rong-Sen Yang
- Departments of Orthopaedic, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan
| | - Yuan-Peng Yen
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Yuan Chiu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Keh-Sung Tsai
- Departments of Laboratory Medicine, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan
| | - Kuo-Cheng Lan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
65
|
Ahima RS, Park HK. Connecting Myokines and Metabolism. Endocrinol Metab (Seoul) 2015; 30:235-45. [PMID: 26248861 PMCID: PMC4595346 DOI: 10.3803/enm.2015.30.3.235] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 06/22/2015] [Accepted: 06/29/2015] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle is the largest organ of the body in non-obese individuals and is now considered to be an endocrine organ. Hormones (myokines) secreted by skeletal muscle mediate communications between muscle and liver, adipose tissue, brain, and other organs. Myokines affect muscle mass and myofiber switching, and have profound effects on glucose and lipid metabolism and inflammation, thus contributing to energy homeostasis and the pathogenesis of obesity, diabetes, and other diseases. In this review, we summarize recent findings on the biology of myokines and provide an assessment of their potential as therapeutic targets.
Collapse
Affiliation(s)
- Rexford S Ahima
- Division of Endocrinology, Diabetes and Metabolism, and the Institute for Diabetes, Obesity and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Hyeong Kyu Park
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Seoul, Korea
| |
Collapse
|
66
|
Gao CQ, Zhang HJ, Yan HC, Yuan L, Dahanayaka S, Li HC, Wang XQ. Satellite cells isolated from skeletal muscle will proliferate faster in WENS yellow feather chicks. Anim Sci J 2015; 87:126-33. [DOI: 10.1111/asj.12403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 01/31/2015] [Accepted: 02/03/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Chun-Qi Gao
- College of Animal Science; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding/ Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture / South China Collaborative Innovation Center for Poultry Disease Control and Product Safety; South China Agricultural University; Guangzhou China
| | - Hao-Jie Zhang
- College of Animal Science; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding/ Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture / South China Collaborative Innovation Center for Poultry Disease Control and Product Safety; South China Agricultural University; Guangzhou China
| | - Hui-Chao Yan
- College of Animal Science; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding/ Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture / South China Collaborative Innovation Center for Poultry Disease Control and Product Safety; South China Agricultural University; Guangzhou China
| | - Li Yuan
- State Key Laboratory of Cellular Stress Biology and Department of Biomedical Sciences; School of Life Sciences; Xiamen University; Xiamen China
| | - Sudath Dahanayaka
- Department of Animal Sciences; Texas A&M University; College Station TX USA
| | - Hai-Chang Li
- Davis Heart & Lung Research Institute; Wexner Medical Center at the Ohio State University; Columbus OH USA
| | - Xiu-Qi Wang
- College of Animal Science; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding/ Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture / South China Collaborative Innovation Center for Poultry Disease Control and Product Safety; South China Agricultural University; Guangzhou China
| |
Collapse
|
67
|
Wu R, Li H, Li T, Zhang Y, Zhu D. Myostatin regulates miR-431 expression via the Ras-Mek-Erk signaling pathway. Biochem Biophys Res Commun 2015; 461:224-9. [DOI: 10.1016/j.bbrc.2015.03.150] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 03/26/2015] [Indexed: 11/26/2022]
|
68
|
Yu M, Wang H, Xu Y, Yu D, Li D, Liu X, Du W. Insulin-like growth factor-1 (IGF-1) promotes myoblast proliferation and skeletal muscle growth of embryonic chickens via the PI3K/Akt signalling pathway. Cell Biol Int 2015; 39:910-22. [PMID: 25808997 DOI: 10.1002/cbin.10466] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/26/2015] [Indexed: 11/07/2022]
Abstract
During embryonic development, IGF-1 fulfils crucial roles in skeletal myogenesis. However, the involvement of IGF-1-induced myoblast proliferation in muscle growth is still unclear. In the present study, we have characterised the role of IGF-1 in myoblast proliferation both in vitro and in vivo and have revealed novel details of how exogenous IGF-1 influences myogenic genes in chicken embryos. The results show that IGF-1 significantly induces the proliferation of cultured myoblasts in a dose-dependent manner. Additionally, the IGF-1 treatment significantly promoted myoblasts entering a new cell cycle and increasing the mRNA expression levels of cell cycle-dependent genes. However, these effects were inhibited by the PI3K inhibitor LY294002 and the Akt inhibitor KP372-1. These data indicated that the pro-proliferative effect of IGF-1 was mediated in response to the PI3K/Akt signalling pathway. Moreover, we also showed that exogenous IGF-1 stimulated myoblast proliferation in vivo. IGF-1 administration obviously promoted the incorporation of BrdU and remarkably increased the number of PAX7-positive cells in the skeletal muscle of chicken embryos. Administration of IGF-1 also significantly induced the upregulation of myogenic factors gene, the enhancement of c-Myc and the inhibition of myostatin (Mstn) expression. These findings demonstrate that IGF-1 has strong activity as a promoter of myoblast expansion and muscle fiber formation during early myogenesis. Therefore, this study offers insight into the mechanisms responsible for IGF-1-mediated stimulation of embryonic skeletal muscle development, which could have important implications for the improvement of chicken meat production.
Collapse
Affiliation(s)
- Minli Yu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, PR China
| | - Huan Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, PR China
| | - Yali Xu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, PR China
| | - Debing Yu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, PR China
| | - Dongfeng Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, PR China
| | - Xiuhong Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, PR China
| | - Wenxing Du
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, PR China
| |
Collapse
|
69
|
Wei C, Ren H, Xu L, Li L, Liu R, Zhang L, Zhao F, Lu J, Zhang X, Du L. Signals of Ezh2, Src, and Akt Involve in myostatin-Pax7 pathways regulating the myogenic fate determination during the sheep myoblast proliferation and differentiation. PLoS One 2015; 10:e0120956. [PMID: 25811841 PMCID: PMC4374906 DOI: 10.1371/journal.pone.0120956] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 02/11/2015] [Indexed: 12/19/2022] Open
Abstract
Myostatin and Pax7 have been well documented individually, however, the mechanism by which Myostatin regulates Pax7 is seldom reported. Here, based on muscle transcriptome analysis in Texel (Myostatin mutant) and Ujumqin (wild type) sheep across the five fetal stages, we constructed and examined the Myostatin-Pax7 pathways in muscle. Then we validated the signals by RNAi in the proliferating and differentiating sheep myoblasts in vitro at mRNA, protein, and cell morphological levels. We reveal that Myostatin signals to Pax7 at least through Ezh2, Src, and Akt during the sheep myoblast proliferation and differentiation. Other signals such as p38MAPK, mTOR, Erk1/2, Wnt, Bmp2, Smad, Tgfb1, and p21 are most probably involved in the Myostatin-affected myogenic events. Myostatin knockdown significantly reduces the counts of nucleus and myotube, but not the fusion index of myoblasts during cell differentiation. In addition, findings also indicate that Myostatin is required for normal myogenic differentiation of the sheep myoblasts, which is different from the C2C12 myoblasts. We expand the regulatory network of Myostatin-Pax7 pathways and first illustrate that Myostatin as a global regulator participates in the epigenetic events involved in myogenesis, which contributes to understand the molecular mechanism of Myostatin in regulation of myogenesis.
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
| | - Hangxing Ren
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, China
| | - Lingyang Xu
- National Center for Molecular Genetics and Breeding of Animal, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Li Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Ruizao Liu
- 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
| | - Fuping Zhao
- 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
| | - Xiaoning Zhang
- National Center for Molecular Genetics and Breeding of Animal, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lixin Du
- National Center for Molecular Genetics and Breeding of Animal, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- * E-mail:
| |
Collapse
|
70
|
Kamiide Y, Furuya M, Inomata N, Yada T. Chronic exposure to cigarette smoke causes extrapulmonary abnormalities in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 39:864-70. [PMID: 25770835 DOI: 10.1016/j.etap.2015.02.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/19/2015] [Accepted: 02/24/2015] [Indexed: 06/04/2023]
Abstract
Pathophysiological features of chronic obstructive pulmonary disease (COPD) include systemic abnormalities, such as weight loss and skeletal muscle wasting. Although cigarette smoke (CS) is a major risk factor in COPD, the systemic effects of CS exposure remain to be elucidated. In this study, rats were exposed to CS or smoke-free air for 12 weeks. CS-exposed rats developed emphysema and had significantly lower body weight and food intake than control rats. The plasma ghrelin levels significantly increased with an upregulation of gastric ghrelin mRNA expression induced by CS exposure. Further, we observed low plasma insulin-like growth factor-1 levels and high tumor necrosis factor-α levels. A significant reduction of skeletal muscle strength and an increase in the mRNA expression of catabolic factors was observed in CS-exposed rats. These results indicated that chronic CS exposure induced not only pulmonary emphysema but also systemic abnormalities related to muscle catabolism associated with inflammatory responses.
Collapse
Affiliation(s)
- Yoshiyuki Kamiide
- Faculty of Pharmacology I, Asubio Pharma Co., Ltd., 6-4-3, Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, 3311-1, Shimotsuke, Tochigi 329-0498, Japan.
| | - Mayumi Furuya
- Faculty of Pharmacology I, Asubio Pharma Co., Ltd., 6-4-3, Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| | - Norio Inomata
- Faculty of Pharmacology I, Asubio Pharma Co., Ltd., 6-4-3, Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| | - Toshihiko Yada
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, 3311-1, Shimotsuke, Tochigi 329-0498, Japan.
| |
Collapse
|
71
|
Mendias CL, Lynch EB, Gumucio JP, Flood MD, Rittman DS, Van Pelt DW, Roche SM, Davis CS. Changes in skeletal muscle and tendon structure and function following genetic inactivation of myostatin in rats. J Physiol 2015; 593:2037-52. [PMID: 25640143 DOI: 10.1113/jphysiol.2014.287144] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/23/2015] [Indexed: 11/08/2022] Open
Abstract
Myostatin is a negative regulator of skeletal muscle and tendon mass. Myostatin deficiency has been well studied in mice, but limited data are available on how myostatin regulates the structure and function of muscles and tendons of larger animals. We hypothesized that, in comparison to wild-type (MSTN(+/+) ) rats, rats in which zinc finger nucleases were used to genetically inactivate myostatin (MSTN(Δ/Δ) ) would exhibit an increase in muscle mass and total force production, a reduction in specific force, an accumulation of type II fibres and a decrease and stiffening of connective tissue. Overall, the muscle and tendon phenotype of myostatin-deficient rats was markedly different from that of myostatin-deficient mice, which have impaired contractility and pathological changes to fibres and their extracellular matrix. Extensor digitorum longus and soleus muscles of MSTN(Δ/Δ) rats demonstrated 20-33% increases in mass, 35-45% increases in fibre number, 20-57% increases in isometric force and no differences in specific force. The insulin-like growth factor-1 pathway was activated to a greater extent in MSTN(Δ/Δ) muscles, but no substantial differences in atrophy-related genes were observed. Tendons of MSTN(Δ/Δ) rats had a 20% reduction in peak strain, with no differences in mass, peak stress or stiffness. The general morphology and gene expression patterns were similar between tendons of both genotypes. This large rodent model of myostatin deficiency did not have the negative consequences to muscle fibres and extracellular matrix observed in mouse models, and suggests that the greatest impact of myostatin in the regulation of muscle mass may not be to induce atrophy directly, but rather to block hypertrophy signalling.
Collapse
Affiliation(s)
- Christopher L Mendias
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Baán JA, Varga ZV, Leszek P, Kuśmierczyk M, Baranyai T, Dux L, Ferdinandy P, Braun T, Mendler L. Myostatin and IGF-I signaling in end-stage human heart failure: a qRT-PCR study. J Transl Med 2015; 13:1. [PMID: 25591711 PMCID: PMC4301667 DOI: 10.1186/s12967-014-0365-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/16/2014] [Indexed: 12/31/2022] Open
Abstract
Background Myostatin (Mstn) is a key regulator of heart metabolism and cardiomyocyte growth interacting tightly with insulin-like growth factor I (IGF-I) under physiological conditions. The pathological role of Mstn has also been suggested since Mstn protein was shown to be upregulated in the myocardium of end-stage heart failure. However, no data are available about the regulation of gene expression of Mstn and IGF-I in different regions of healthy or pathologic human hearts, although they both might play a crucial role in the pathomechanism of heart failure. Methods In the present study, heart samples were collected from left ventricles, septum and right ventricles of control healthy individuals as well as from failing hearts of dilated (DCM) or ischemic cardiomyopathic (ICM) patients. A comprehensive qRT-PCR analysis of Mstn and IGF-I signaling was carried out by measuring expression of Mstn, its receptor Activin receptor IIB (ActRIIB), IGF-I, IGF-I receptor (IGF-IR), and the negative regulator of Mstn miR-208, respectively. Moreover, we combined the measured transcript levels and created complex parameters characterizing either Mstn- or IGF-I signaling in the different regions of healthy or failing hearts. Results We have found that in healthy control hearts, the ratio of Mstn/IGF-I signaling was significantly higher in the left ventricle/septum than in the right ventricle. Moreover, Mstn transcript levels were significantly upregulated in all heart regions of DCM but not ICM patients. However, the ratio of Mstn/IGF-I signaling remained increased in the left ventricle/septum compared to the right ventricle of DCM patients (similarly to the healthy hearts). In contrast, in ICM hearts significant transcript changes were detected mainly in IGF-I signaling. In paralell with these results miR-208 showed mild upregulation in the left ventricle of both DCM and ICM hearts. Conclusions This is the first demonstration of a spatial asymmetry in the expression pattern of Mstn/IGF-I in healthy hearts, which is likely to play a role in the different growth regulation of left vs. right ventricle. Moreover, we identified Mstn as a massively regulated gene in DCM but not in ICM as part of possible compensatory mechanisms in the failing heart.
Collapse
|
73
|
Jeffery N, Mendias C. Endocranial and masticatory muscle volumes in myostatin-deficient mice. ROYAL SOCIETY OPEN SCIENCE 2014; 1:140187. [PMID: 26064569 PMCID: PMC4448778 DOI: 10.1098/rsos.140187] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 08/15/2014] [Indexed: 06/04/2023]
Abstract
Structural and functional trade-offs are integral to the evolution of the mammalian skull and its development. This paper examines the potential for enlargement of the masticatory musculature to limit the size of the endocranial cavity by studying a myostatin-deficient mouse model of hypermuscularity (MSTN-/-). The study tests the null prediction that the larger MSTN-/- mice have larger brains compared with wild-type (WT) mice in order to service the larger muscles. Eleven post-mortem MSTN-/- mice and 12 WT mice were imaged at high resolution using contrast enhanced micro-CT. Masticatory muscle volumes (temporalis, masseter, internal and external pterygoids) and endocranial volumes were measured on the basis of two-dimensional manual tracings and the Cavalieri principle. Volumes were compared using Kruskal-Wallis and Student's t-tests. Results showed that the masticatory muscles of the MSTN-/- mice were significantly larger than in the WT mice. Increases were in the region of 17-36% depending on the muscle. Muscles increased in proportion to each other, maintaining percentages in the region of 5, 10, 21 and 62% of total muscle volume for the external ptyergoid, internal pterygoid, temporalis and masseter, respectively. Kruskal-Wallis and t-tests demonstrated that the endocranial volume was significantly larger in the WT mice, approximately 16% larger on average than that seen in the MSTN-/- mice. This comparative reduction of MSTN-/- endocranial size could not be explained in terms of observer bias, ageing, sexual dimorphism or body size scaling. That the results showed a reduction of brain size associated with an increase of muscle size falsifies the null prediction and lends tentative support to the view that the musculature influences brain growth. It remains to be determined whether the observed effect is primarily physical, nutritional, metabolic or molecular in nature.
Collapse
Affiliation(s)
- Nathan Jeffery
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, UK
| | - Christopher Mendias
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
74
|
Rodriguez J, Vernus B, Chelh I, Cassar-Malek I, Gabillard JC, Hadj Sassi A, Seiliez I, Picard B, Bonnieu A. Myostatin and the skeletal muscle atrophy and hypertrophy signaling pathways. Cell Mol Life Sci 2014; 71:4361-71. [PMID: 25080109 PMCID: PMC11113773 DOI: 10.1007/s00018-014-1689-x] [Citation(s) in RCA: 286] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 07/16/2014] [Accepted: 07/17/2014] [Indexed: 12/16/2022]
Abstract
Myostatin, a member of the transforming growth factor-β superfamily, is a potent negative regulator of skeletal muscle growth and is conserved in many species, from rodents to humans. Myostatin inactivation can induce skeletal muscle hypertrophy, while its overexpression or systemic administration causes muscle atrophy. As it represents a potential target for stimulating muscle growth and/or preventing muscle wasting, myostatin regulation and functions in the control of muscle mass have been extensively studied. A wealth of data strongly suggests that alterations in skeletal muscle mass are associated with dysregulation in myostatin expression. Moreover, myostatin plays a central role in integrating/mediating anabolic and catabolic responses. Myostatin negatively regulates the activity of the Akt pathway, which promotes protein synthesis, and increases the activity of the ubiquitin-proteasome system to induce atrophy. Several new studies have brought new information on how myostatin may affect both ribosomal biogenesis and translation efficiency of specific mRNA subclasses. In addition, although myostatin has been identified as a modulator of the major catabolic pathways, including the ubiquitin-proteasome and the autophagy-lysosome systems, the underlying mechanisms are only partially understood. The goal of this review is to highlight outstanding questions about myostatin-mediated regulation of the anabolic and catabolic signaling pathways in skeletal muscle. Particular emphasis has been placed on (1) the cross-regulation between myostatin, the growth-promoting pathways and the proteolytic systems; (2) how myostatin inhibition leads to muscle hypertrophy; and (3) the regulation of translation by myostatin.
Collapse
Affiliation(s)
- J. Rodriguez
- INRA, UMR866 Dynamique Musculaire Et Métabolisme, Université Montpellier 1, Université Montpellier 2, 2 Place Viala, 34060 Montpellier, France
| | - B. Vernus
- INRA, UMR866 Dynamique Musculaire Et Métabolisme, Université Montpellier 1, Université Montpellier 2, 2 Place Viala, 34060 Montpellier, France
| | - I. Chelh
- INRA, VetAgro Sup, UMR1213 Herbivores, 63122 Saint-Genès-Champanelle, France
| | - I. Cassar-Malek
- INRA, VetAgro Sup, UMR1213 Herbivores, 63122 Saint-Genès-Champanelle, France
| | - J. C. Gabillard
- INRA, UR1037, Fish Physiology and Genomics, Campus de Beaulieu, 35000 Rennes, France
| | - A. Hadj Sassi
- INRA-USC2009, Université Bordeaux 1, Avenue des Facultés, 33405 Talence, France
| | - I. Seiliez
- INRA, UR1067 Nutrition, Métabolisme, Aquaculture, 64310 Saint-Pée-sur-Nivelle, France
| | - B. Picard
- INRA, VetAgro Sup, UMR1213 Herbivores, 63122 Saint-Genès-Champanelle, France
| | - A. Bonnieu
- INRA, UMR866 Dynamique Musculaire Et Métabolisme, Université Montpellier 1, Université Montpellier 2, 2 Place Viala, 34060 Montpellier, France
| |
Collapse
|
75
|
Sakuma K, Aoi W, Yamaguchi A. The intriguing regulators of muscle mass in sarcopenia and muscular dystrophy. Front Aging Neurosci 2014; 6:230. [PMID: 25221510 PMCID: PMC4148637 DOI: 10.3389/fnagi.2014.00230] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 08/10/2014] [Indexed: 12/25/2022] Open
Abstract
Recent advances in our understanding of the biology of muscle have led to new interest in the pharmacological treatment of muscle wasting. Loss of muscle mass and increased intramuscular fibrosis occur in both sarcopenia and muscular dystrophy. Several regulators (mammalian target of rapamycin, serum response factor, atrogin-1, myostatin, etc.) seem to modulate protein synthesis and degradation or transcription of muscle-specific genes during both sarcopenia and muscular dystrophy. This review provides an overview of the adaptive changes in several regulators of muscle mass in both sarcopenia and muscular dystrophy.
Collapse
Affiliation(s)
- Kunihiro Sakuma
- Research Center for Physical Fitness, Sports and Health, Toyohashi University of Technology, Toyohashi, Japan
| | - Wataru Aoi
- Laboratory of Health Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Akihiko Yamaguchi
- Department of Physical Therapy, Health Sciences University of Hokkaido, Kanazawa, Japan
| |
Collapse
|
76
|
Wu Q, He J, Mei W, Zhang Z, Wu X, Sun F. Arene ruthenium(ii) complex, a potent inhibitor against proliferation, migration and invasion of breast cancer cells, reduces stress fibers, focal adhesions and invadopodia. Metallomics 2014; 6:2204-12. [PMID: 25142071 DOI: 10.1039/c4mt00158c] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Effective chemotherapy drugs for cancer that would inhibit tumor growth and suppress metastasis are currently lacking. In this study, a series of arene ruthenium complexes, [(η6-arene)Ru(H2iip)Cl]Cl (arene = p-cymene, RAWQ03; CH3C6H5, RAWQ04; and C6H6, RAWQ11), were synthesized and their inhibitory activity against tumor cells were evaluated. The results showed that the complex RAWQ11 inhibited the growth of MDA-MB-231 breast cancer cells by inducing S-phase arrest, which is closely related to the inhibition of cell mitosis-mediated cell nucleus damage. Further studies showed that RAWQ11 can inhibit the invasion and metastasis of MDA-MB-231 cells. The morphology of MDA-MB-231 cells changed, the number of focal adhesions decreased, and the stress fibers de-polymerized upon dealing with the complex RAWQ11. The FITC-gelatin assay confirmed that the formation of invadopodia in MDA-MB-231 cells was significantly blocked by RAWQ11. Furthermore, RAWQ11 can block the AKT signal pathway by upregulating the PTEN expression through binding and downregulating miR-21. These results demonstrated that this type of arene ruthenium(ii) complex can block the invadopodia formation by regulating the PTEN/AKT signal pathway mediated by miR-21 to inhibit the invasion and metastasis of breast cancer cells. Therefore, this complex can be used as a potential dual functional agent to inhibit the growth and metastasis of tumor cells.
Collapse
Affiliation(s)
- Qiong Wu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China.
| | | | | | | | | | | |
Collapse
|
77
|
Kunert-Keil C, Botzenhart U, Gedrange T, Gredes T. Interrelationship between bone substitution materials and skeletal muscle tissue. Ann Anat 2014; 199:73-8. [PMID: 25159858 DOI: 10.1016/j.aanat.2014.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 07/18/2014] [Accepted: 07/30/2014] [Indexed: 01/25/2023]
Abstract
Bone density and quantity are primary conditions for the insertion and stability of dental implants. In cases of a lack of adequate maxillary or mandibulary bone, bone augmentation will be necessary. The use of synthetic bioactive bone substitution materials is of increasing importance as alternatives to autogenously bone grafts. It is well known that bone can influence muscle function and muscle function can influence bone structures. Muscles have a considerable potential of adaptation and muscle tissue surrounding an inserted implant or bone surrogate can integrate changes in mechanical load of the muscle and hereupon induce signaling cascades with protein synthesis and arrangement of the cytoskeleton. The Musculus latissimus dorsi is very often used for the analyses of the in vivo biocompatibility of newly designed biomaterials. Beside macroscopically and histologically examination, biocompatibility can be assessed by analyses of the biomaterial influence of gene expression. This review discusses changes in the fiber type distribution, myosin heavy chain isoform composition, histological appearance and vascularization of the skeletal muscle after implantation of bone substitution materials. Especially, the effects of bone surrogates should be described at the molecular-biological and cellular level.
Collapse
Affiliation(s)
- Christiane Kunert-Keil
- Department of Orthodontics, Carl Gustav Carus Campus, Technische Universität Dresden, Fetscherstr. 74, Haus 28, D-01307 Dresden, Germany.
| | - Ute Botzenhart
- Department of Orthodontics, Carl Gustav Carus Campus, Technische Universität Dresden, Fetscherstr. 74, Haus 28, D-01307 Dresden, Germany
| | - Tomasz Gedrange
- Department of Orthodontics, Carl Gustav Carus Campus, Technische Universität Dresden, Fetscherstr. 74, Haus 28, D-01307 Dresden, Germany
| | - Tomasz Gredes
- Department of Orthodontics, Carl Gustav Carus Campus, Technische Universität Dresden, Fetscherstr. 74, Haus 28, D-01307 Dresden, Germany
| |
Collapse
|
78
|
Liu B, Chen X, Wang ZQ, Tong WM. Nbn gene inactivation in the CNS of mouse inhibits the myelinating ability of the mature cortical oligodendrocytes. Glia 2014; 62:133-44. [PMID: 24272708 DOI: 10.1002/glia.22593] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 10/07/2013] [Accepted: 10/17/2013] [Indexed: 01/12/2023]
Abstract
Nijmegen Breakage Syndrome (NBS) is a recessive genetic disorder characterized by immunodeficiency, elevated sensitivity to ionizing radiation, chromosomal instability, microcephaly, and high predisposition to malignancies. Since the underlying molecular mechanisms of the NBS microcephaly are still obscure, thus our group previously inactivated the Nbn gene in the central nervous system (CNS) of mice by nestin-Cre targeting gene system, and generated Nbn(CNS-del) mice. Interestingly, the newborn Nbn(CNS-del) mice exhibit obvious microcephaly, which is accompanied by severe ataxia and balance deficiency. In this study presented here, we report that Nbn-deficiency induces the enhanced apoptosis of the mature oligodendrocytes at postnatal day 7, which further affects the myelination of the nerve fibers of cerebrum and corpus callosum.The distinct regulatory roles of Ataxia telangiectasia mutated (ATM) signaling and protein kinase B(Akt)/the mammalian target of Rapamycin (AKT/mTOR) signaling are responsible for the enhanced apoptosis of the Nbn-deficient oligodendrocytes. In addition, a series of transcriptional factors including histonedeacetylase (HDAC), zinc finger protein 191 (ZFP-191) and myelin sheath regulatory factor (MRF) play distinct roles in regulating the myelination of the Nbn-deficient oligodendrocytes. Based on these results, it concludes that ATM-Chk2-P53-P21 signaling pathway and the AKT/mTOR signaling pathway are both responsible for the enhanced apoptosis of the Nbn-deficient oligodendrocytes. HDAC, ZFP-191, and MRF are also involved in the pathogenesis of the hypomyelination of the Nbn-deficient oligodendrocytes.
Collapse
Affiliation(s)
- Bo Liu
- Department of Pathology, Center for Experimental Animal Research, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | |
Collapse
|
79
|
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]
|
80
|
Current understanding of sarcopenia: possible candidates modulating muscle mass. Pflugers Arch 2014; 467:213-29. [PMID: 24797147 DOI: 10.1007/s00424-014-1527-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 04/21/2014] [Accepted: 04/22/2014] [Indexed: 12/17/2022]
Abstract
The world's elderly population is expanding rapidly, and we are now faced with the significant challenge of maintaining or improving physical activity, independence, and quality of life in the elderly. Sarcopenia, the age-related loss of skeletal muscle mass, is characterized by a deterioration of muscle quantity and quality leading to a gradual slowing of movement, a decline in strength and power, increased risk of fall-related injury, and often, frailty. Since sarcopenia is largely attributed to various molecular mediators affecting fiber size, mitochondrial homeostasis, and apoptosis, the mechanisms responsible for these deleterious changes present numerous therapeutic targets for drug discovery. Muscle loss has been linked with several proteolytic systems, including the ubuiquitin-proteasome, lysosome-autophagy, and tumor necrosis factor (TNF)-α/nuclear factor-kappaB (NF-κB) systems. Although many factors are considered to regulate age-dependent muscle loss, this gentle atrophy is not affected by factors known to enhance rapid atrophy (denervation, hindlimb suspension, etc.). In addition, defects in Akt-mammalian target of rapamycin (mTOR) and serum response factor (SRF)-dependent signaling have been found in sarcopenic muscle. Intriguingly, more recent studies indicated an apparent functional defect in autophagy- and myostatin-dependent signaling in sarcopenic muscle. In this review, we summarize the current understanding of the adaptation of many regulators in sarcopenia.
Collapse
|
81
|
Xiong CJ, Li PF, Song YL, Xue LX, Jia ZQ, Yao CX, Wei QX, Zhang SF, Zhang SF, Zhang YY, Zhao JM, Wang TQ, Guo MF, Zang MX. Insulin induces C2C12 cell proliferation and apoptosis through regulation of cyclin D1 and BAD expression. J Cell Biochem 2014; 114:2708-17. [PMID: 23794242 DOI: 10.1002/jcb.24619] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Accepted: 06/17/2013] [Indexed: 11/06/2022]
Abstract
Insulin is a secreted peptide hormone identified in human pancreas to promote glucose utilization. Insulin has been observed to induce cell proliferation and myogenesis in C2C12 cells. The precise mechanisms underlying the proliferation of C2C12 cells induced by insulin remain unclear. In this study, we observed for the first time that 10 nM insulin treatment promotes C2C12 cell proliferation. Additionally, 50 and 100 nM insulin treatment induces C2C12 cell apoptosis. By utilizing real-time PCR and Western blotting analysis, we found that the mRNA levels of cyclinD1 and BAD are induced upon 10 and 50 nM/100 nM insulin treatment, respectively. The similar results were observed in C2C12 cells expressing GATA-6 or PPARα. Our results identify for the first time the downstream targets of insulin, cyclin D1, and BAD, elucidate a new molecular mechanism of insulin in promoting cell proliferation and apoptosis.
Collapse
Affiliation(s)
- Cheng-Juan Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou City, Henan, 450001, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Watts R, Ghozlan M, Hughey CC, Johnsen VL, Shearer J, Hittel DS. Myostatin inhibits proliferation and insulin-stimulated glucose uptake in mouse liver cells. Biochem Cell Biol 2014; 92:226-34. [PMID: 24882465 DOI: 10.1139/bcb-2014-0004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although myostatin functions primarily as a negative regulator of skeletal muscle growth and development, accumulating biological and epidemiological evidence indicates an important contributing role in liver disease. In this study, we demonstrate that myostatin suppresses the proliferation of mouse Hepa-1c1c7 murine-derived liver cells (50%; p < 0.001) in part by reducing the expression of the cyclins and cyclin-dependent kinases that elicit G1-S phase transition of the cell cycle (p < 0.001). Furthermore, real-time PCR-based quantification of the long noncoding RNA metastasis associated lung adenocarcinoma transcript 1 (Malat1), recently identified as a myostatin-responsive transcript in skeletal muscle, revealed a significant downregulation (25% and 50%, respectively; p < 0.05) in the livers of myostatin-treated mice and liver cells. The importance of Malat1 in liver cell proliferation was confirmed via arrested liver cell proliferation (p < 0.05) in response to partial Malat1 siRNA-mediated knockdown. Myostatin also significantly blunted insulin-stimulated glucose uptake and Akt phosphorylation in liver cells while increasing the phosphorylation of myristoylated alanine-rich C-kinase substrate (MARCKS), a protein that is essential for cancer cell proliferation and insulin-stimulated glucose transport. Together, these findings reveal a plausible mechanism by which circulating myostatin contributes to the diminished regenerative capacity of the liver and diseases characterized by liver insulin resistance.
Collapse
Affiliation(s)
- Rani Watts
- a Faculty of Kinesiology, University of Calgary, 2500 University Dr. Calgary, AB T2N 1N4, Canada
| | | | | | | | | | | |
Collapse
|
83
|
Effects of Apelin-13 on Rat Bone Marrow-Derived Mesenchymal Stem Cell Proliferation Through the AKT/GSK3β/Cyclin D1 Pathway. Int J Pept Res Ther 2014. [DOI: 10.1007/s10989-014-9404-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
84
|
Cleasby ME, Jarmin S, Eilers W, Elashry M, Andersen DK, Dickson G, Foster K. Local overexpression of the myostatin propeptide increases glucose transporter expression and enhances skeletal muscle glucose disposal. Am J Physiol Endocrinol Metab 2014; 306:E814-23. [PMID: 24473441 PMCID: PMC3962614 DOI: 10.1152/ajpendo.00586.2013] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/28/2014] [Indexed: 12/20/2022]
Abstract
Insulin resistance (IR) in skeletal muscle is a prerequisite for type 2 diabetes and is often associated with obesity. IR also develops alongside muscle atrophy in older individuals in sarcopenic obesity. The molecular defects that underpin this syndrome are not well characterized, and there is no licensed treatment. Deletion of the transforming growth factor-β family member myostatin, or sequestration of the active peptide by overexpression of the myostatin propeptide/latency-associated peptide (ProMyo) results in both muscle hypertrophy and reduced obesity and IR. We aimed to establish whether local myostatin inhibition would have a paracrine/autocrine effect to enhance glucose disposal beyond that simply generated by increased muscle mass, and the mechanisms involved. We directly injected adeno-associated virus expressing ProMyo in right tibialis cranialis/extensor digitorum longus muscles of rats and saline in left muscles and compared the effects after 17 days. Both test muscles were increased in size (by 7 and 11%) and showed increased radiolabeled 2-deoxyglucose uptake (26 and 47%) and glycogen storage (28 and 41%) per unit mass during an intraperitoneal glucose tolerance test. This was likely mediated through increased membrane protein levels of GLUT1 (19% higher) and GLUT4 (63% higher). Interestingly, phosphorylation of phosphoinositol 3-kinase signaling intermediates and AMP-activated kinase was slightly decreased, possibly because of reduced expression of insulin-like growth factor-I in these muscles. Thus, myostatin inhibition has direct effects to enhance glucose disposal in muscle beyond that expected of hypertrophy alone, and this approach may offer potential for the therapy of IR syndromes.
Collapse
Affiliation(s)
- M. E. Cleasby
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, United Kingdom
| | - S. Jarmin
- School of Biological Sciences, Royal Holloway, University of London, Egham, Surrey, United Kingdom; and
| | - W. Eilers
- School of Biological Sciences, University of Reading, Reading, Berkshire, United Kingdom
| | - M. Elashry
- School of Biological Sciences, University of Reading, Reading, Berkshire, United Kingdom
| | - D. K. Andersen
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, United Kingdom
| | - G. Dickson
- School of Biological Sciences, Royal Holloway, University of London, Egham, Surrey, United Kingdom; and
| | - K. Foster
- School of Biological Sciences, University of Reading, Reading, Berkshire, United Kingdom
| |
Collapse
|
85
|
Ma G, Wang H, Gu X, Li W, Zhang X, Cui L, Li Y, Zhang Y, Zhao B, Li K. CARP, a myostatin-downregulated gene in CFM Cells, is a novel essential positive regulator of myogenesis. Int J Biol Sci 2014; 10:309-20. [PMID: 24644428 PMCID: PMC3957086 DOI: 10.7150/ijbs.7475] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 02/18/2014] [Indexed: 11/30/2022] Open
Abstract
Myostatin, a member of the TGF-β superfamily, has been shown to act as a negative regulator of myogenesis. Although its role in myogenesis has been clearly documented through genetic analysis, few gene cascades that respond to myostatin signaling and regulate myogenesis have been characterized, especially in avian species. In a previous study, we screened for such genes in chicken fetal myoblasts (CFMs) using the differential display PCR method and found that cardiac ankyrin repeat protein (CARP) was downregulated by myostatin and specifically expressed in chicken skeletal muscle. However, little is known about the potential functions of CARP in chicken skeletal myogenesis. In this study, the expression patterns of chicken CARP and the possible function of this gene in skeletal muscle growth were characterized. Our data showed that CARP was predominantly expressed in postnatal skeletal muscle, and its expression increased during myogenic differentiation in CFM cells. When CARP was overexpressed, CFM cell growth was enhanced by accelerating the cell cycle at the G1 to S phase transition and increasing cyclin D1 expression. CARP knockdown had the opposite effect: while myoblasts underwent differentiation, knockdown of CARP expression induced extensive cell death, suppressed the formation of myotubes, and markedly decreased the expression of differentiation-related genes such as myosin heavy chain (MHC), myoD, and caveolin-3. Our findings indicate that CARP may play a key role in the myostatin signaling cascade that governs chicken skeletal myogenesis through promoting proliferation and avoiding apoptosis during CFM cell differentiation.
Collapse
Affiliation(s)
- Guoda Ma
- 1. Institute of Neurology, Guangdong Medical College, Zhanjiang 524001, China; ; 2. Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, China
| | - Haiyang Wang
- 3. Department of General Surgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xuefeng Gu
- 1. Institute of Neurology, Guangdong Medical College, Zhanjiang 524001, China
| | - Wen Li
- 1. Institute of Neurology, Guangdong Medical College, Zhanjiang 524001, China
| | - Xingli Zhang
- 1. Institute of Neurology, Guangdong Medical College, Zhanjiang 524001, China
| | - Lili Cui
- 1. Institute of Neurology, Guangdong Medical College, Zhanjiang 524001, China
| | - You Li
- 1. Institute of Neurology, Guangdong Medical College, Zhanjiang 524001, China
| | - Yong Zhang
- 4. National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Bin Zhao
- 1. Institute of Neurology, Guangdong Medical College, Zhanjiang 524001, China; ; 2. Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, China
| | - Keshen Li
- 1. Institute of Neurology, Guangdong Medical College, Zhanjiang 524001, China
| |
Collapse
|
86
|
Adipose tissue-derived stem cell secreted IGF-1 protects myoblasts from the negative effect of myostatin. BIOMED RESEARCH INTERNATIONAL 2014; 2014:129048. [PMID: 24575400 PMCID: PMC3920898 DOI: 10.1155/2014/129048] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 12/03/2013] [Indexed: 11/23/2022]
Abstract
Myostatin, a TGF-β family member, is associated with inhibition of muscle growth and differentiation and might interact with the IGF-1 signaling pathway. Since IGF-1 is secreted at a bioactive level by adipose tissue-derived mesenchymal stem cells (ASCs), these cells (ASCs) provide a therapeutic option for Duchenne Muscular Dystrophy (DMD). But the protective effect of stem cell secreted IGF-1 on myoblast under high level of myostatin remains unclear. In the present study murine myoblasts were exposed to myostatin under presence of ASCs conditioned medium and investigated for proliferation and apoptosis. The protective effect of IGF-1 was further examined by using IGF-1 neutralizing and receptor antibodies as well as gene silencing RNAi technology. MyoD expression was detected to identify impact of IGF-1 on myoblasts differentiation when exposed to myostatin. IGF-1 was accountable for 43.6% of the antiapoptotic impact and 48.8% for the proliferative effect of ASCs conditioned medium. Furthermore, IGF-1 restored mRNA and protein MyoD expression of myoblasts under risk. Beside fusion and transdifferentiation the beneficial effect of ASCs is mediated by paracrine secreted cytokines, particularly IGF-1. The present study underlines the potential of ASCs as a therapeutic option for Duchenne muscular dystrophy and other dystrophic muscle diseases.
Collapse
|
87
|
White TA, LeBrasseur NK. Myostatin and sarcopenia: opportunities and challenges - a mini-review. Gerontology 2014; 60:289-93. [PMID: 24457615 DOI: 10.1159/000356740] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/14/2013] [Indexed: 12/27/2022] Open
Abstract
The progressive loss of skeletal muscle mass, strength and/or function with advancing age, termed sarcopenia, poses a major threat to independence and quality of life. Therefore, there is significant merit in better understanding the biology of sarcopenia and developing therapeutic interventions to prevent, slow or reverse its progression. Since the discovery of myostatin, a potent negative regulator of growth that is highly enriched in skeletal muscle, there has been great interest in it as a potential mediator of sarcopenia as well as a therapeutic target. The complex biology of myostatin, the promise of myostatin inhibition as an effective means to counter sarcopenia, and the challenges facing its clinical translation are reviewed herein.
Collapse
Affiliation(s)
- Thomas A White
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minn., USA
| | | |
Collapse
|
88
|
|
89
|
Gabillard JC, Biga PR, Rescan PY, Seiliez I. Revisiting the paradigm of myostatin in vertebrates: insights from fishes. Gen Comp Endocrinol 2013; 194:45-54. [PMID: 24018114 DOI: 10.1016/j.ygcen.2013.08.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 08/09/2013] [Accepted: 08/15/2013] [Indexed: 11/21/2022]
Abstract
In the last decade, myostatin (MSTN), a member of the TGFβ superfamily, has emerged as a strong inhibitor of muscle growth in mammals. In fish many studies reveal a strong conservation of mstn gene organization, sequence, and protein structures. Because of ancient genome duplication, teleostei may have retained two copies of mstn genes and even up to four copies in salmonids due to additional genome duplication event. In sharp contrast to mammals, the different fish mstn orthologs are widely expressed with a tissue-specific expression pattern. Quantification of mstn mRNA in fish under different physiological conditions, demonstrates that endogenous expression of mstn paralogs is rarely related to fish muscle growth rate. In addition, attempts to inhibit MSTN activity did not consistently enhance muscle growth as in mammals. In vitro, MSTN stimulates myotube atrophy and inhibits proliferation but not differentiation of myogenic cells as in mammals. In conclusion, given the strong mstn expression non-muscle tissues of fish, we propose a new hypothesis stating that fish MSTN functions as a general inhibitors of cell proliferation and cell growth to control tissue mass but is not specialized into a strong muscle regulator.
Collapse
Affiliation(s)
- Jean-Charles Gabillard
- INRA, UR1037 Laboratoire de Physiologie et Génomique des Poissons, Equipe Croissance et Qualité de la Chair des Poissons, Campus de Beaulieu, 35000 Rennes, France.
| | | | | | | |
Collapse
|
90
|
Liu B, Chen X, Wang ZQ, Tong WM. DNA damage and oxidative injury are associated with hypomyelination in the corpus callosum of newborn Nbn(CNS-del) mice. J Neurosci Res 2013; 92:254-66. [PMID: 24272991 DOI: 10.1002/jnr.23313] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 09/10/2013] [Accepted: 09/18/2013] [Indexed: 01/16/2023]
Abstract
Nijmegen breakage syndrome (NBS), caused by mutation of the Nbn gene, is a recessive genetic disorder characterized by immunodeficiency, elevated sensitivity to ionizing radiation, chromosomal instability, microcephaly, and high predisposition to malignancies. To explore the underlying molecular mechanisms of NBS microcephaly, Frappart et al. previously inactivated Nbn gene in the central nervous system (CNS) of mice by the nestin-Cre targeting gene system and generated Nbn(CNS-del) mice. Here we first report that Nbn gene inactivation induces the defective proliferation and enhanced apoptosis of the oligodendrocyte precursor cells (OPCs), contributing to the severe hypomyelination of the nerve fibers of the corpus callosum. Under conditions of DNA damage and oxidative stress, the distinct regulatory roles of ATM-Chk2 signaling and AKT/mTOR signaling are responsible for the defective proliferation and enhanced apoptosis of the Nbn-deficient OPCs. In addition, specific HDAC isoforms may play distinctive roles in regulating the myelination of the Nbn-deficient OPCs. However, brain-derived neurotrophic factor and nerve growth factor stimulation attenuates the oxidative stress and thereby increases the proliferation of the Nbn-deficient OPCs, which is accompanied by upregulation of the AKT/mTOR/P70S6K signaling pathway. Taken together, these findings demonstrate that DNA damage and oxidative stress resulting from Nbn gene inactivation are associated with hypomyelination of the nerve fibers of corpus callosum.
Collapse
Affiliation(s)
- B Liu
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | |
Collapse
|
91
|
Adams GR, Bamman MM. Characterization and regulation of mechanical loading-induced compensatory muscle hypertrophy. Compr Physiol 2013; 2:2829-70. [PMID: 23720267 DOI: 10.1002/cphy.c110066] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In mammalian systems, skeletal muscle exists in a dynamic state that monitors and regulates the physiological investment in muscle size to meet the current level of functional demand. This review attempts to consolidate current knowledge concerning development of the compensatory hypertrophy that occurs in response to a sustained increase in the mechanical loading of skeletal muscle. Topics covered include: defining and measuring compensatory hypertrophy, experimental models, loading stimulus parameters, acute responses to increased loading, hyperplasia, myofiber-type adaptations, the involvement of satellite cells, mRNA translational control, mechanotransduction, and endocrinology. The authors conclude with their impressions of current knowledge gaps in the field that are ripe for future study.
Collapse
Affiliation(s)
- Gregory R Adams
- Department of Physiology and Biophysics, University of California Irvine, Irvine, California, USA.
| | | |
Collapse
|
92
|
Numb-deficient satellite cells have regeneration and proliferation defects. Proc Natl Acad Sci U S A 2013; 110:18549-54. [PMID: 24170859 DOI: 10.1073/pnas.1311628110] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The adaptor protein Numb has been implicated in the switch between cell proliferation and differentiation made by satellite cells during muscle repair. Using two genetic approaches to ablate Numb, we determined that, in its absence, muscle regeneration in response to injury was impaired. Single myofiber cultures demonstrated a lack of satellite cell proliferation in the absence of Numb, and the proliferation defect was confirmed in satellite cell cultures. Quantitative RT-PCR from Numb-deficient satellite cells demonstrated highly up-regulated expression of p21 and Myostatin, both inhibitors of myoblast proliferation. Transfection with Myostatin-specific siRNA rescued the proliferation defect of Numb-deficient satellite cells. Furthermore, overexpression of Numb in satellite cells inhibited Myostatin expression. These data indicate a unique function for Numb during the initial activation and proliferation of satellite cells in response to muscle injury.
Collapse
|
93
|
Li WY, Song YL, Xiong CJ, Lu PQ, Xue LX, Yao CX, Wang WP, Zhang SF, Zhang SF, Wei QX, Zhang YY, Zhao JM, Zang MX. Insulin induces proliferation and cardiac differentiation of P19CL6 cells in a dose-dependent manner. Dev Growth Differ 2013; 55:676-86. [PMID: 24020834 DOI: 10.1111/dgd.12075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 07/27/2013] [Accepted: 08/04/2013] [Indexed: 12/29/2022]
Abstract
Insulin is a peptide hormone produced by beta cells of the pancreas. The roles of insulin in energy metabolism have been well studied, with most of the attention focused on glucose utilization, but the roles of insulin in cell proliferation and differentiation remain unclear. In this study, we observed for the first time that 10 nmol/L insulin treatment induces cell proliferation and cardiac differentiation of P19CL6 cells, whereas 50 and 100 nmol/L insulin treatment induces P19CL6 cell apoptosis and blocks cardiac differentiation of P19CL6 cells. By using real-time polymerase chain reaction (PCR) and Western blotting analysis, we found that the mRNA levels of cyclin D1 and α myosin heavy chain (α-MHC) are induced upon 10 nmol/L insulin stimulation and inhibited upon 50/100 nmol/L insulin treatment, whereas the mRNA levels of BCL-2-antagonist of cell death (BAD) exists a reverse trend. The similar results were observed in P19CL6 cells expressing GATA-6 or peroxisome proliferator-activated receptor α (PPARα). Our results identified the downstream targets of insulin, cyclin D1, BAD, α-MHC, and GATA-4, elucidate a novel molecular mechanism of insulin in promoting cell proliferation and differentiation.
Collapse
Affiliation(s)
- Wen-Yan Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
|
95
|
Valdés JA, Flores S, Fuentes EN, Osorio-Fuentealba C, Jaimovich E, Molina A. IGF-1 induces IP3-dependent calcium signal involved in the regulation of myostatin gene expression mediated by NFAT during myoblast differentiation. J Cell Physiol 2013; 228:1452-63. [DOI: 10.1002/jcp.24298] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 11/27/2012] [Indexed: 12/20/2022]
|
96
|
Watts R, Johnsen VL, Shearer J, Hittel DS. Myostatin-induced inhibition of the long noncoding RNA Malat1 is associated with decreased myogenesis. Am J Physiol Cell Physiol 2013; 304:C995-1001. [PMID: 23485710 DOI: 10.1152/ajpcell.00392.2012] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Myostatin, a member of the transforming growth factor-β (TGF-β) superfamily of secreted proteins, is a potent negative regulator of myogenesis. Free myostatin induces the phosphorylation of the Smad family of transcription factors, which, in turn, regulates gene expression, via the canonical TGF-β signaling pathway. There is, however, emerging evidence that myostatin can regulate gene expression independent of Smad signaling. As such, we acquired global gene expression data from the gastrocnemius muscle of C57BL/6 mice following a 6-day treatment with recombinant myostatin compared with vehicle-treated animals. Of the many differentially expressed genes, the myostatin-associated decrease (-11.20-fold; P < 0.05) in the noncoding metastasis-associated lung adenocarcinoma transcript 1 (Malat1) was the most significant and the most intriguing because of numerous reports describing its novel role in regulating cell growth. We therefore sought to further characterize the role of Malat1 expression in skeletal muscle myogenesis. RT-PCR-based quantification of C2C12 and primary human skeletal muscle cells revealed a significant and persistent upregulation (4- to 7-fold; P < 0.05) of Malat1 mRNA during the differentiation of myoblasts into myotubes. Conversely, targeted knockdown of Malat1 using siRNA suppressed myoblast proliferation by arresting cell growth in the G(0)/G(1) phase. These results reveal Malat1 as novel downstream target of myostatin with a considerable ability to regulate myogenesis. The identification of new targets of myostatin will have important repercussions for regenerative biology through inhibition and/or reversal of muscle atrophy and wasting diseases.
Collapse
Affiliation(s)
- Rani Watts
- Faculty of Kinesiology, University of Calgary, Calgary, Canada
| | | | | | | |
Collapse
|
97
|
Liu Y, Cheng H, Zhou Y, Zhu Y, Bian R, Chen Y, Li C, Ma Q, Zheng Q, Zhang Y, Jin H, Wang X, Chen Q, Zhu D. Myostatin induces mitochondrial metabolic alteration and typical apoptosis in cancer cells. Cell Death Dis 2013; 4:e494. [PMID: 23412387 PMCID: PMC3734823 DOI: 10.1038/cddis.2013.31] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Myostatin, a member of the transforming growth factor-β superfamily, regulates the glucose metabolism of muscle cells, while dysregulated myostatin activity is associated with a number of metabolic disorders, including muscle cachexia, obesity and type II diabetes. We observed that myostatin induced significant mitochondrial metabolic alterations and prolonged exposure of myostatin induced mitochondria-dependent apoptosis in cancer cells addicted to glycolysis. To address the underlying mechanism, we found that the protein levels of Hexokinase II (HKII) and voltage-dependent anion channel 1 (VDAC1), two key regulators of glucose metabolisms as well as metabolic stress-induced apoptosis, were negatively correlated. In particular, VDAC1 was dramatically upregulated in cells that are sensitive to myostatin treatment whereas HKII was downregulated and dissociated from mitochondria. Myostatin promoted the translocation of Bax from cytosol to mitochondria, and knockdown of VDAC1 inhibited myostatin-induced Bax translocation and apoptosis. These apoptotic changes can be partially rescued by repletion of ATP, or by ectopic expression of HKII, suggesting that perturbation of mitochondrial metabolism is causally linked with subsequent apoptosis. Our findings reveal novel function of myostatin in regulating mitochondrial metabolism and apoptosis in cancer cells.
Collapse
Affiliation(s)
- Y Liu
- The State Key Laboratory of Biomembrane and Membrane Biotechnology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
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
|
99
|
Lu J, Zhao H, Xu J, Zhang L, Yan L, Shen Z. Elevated cyclin D1 expression is governed by plasma IGF-1 through Ras/Raf/MEK/ERK pathway in rumen epithelium of goats supplying a high metabolizable energy diet. J Anim Physiol Anim Nutr (Berl) 2012; 97:1170-8. [DOI: 10.1111/jpn.12026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 11/21/2012] [Indexed: 12/15/2022]
Affiliation(s)
- J. Lu
- Lab of Animal Physiology and Biochemistry; College of Veterinary Medicine; Nanjing Agricultural University; Nanjing China
- Jiangsu Animal Husbandry & Veterinary College; Nanjing China
| | - H. Zhao
- Lab of Animal Physiology and Biochemistry; College of Veterinary Medicine; Nanjing Agricultural University; Nanjing China
| | - J. Xu
- Lab of Animal Physiology and Biochemistry; College of Veterinary Medicine; Nanjing Agricultural University; Nanjing China
| | - L. Zhang
- Lab of Animal Physiology and Biochemistry; College of Veterinary Medicine; Nanjing Agricultural University; Nanjing China
| | - L. Yan
- Lab of Animal Physiology and Biochemistry; College of Veterinary Medicine; Nanjing Agricultural University; Nanjing China
| | - Z. Shen
- Lab of Animal Physiology and Biochemistry; College of Veterinary Medicine; Nanjing Agricultural University; Nanjing China
| |
Collapse
|
100
|
Monestier O, Brun C, Heu K, Passet B, Malhouroux M, Magnol L, Vilotte JL, Blanquet V. Ubiquitous Gasp1 overexpression in mice leads mainly to a hypermuscular phenotype. BMC Genomics 2012; 13:541. [PMID: 23046573 PMCID: PMC3575399 DOI: 10.1186/1471-2164-13-541] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 10/03/2012] [Indexed: 11/10/2022] Open
Abstract
Background Myostatin, a member of the TGFβ superfamily, is well known as a potent and specific negative regulator of muscle growth. Targeting the myostatin signalling pathway may offer promising therapeutic strategies for the treatment of muscle-wasting disorders. In the last decade, various myostatin-binding proteins have been identified to be able to inhibit myostatin activity. One of these is GASP1 (Growth and Differentiation Factor-Associated Serum Protein-1), a protein containing a follistatin domain as well as multiple domains associated with protease inhibitors. Despite in vitro data, remarkably little is known about in vivo functions of Gasp1. To further address the role of GASP1 during mouse development and in adulthood, we generated a gain-of-function transgenic mouse model that overexpresses Gasp1 under transcriptional control of the human cytomegalovirus immediate-early promoter/enhancer. Results Overexpression of Gasp1 led to an increase in muscle mass observed not before day 15 of postnatal life. The surGasp1 transgenic mice did not display any other gross abnormality. Histological and morphometric analysis of surGasp1 rectus femoris muscles revealed an increase in myofiber size without a corresponding increase in myofiber number. Fiber-type distribution was unaltered. Interestingly, we do not detect a change in total fat mass and lean mass. These results differ from those for myostatin knockout mice, transgenic mice overexpressing the myostatin propeptide or follistatin which exhibit both muscle hypertrophy and hyperplasia, and show minimal fat deposition. Conclusions Altogether, our data give new insight into the in vivo functions of Gasp1. As an extracellular regulatory factor in the myostatin signalling pathway, additional studies on GASP1 and its homolog GASP2 are required to elucidate the crosstalk between the different intrinsic inhibitors of the myostatin.
Collapse
Affiliation(s)
- Olivier Monestier
- INRA, UMR1061 Unité de Génétique Moléculaire Animale, Limoges, 87060, France
| | | | | | | | | | | | | | | |
Collapse
|