201
|
Zhu Y, Deng D, Long C, Jin G, Zhang Q, Shen H. Abnormal expression of seven myogenesis-related genes in extraocular muscles of patients with concomitant strabismus. Mol Med Rep 2012; 7:217-22. [PMID: 23128899 DOI: 10.3892/mmr.2012.1149] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 10/03/2012] [Indexed: 11/06/2022] Open
Abstract
Hyperplasia or hypoplasia of muscles gradually leads to strabismus. Myogenesis-related genes are involved in extraocular muscle development, including myogenic differentiation 1 (MYOD1), myogenin (MYOG), retinoblastoma 1 (RB1), cyclin-dependent kinase inhibitor 1A (P21), cyclin‑dependent kinase inhibitor 1C (P57), insulin-like growth factor 1 (IGF1) and muscle creatine kinase (MCK). This study evaluated the expression of the above seven myogenesis-related genes by real-time quantitative RT-PCR in 18 resected extrocular muscles of patients with concomitant strabismus and 12 normal control muscle samples from one presumably healthy male 6 h after sudden mortality. We found that although there was a great divergence among the expression levels of 6 myogenesis-related regulatory factors, the relative expression patterns were similar in all the normal muscles, including the synergistic, antagonistic and yoke muscles. However, their expression levels in the 18 diseased extraocular muscles were abnormal; the expression levels of all the genes, with the exception of P57, were reduced in most of the diseased muscle tissues. These results imply that the abnormal expression of these myogenesis-related genes may contribute to concomitant strabismus.
Collapse
Affiliation(s)
- Yujuan Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, PR China
| | | | | | | | | | | |
Collapse
|
202
|
Nizzardo M, Simone C, Falcone M, Riboldi G, Comi GP, Bresolin N, Corti S. Direct reprogramming of adult somatic cells into other lineages: past evidence and future perspectives. Cell Transplant 2012; 22:921-44. [PMID: 23044010 DOI: 10.3727/096368912x657477] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Direct reprogramming of an adult cell into another differentiated lineage-such as fibroblasts into neurons, cardiomyocytes, or blood cells-without passage through an undifferentiated pluripotent stage is a new area of research that has recently emerged alongside stem cell technology and induced pluripotent stem cell reprogramming; indeed, this avenue of investigation has begun to play a central role in basic biological research and regenerative medicine. Even though the field seems new, its origins go back to the 1980s when it was demonstrated that differentiated adult cells can be converted into another cell lineage through the overexpression of transcription factors, establishing mature cell plasticity. Here, we retrace transdifferentiation experiments from the discovery of master control genes to recent in vivo reprogramming of one somatic cell into another from the perspective of possible applications for the development of new therapeutic approaches for human diseases.
Collapse
Affiliation(s)
- Monica Nizzardo
- Department of Neurological Sciences, Dino Ferrari Centre, University of Milan, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
203
|
Wobus AM, Rohwedel J, Maltsev V, Hescheler J. In vitro cellular models for cardiac development and pharmacotoxicology. Toxicol In Vitro 2012; 9:477-88. [PMID: 20650116 DOI: 10.1016/0887-2333(95)00023-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Permanent cultures of cardiac cells described so far have limited value for studying cell biology and pharmacology of the developing heart because of the loss of proliferative capacity and cardiac-specific properties of cardiomyocytes during long-term cultivation. Pluripotent embryonic carcinoma (EC) and embryonic stem (ES) cells cultivated as permanent lines offer a new approach for studying cardiogenic differentiation in vitro. We describe cardiogenesis in vitro by differentiating EC and ES cells by way of embryo-like aggregates (embryoid bodies) into spontaneously beating cardiomyocytes. During cardiomyocyte differentiation three distinct developmental stages were defined by expression of specific action potentials and ionic currents measured by the whole-cell patch-clamp technique. Whereas early differentiated cardiomyocytes are characterized by action potentials and ionic currents typical for early pacemaker cells, terminally differentiated cardiomyocytes show action potentials and ionic currents inherent to ventricular-, atrial- or sinus nodal-like cells. These functional characteristics are in accordance with the expression of alpha- and beta-cardiac myosin heavy chain at early differentiation stages and the additional expression of ventricular-specific MLC-2V and atrial-specific ANF genes at terminal stages demonstrated by reverse transcription polymerase chain reaction (RT-PCR) analysis. Pharmacological studies performed by measuring chronotropic responses and by analysing the Ca(2+) channel activity correspond to data obtained with cardiac cells from living organisms. For testing the influence of exogenous compounds on cardiac differentiation the teratogenic compound retinoic acid (RA) was applied during distinct stages of embryoid body development. A temporally controlled influence of RA on cardiac differentiation and expression of cardiac-specific genes was found. We conclude that ES cell-derived cardiomyocytes provide an excellent cellular model to study early cardiac development and to perform pharmacological and embryotoxicological investigations.
Collapse
Affiliation(s)
- A M Wobus
- Institute of Plant Genetics and Crop Plant Research, D-06466 Gatersleben, Germany
| | | | | | | |
Collapse
|
204
|
Abstract
In 1961, the satellite cell was first identified when electron microscopic examination of skeletal muscle demonstrated a cell wedged between the plasma membrane of the muscle fiber and the basement membrane. In recent years it has been conclusively demonstrated that the satellite cell is the primary cellular source for muscle regeneration and is equipped with the potential to self renew, thus functioning as a bona fide skeletal muscle stem cell (MuSC). As we move past the 50(th) anniversary of the satellite cell, we take this opportunity to discuss the current state of the art and dissect the unknowns in the MuSC field.
Collapse
|
205
|
Hsu CL, Muerdter CP, Knickerbocker AD, Walsh RM, Zepeda-Rivera MA, Depner KH, Sangesland M, Cisneros TB, Kim JY, Sanchez-Vazquez P, Cherezova L, Regan RD, Bahrami NM, Gray EA, Chan AY, Chen T, Rao MY, Hille MB. Cdc42 GTPase and Rac1 GTPase act downstream of p120 catenin and require GTP exchange during gastrulation of zebrafish mesoderm. Dev Dyn 2012; 241:1545-61. [PMID: 22911626 DOI: 10.1002/dvdy.23847] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We investigated the roles of p120 catenin, Cdc42, Rac1, and RhoA GTPases in regulating migration of presomitic mesoderm cells in zebrafish embryos. p120 catenin has dual roles: It binds the intracellular and juxtamembrane region of cadherins to stabilize cadherin-mediated adhesion with the aid of RhoA GTPase, and it activates Cdc42 GTPase and Rac1 GTPase in the cytosol to initiate cell motility. RESULTS During gastrulation of zebrafish embryos, knockdown of the synthesis of zygotic p120 catenin δ1 mRNAs with a splice-site morpholino caused lateral widening and anterior-posterior shortening of the presomitic mesoderm and somites and a shortened anterior-posterior axis. These phenotypes indicate a cell-migration effect. Co-injection of low amounts of wild-type Cdc42 or wild-type Rac1 or dominant-negative RhoA mRNAs, but not constitutively-active Cdc42 mRNA, rescued these p120 catenin δ1-depleted embryos. CONCLUSIONS These downstream small GTPases require appropriate spatiotemporal stimulation or cycling of GTP to guide mesodermal cell migration. A delicate balance of Rho GTPases and p120 catenin underlies normal development.
Collapse
Affiliation(s)
- Cynthia L Hsu
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Maguire RJ, Isaacs HV, Pownall ME. Early transcriptional targets of MyoD link myogenesis and somitogenesis. Dev Biol 2012; 371:256-68. [PMID: 22954963 DOI: 10.1016/j.ydbio.2012.08.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 07/10/2012] [Accepted: 08/22/2012] [Indexed: 12/20/2022]
Abstract
In order to identify early transcriptional targets of MyoD prior to skeletal muscle differentiation, we have undertaken a transcriptomic analysis on gastrula stage Xenopus embryos in which MyoD has been knocked-down. Our validated list of genes transcriptionally regulated by MyoD includes Esr1 and Esr2, which are known targets of Notch signalling, and Tbx6, mesogenin, and FoxC1; these genes are all are known to be essential for normal somitogenesis but are expressed surprisingly early in the mesoderm. In addition we found that MyoD is required for the expression of myf5 in the early mesoderm, in contrast to the reverse relationship of these two regulators in amniote somites. These data highlight a role for MyoD in the early mesoderm in regulating a set of genes that are essential for both myogenesis and somitogenesis.
Collapse
Affiliation(s)
- Richard J Maguire
- Biology Department, University of York, Heslington, York, North Yorkshire YO10 5YW, UK
| | | | | |
Collapse
|
207
|
Ijuin T, Takenawa T. Role of phosphatidylinositol 3,4,5-trisphosphate (PIP3) 5-phosphatase skeletal muscle- and kidney-enriched inositol polyphosphate phosphatase (SKIP) in myoblast differentiation. J Biol Chem 2012; 287:31330-41. [PMID: 22815484 DOI: 10.1074/jbc.m112.388785] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Insulin-like growth factors (IGFs) are essential for the development, regeneration, and hypertrophy of skeletal muscles. IGF-II promotes myoblast differentiation through phosphatidylinositol 3-kinase (PI 3-kinase), Akt, and mTOR signaling. Here, we report that skeletal muscle- and kidney-enriched inositol polyphosphate phosphatase (SKIP) negatively regulates myogenesis through inhibition of IGF-II production and attenuation of the IGF-II-Akt-mTOR signaling pathway. We also demonstrate that SKIP expression, which was markedly elevated during differentiation, was controlled by MyoD in C2C12 cells. Expression of SKIP inhibited IGF-II at the transcription level. These results indicate that SKIP regulates MyoD-mediated muscle differentiation. Silencing of SKIP increased IGF-II transcription and myoblast differentiation. Furthermore, knockdown of SKIP resulted in thick myotubes with a larger number of nuclei than that in control C2C12 cells. Taken together, these data indicate that SKIP controls the IGF-II-PI 3-kinase-Akt-mTOR auto-regulation loop during myogenesis. Our findings identify SKIP as a key regulator of muscle cell differentiation.
Collapse
Affiliation(s)
- Takeshi Ijuin
- Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | | |
Collapse
|
208
|
Singh MK, Singh KP, Kumar D, Shah RA, Anand T, Chauhan MS, Manik RS, Singla SK, Palta P. Buffalo (Bubalus bubalis) ES cell-like cells are capable of in vitro skeletal myogenic differentiation. Reprod Domest Anim 2012; 48:284-91. [PMID: 22788718 DOI: 10.1111/j.1439-0531.2012.02146.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
When buffalo embryonic stem (ES) cell-like cells that expressed surface markers SSEA-4, TRA-1-60, TRA-1-81, CD9 and CD90 and intracellular markers OCT4, SOX2 and FOXD3, as shown by immunofluorescence, and that expressed REX-1 and NUCLEOSTEMIN as confirmed by RT-PCR, were subjected to suspension culture in hanging drops in absence of LIF and buffalo foetal fibroblast feeder layer support, they differentiated to form three-dimensional embryoid bodies (EBs). Of 231 EBs examined on Day 3 of suspension culture, 141 (61.3 ± 3.09%) were of compact type, whereas 90 (38.4 ± 3.12%) were of cystic type. The cells obtained from EBs were found to express NF-68 and NESTIN (ectodermal lineage), BMP-4 and α-skeletal actin (mesodermal lineage), and α-fetoprotein, GATA-4 and HNF-4 (endodermal lineage). When these EBs were cultured on gelatin-coated dishes, they spontaneously differentiated to several cell types such as epithelial- and neuron-like cells. When EBs were cultured in the presence of 1 or 2% DMSO or 10(-8) M or 10(-7) M retinoic acid for 25 days, ES cells could be directed to form muscle cell-like cells, the identity of which was confirmed by expression of α-actinin by immunofluorescence and of MYF-5, MYOD and MYOGENIN genes by RT-PCR. MYOD was first detected on Day 10 in both treatment groups and on Day 15 in controls, whereas MYOGENIN was first detected on Day 10, Day 15 and Day 25 in the presence of retinoic acid, in the presence of DMSO and in controls, respectively. The present study demonstrates the ability of buffalo ES cell-like cells to undergo directed differentiation to cells of skeletal myogenic lineage.
Collapse
Affiliation(s)
- M K Singh
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Wang L, Jia Y, Rogers H, Wu YP, Huang S, Noguchi CT. GATA-binding protein 4 (GATA-4) and T-cell acute leukemia 1 (TAL1) regulate myogenic differentiation and erythropoietin response via cross-talk with Sirtuin1 (Sirt1). J Biol Chem 2012; 287:30157-69. [PMID: 22773876 DOI: 10.1074/jbc.m112.376640] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Erythropoietin (EPO), the cytokine required for erythrocyte production, contributes to muscle progenitor cell proliferation and delay myogenic differentiation. However, the underlying mechanism is not yet fully understood. Here, we report that EPO changes the skeletal myogenic regulatory factor expression program and delays differentiation via induction of GATA-4 and the basic helix-loop-helix TAL1 and that knockdown of both factors promotes differentiation. EPO increases the Sirt1 level, a NAD(+)-dependent deacetylase, and also induces the NAD(+)/NADH ratio that further increases Sirt1 activity. Sirt1 knockdown reduced GATA-4 and TAL1 expression, impaired EPO effect on delayed myogenic differentiation, and the Sirt1 knockdown effect was abrogated when combined with overexpression of GATA-4 or TAL1. GATA-4 interacts with Sirt1 and targets Sirt1 to the myogenin promoter and represses myogenin expression, whereas TAL1 inhibits myogenin expression by decreasing MyoD binding to and activation of the myogenin promoter. Sirt1 was found to bind to the GATA-4 promoter to directly regulate GATA-4 expression and GATA-4 binds to the TAL1 promoter to regulate TAL1 expression positively. These data suggest that GATA-4, TAL1, and Sirt1 cross-talk each other to regulate myogenic differentiation and mediate EPO activity during myogenic differentiation with Sirt1 playing a role upstream of GATA-4 and TAL1. Taken together, our findings reveal a novel role for GATA-4 and TAL1 to affect skeletal myogenic differentiation and EPO response via cross-talk with Sirt1.
Collapse
Affiliation(s)
- Li Wang
- Molecular Medicine Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892-1822, USA
| | | | | | | | | | | |
Collapse
|
210
|
Polyethylene glycol-based protein nanocapsules for functional delivery of a differentiation transcription factor. Biomaterials 2012; 33:5459-67. [DOI: 10.1016/j.biomaterials.2012.04.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 04/01/2012] [Indexed: 12/21/2022]
|
211
|
Tackling skeletal muscle cells epigenome in the next-generation sequencing era. Comp Funct Genomics 2012; 2012:979168. [PMID: 22701348 PMCID: PMC3371680 DOI: 10.1155/2012/979168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 04/03/2012] [Indexed: 11/21/2022] Open
Abstract
Recent advances in high-throughput technologies have transformed methodologies employed to study cell-specific epigenomes and the approaches to investigate complex cellular phenotypes. Application of next-generation sequencing technology in the skeletal muscle differentiation field is rapidly extending our knowledge on how chromatin modifications, transcription factors and chromatin regulators orchestrate gene expression pathways guiding myogenesis. Here, we review recent biological insights gained by the application of next-generation sequencing techniques to decode the epigenetic profile and gene regulatory networks underlying skeletal muscle differentiation.
Collapse
|
212
|
Acharya A, Baek ST, Huang G, Eskiocak B, Goetsch S, Sung CY, Banfi S, Sauer MF, Olsen GS, Duffield JS, Olson EN, Tallquist MD. The bHLH transcription factor Tcf21 is required for lineage-specific EMT of cardiac fibroblast progenitors. Development 2012; 139:2139-49. [PMID: 22573622 PMCID: PMC3357908 DOI: 10.1242/dev.079970] [Citation(s) in RCA: 363] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2012] [Indexed: 01/20/2023]
Abstract
The basic helix-loop-helix (bHLH) family of transcription factors orchestrates cell-fate specification, commitment and differentiation in multiple cell lineages during development. Here, we describe the role of a bHLH transcription factor, Tcf21 (epicardin/Pod1/capsulin), in specification of the cardiac fibroblast lineage. In the developing heart, the epicardium constitutes the primary source of progenitor cells that form two cell lineages: coronary vascular smooth muscle cells (cVSMCs) and cardiac fibroblasts. Currently, there is a debate regarding whether the specification of these lineages occurs early in the formation of the epicardium or later after the cells have entered the myocardium. Lineage tracing using a tamoxifen-inducible Cre expressed from the Tcf21 locus demonstrated that the majority of Tcf21-expressing epicardial cells are committed to the cardiac fibroblast lineage prior to initiation of epicardial epithelial-to-mesenchymal transition (EMT). Furthermore, Tcf21 null hearts fail to form cardiac fibroblasts, and lineage tracing of the null cells showed their inability to undergo EMT. This is the first report of a transcription factor essential for the development of cardiac fibroblasts. We demonstrate a unique role for Tcf21 in multipotent epicardial progenitors, prior to the process of EMT that is essential for cardiac fibroblast development.
Collapse
Affiliation(s)
- Asha Acharya
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas TX-75390, USA
| | - Seung Tae Baek
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas TX-75390, USA
| | - Guo Huang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas TX-75390, USA
| | - Banu Eskiocak
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas TX-75390, USA
| | - Sean Goetsch
- Department of Cardiology, University of Texas Southwestern Medical Center, Dallas TX-75390, USA
| | - Caroline Y. Sung
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas TX-75390, USA
| | - Serena Banfi
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas TX-75390, USA
| | - Marion F. Sauer
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas TX-75390, USA
| | - Gregory S. Olsen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas TX-75390, USA
| | | | - Eric N. Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas TX-75390, USA
| | - Michelle D. Tallquist
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas TX-75390, USA
| |
Collapse
|
213
|
DNA methyltransferase inhibitor CDA-II inhibits myogenic differentiation. Biochem Biophys Res Commun 2012; 422:522-6. [DOI: 10.1016/j.bbrc.2012.05.068] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 05/12/2012] [Indexed: 12/26/2022]
|
214
|
Wang D, Bai X, Tian Q, Lai Y, Lin EA, Shi Y, Mu X, Feng JQ, Carlson CS, Liu CJ. GEP constitutes a negative feedback loop with MyoD and acts as a novel mediator in controlling skeletal muscle differentiation. Cell Mol Life Sci 2012; 69:1855-73. [PMID: 22179841 PMCID: PMC3319484 DOI: 10.1007/s00018-011-0901-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Revised: 11/22/2011] [Accepted: 11/28/2011] [Indexed: 01/16/2023]
Abstract
Granulin-epithelin precursor (GEP) is an autocrine growth factor that has been implicated in embryonic development, tissue repair, tumorigenesis, and inflammation. Here we report that GEP was expressed in skeletal muscle tissue and its level was differentially altered in the course of C2C12 myoblast fusion. The GEP expression during myoblast fusion was a consequence of MyoD transcription factor binding to several E-box (CANNTG) sequences in the 5'-flanking regulatory region of GEP gene, followed by transcription. Recombinant GEP potently inhibited myotube formation from C2C12 myoblasts whereas the knockdown of endogenous of GEP via a siRNA approach accelerated the fusion of myoblasts to myotubes. Interestingly, the muscle fibers of GEP knockdown mice were larger in number but noticeably smaller in size when compared to the wild-type. Mechanistic studies revealed that during myoblast fusion, the addition of GEP led to remarkable reductions in the expressions of muscle-specific transcription factors, including MyoD. In addition, the regulation of myotube formation by GEP is mediated by the anti-myogenic factor JunB, which is upregulated following GEP stimulation. Thus, GEP growth factor, JunB, and MyoD transcription factor form a regulatory loop and act in concert in the course of myogenesis.
Collapse
Affiliation(s)
- Dawei Wang
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
- Department of Orthopedics, Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Xiaohui Bai
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
- Department of Otorhinolaryngology Head and Neck Surgery, Provincial Hospital affiliated to Shandong University, Jinan 250021, China
| | - Qingyun Tian
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Yongjie Lai
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Edward A. Lin
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Yongxiang Shi
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Xiaodong Mu
- Stem Cell Research Center, Children’s Hospital of Pittsburgh and Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219
| | - Jian Q. Feng
- Baylor College of Dentistry, Texas A&M Health Science Center, Dallas, TX 75246
| | - Cathy S. Carlson
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - Chuan-ju Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
215
|
Pavan WJ, Raible DW. Specification of neural crest into sensory neuron and melanocyte lineages. Dev Biol 2012; 366:55-63. [PMID: 22465373 PMCID: PMC3351495 DOI: 10.1016/j.ydbio.2012.02.038] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 02/29/2012] [Indexed: 11/27/2022]
Abstract
Elucidating the mechanisms by which multipotent cells differentiate into distinct lineages is a common theme underlying developmental biology investigations. Progress has been made in understanding some of the essential factors and pathways involved in the specification of different lineages from the neural crest. These include gene regulatory networks involving transcription factor hierarchies and input from signaling pathways mediated from environmental cues. In this review, we examine the mechanisms for two lineages that are derived from the neural crest, peripheral sensory neurons and melanocytes. Insights into the specification of these cell types may reveal common themes in the specification processes that occur throughout development.
Collapse
Affiliation(s)
- William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
216
|
Yusuf F, Brand-Saberi B. Myogenesis and muscle regeneration. Histochem Cell Biol 2012; 138:187-99. [DOI: 10.1007/s00418-012-0972-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2012] [Indexed: 12/27/2022]
|
217
|
Woo DH, Yun SJ, Kim EK, Ha JM, Shin HK, Bae SS. Regulation of Skeletal Muscle Differentiation by Akt. ACTA ACUST UNITED AC 2012. [DOI: 10.5352/jls.2012.22.4.447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
218
|
Macquarrie KL, Yao Z, Young JM, Cao Y, Tapscott SJ. miR-206 integrates multiple components of differentiation pathways to control the transition from growth to differentiation in rhabdomyosarcoma cells. Skelet Muscle 2012; 2:7. [PMID: 22541669 PMCID: PMC3417070 DOI: 10.1186/2044-5040-2-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 04/29/2012] [Indexed: 01/02/2023] Open
Abstract
Background Similar to replicating myoblasts, many rhabdomyosarcoma cells express the myogenic determination gene MyoD. In contrast to myoblasts, rhabdomyosarcoma cells do not make the transition from a regulative growth phase to terminal differentiation. Previously we demonstrated that the forced expression of MyoD with its E-protein dimerization partner was sufficient to induce differentiation and suppress multiple growth-promoting genes, suggesting that the dimer was targeting a switch that regulated the transition from growth to differentiation. Our data also suggested that a balance between various inhibitory transcription factors and MyoD activity kept rhabdomyosarcomas trapped in a proliferative state. Methods Potential myogenic co-factors were tested for their ability to drive differentiation in rhabdomyosarcoma cell culture models, and their relation to MyoD activity determined through molecular biological experiments. Results Modulation of the transcription factors RUNX1 and ZNF238 can induce differentiation in rhabdomyosarcoma cells and their activity is integrated, at least in part, through the activation of miR-206, which acts as a genetic switch to transition the cell from a proliferative growth phase to differentiation. The inhibitory transcription factor MSC also plays a role in controlling miR-206, appearing to function by occluding a binding site for MyoD in the miR-206 promoter. Conclusions These findings support a network model composed of coupled regulatory circuits with miR-206 functioning as a switch regulating the transition from one stable state (growth) to another (differentiation).
Collapse
Affiliation(s)
- Kyle L Macquarrie
- Human Biology Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, C3-168, Seattle, WA, 98109, USA.
| | | | | | | | | |
Collapse
|
219
|
Havis E, Coumailleau P, Bonnet A, Bismuth K, Bonnin MA, Johnson R, Fan CM, Relaix F, Shi DL, Duprez D. Sim2 prevents entry into the myogenic program by repressing MyoD transcription during limb embryonic myogenesis. Development 2012; 139:1910-20. [PMID: 22513369 DOI: 10.1242/dev.072561] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The basic helix-loop-helix transcription factor MyoD is a central actor that triggers the skeletal myogenic program. Cell-autonomous and non-cell-autonomous regulatory pathways must tightly control MyoD expression to ensure correct initiation of the muscle program at different places in the embryo and at different developmental times. In the present study, we have addressed the involvement of Sim2 (single-minded 2) in limb embryonic myogenesis. Sim2 is a bHLH-PAS transcription factor that inhibits transcription by active repression and displays enhanced expression in ventral limb muscle masses during chick and mouse embryonic myogenesis. We have demonstrated that Sim2 is expressed in muscle progenitors that have not entered the myogenic program, in different experimental conditions. MyoD expression is transiently upregulated in limb muscle masses of Sim2(-/-) mice. Conversely, Sim2 gain-of-function experiments in chick and Xenopus embryos showed that Sim2 represses MyoD expression. In addition, we show that Sim2 represses the activity of the mouse MyoD promoter in primary myoblasts and is recruited to the MyoD core enhancer in embryonic mouse limbs. Sim2 expression is non-autonomously and negatively regulated by the dorsalising factor Lmx1b. We propose that Sim2 represses MyoD transcription in limb muscle masses, through Sim2 recruitment to the MyoD core enhancer, in order to prevent premature entry into the myogenic program. This MyoD repression is predominant in ventral limb regions and is likely to contribute to the differential increase of the global mass of ventral muscles versus dorsal muscles.
Collapse
|
220
|
Abe H, Tominaga T, Matsubara T, Abe N, Kishi S, Nagai K, Murakami T, Araoka T, Doi T. Scleraxis modulates bone morphogenetic protein 4 (BMP4)-Smad1 protein-smooth muscle α-actin (SMA) signal transduction in diabetic nephropathy. J Biol Chem 2012; 287:20430-42. [PMID: 22474292 DOI: 10.1074/jbc.m111.275610] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Activation of mesangial cells (MCs), which is characterized by induction of smooth muscle α-actin (SMA) expression, contributes to a key event in various renal diseases; however, the mechanisms controlling MC differentiation are still largely undefined. Activated Smad1 induced SMA in a dose-dependent manner in MCs. As a direct regulating molecule for SMA, we identified and characterized scleraxis (Scx) as a new phenotype modulator in advanced glycation end product (AGE)-exposed MCs. Scx physically associated with E12 and bound the E-box in the promoter of SMA and negatively regulated the AGE-induced SMA expression. Scx induced expression and secretion of bone morphogenetic protein 4 (BMP4), thereby controlling the Smad1 activation in AGE-treated MCs. In diabetic mice, Scx was concomitantly expressed with SMA in the glomeruli. Inhibitor of differentiation 1 (Id1) was further induced by extended treatment with AGE, thereby dislodging Scx from the SMA promoter. These data suggest that Scx and Id1 are involved in the BMP4-Smad1-SMA signal transduction pathway besides the TGFβ1-Smad1-SMA signaling pathway and modulate phenotypic changes in MCs in diabetic nephropathy.
Collapse
Affiliation(s)
- Hideharu Abe
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School of Medicine, Tokushima 770-8503, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
221
|
Abstract
Classic experiments such as somatic cell nuclear transfer into oocytes and cell fusion demonstrated that differentiated cells are not irreversibly committed to their fate. More recent work has built on these conclusions and discovered defined factors that directly induce one specific cell type from another, which may be as distantly related as cells from different germ layers. This suggests the possibility that any specific cell type may be directly converted into any other if the appropriate reprogramming factors are known. Direct lineage conversion could provide important new sources of human cells for modeling disease processes or for cellular-replacement therapies. For future applications, it will be critical to carefully determine the fidelity of reprogramming and to develop methods for robustly and efficiently generating human cell types of interest.
Collapse
Affiliation(s)
- Thomas Vierbuchen
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | |
Collapse
|
222
|
Bentzinger CF, Wang YX, Rudnicki MA. Building muscle: molecular regulation of myogenesis. Cold Spring Harb Perspect Biol 2012; 4:4/2/a008342. [PMID: 22300977 DOI: 10.1101/cshperspect.a008342] [Citation(s) in RCA: 777] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The genesis of skeletal muscle during embryonic development and postnatal life serves as a paradigm for stem and progenitor cell maintenance, lineage specification, and terminal differentiation. An elaborate interplay of extrinsic and intrinsic regulatory mechanisms controls myogenesis at all stages of development. Many aspects of adult myogenesis resemble or reiterate embryonic morphogenetic episodes, and related signaling mechanisms control the genetic networks that determine cell fate during these processes. An integrative view of all aspects of myogenesis is imperative for a comprehensive understanding of muscle formation. This article provides a holistic overview of the different stages and modes of myogenesis with an emphasis on the underlying signals, molecular switches, and genetic networks.
Collapse
Affiliation(s)
- C Florian Bentzinger
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Health Research Institute, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
223
|
Liu HH, Wang JW, Li L, Han CC, Huang KL, Si JM, He H, Xu F. Molecular evolutionary analysis of the duck MYOD gene family and its differential expression pattern in breast muscle development. Br Poult Sci 2012; 52:423-31. [PMID: 21919569 DOI: 10.1080/00071668.2011.590795] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
1. The objective of the research was to investigate the molecular evolutionary relationships between the duck myogenic determination factors (MYOD) gene family members and their roles in muscle development. 2. The four members of the duck MYOD gene family were cloned using RT-PCR, and their relative mRNA expression during duck muscle development was measured using qRT-PCR. 3. The results showed that MyoD and Myf5 clustered together, as did MyoG and MRF4 based on their complete amino acid sequence and the basic helix-loop-helix domain. Results of the evolutionary level analysis were consistent with that of the differential expression patterns during duck breast muscle development. As determined by qRT-PCR, MyoD and Myf5 were highly expressed in 22-day embryos, while MyoG and MRF4 expression was high in 14-day embryos. 4. We conclude that the entire MYOD gene family in the duck originated from a common ancestral gene and evolved after two duplication events. The roles of the MYOD gene family members in duck muscle development are similar to those in mammals.
Collapse
Affiliation(s)
- He-He Liu
- Institute of Animal breeding & Genetic, Sichuan Agricultural University, Ya'an, Sichuan 625014, P R China
| | | | | | | | | | | | | | | |
Collapse
|
224
|
Abstract
The main route for CO(2) and water vapor exchange between a plant and the environment is through small pores called stomata. The accessibility of stomata and predictable division series that characterize their development provides an excellent system to address fundamental questions in biology. Stomatal cell-state transition and specification are regulated by a suite of transcription factors controlled by positional signaling via peptide ligands and transmembrane receptors. Downstream effectors include several members of the core cell-cycle genes. Environmentally induced signals are integrated into this essential developmental program to modulate stomatal development or function in response to changes in the abiotic environment. In addition, the recent identification of premitotic polarly localized proteins from both Arabidopsis and maize has laid a foundation for the future understanding of intrinsic cell polarity in plants. This review highlights the mechanisms of stomatal development through characterization of genes controlling cell-fate specification, cell polarity, cell division, and cell-cell communication during stomatal development and discusses the genetic framework linking these molecular processes with the correct spacing, density, and differentiation of stomata.
Collapse
Affiliation(s)
- Lynn Jo Pillitteri
- Department of Biology, Western Washington University, Bellingham, WA 98225, USA.
| | | |
Collapse
|
225
|
Takagaki Y, Yamagishi H, Matsuoka R. Factors Involved in Signal Transduction During Vertebrate Myogenesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 296:187-272. [DOI: 10.1016/b978-0-12-394307-1.00004-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
226
|
Mancinelli R, Pietrangelo T, Burnstock G, Fanò G, Fulle S. Transcriptional profile of GTP-mediated differentiation of C2C12 skeletal muscle cells. Purinergic Signal 2011; 8:207-21. [PMID: 22127439 DOI: 10.1007/s11302-011-9266-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 10/03/2011] [Indexed: 02/01/2023] Open
Abstract
Several purine receptors have been localised on skeletal muscle membranes. Previous data support the hypothesis that extracellular guanosine 5'-triphosphate (GTP) is an important regulatory factor in the development and function of muscle tissue. We have previously described specific extracellular binding sites for GTP on the plasma membrane of mouse skeletal muscle (C2C12) cells. Extracellular GTP induces an increase in intracellular Ca(2+) concentrations that results in membrane hyperpolarisation through Ca(2+)-activated K(+) channels, as has been demonstrated by patch-clamp experiments. This GTP-evoked increase in intracellular Ca(2+) is due to release of Ca(2+) from intracellular inositol-1,4,5-trisphosphate-sensitive stores. This enhances the expression of the myosin heavy chain in these C2C12 myoblasts and commits them to fuse into multinucleated myotubes, probably via a phosphoinositide-3-kinase-dependent signal-transduction mechanism. To define the signalling of extracellular GTP as an enhancer or modulator of myogenesis, we investigated whether the gene-expression profile of differentiated C2C12 cells (4 and 24 h in culture) is affected by extracellular GTP. To investigate the nuclear activity and target genes modulated by GTP, transcriptional profile analysis and real-time PCR were used. We demonstrate that in the early stages of differentiation, GTP up-regulates genes involved in different pathways associated with myogenic processes, including cytoskeleton structure, the respiratory chain, myogenesis, chromatin reorganisation, cell adhesion, and the Jak/Stat pathway, and down-regulates the mitogen-activated protein kinase pathway. GTP also increases the expression of three genes involved in myogenesis, Pp3ca, Gsk3b, and Pax7. Our data suggests that in the myogenic C2C12 cell line, extracellular GTP acts as a differentiative factor in the induction and sustaining of myogenesis.
Collapse
Affiliation(s)
- Rosa Mancinelli
- Department of Neuroscience and Imaging, University G. d'Annunzio Chieti-Pescara, Chieti, Italy.
| | | | | | | | | |
Collapse
|
227
|
Scimone ML, Srivastava M, Bell GW, Reddien PW. A regulatory program for excretory system regeneration in planarians. Development 2011; 138:4387-98. [PMID: 21937596 DOI: 10.1242/dev.068098] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Planarians can regenerate any missing body part, requiring mechanisms for the production of organ systems in the adult, including their prominent tubule-based filtration excretory system called protonephridia. Here, we identify a set of genes, Six1/2-2, POU2/3, hunchback, Eya and Sall, that encode transcription regulatory proteins that are required for planarian protonephridia regeneration. During regeneration, planarian stem cells are induced to form a cell population in regeneration blastemas expressing Six1/2-2, POU2/3, Eya, Sall and Osr that is required for excretory system formation. POU2/3 and Six1/2-2 are essential for these precursor cells to form. Eya, Six1/2-2, Sall, Osr and POU2/3-related genes are required for vertebrate kidney development. We determined that planarian and vertebrate excretory cells express homologous proteins involved in reabsorption and waste modification. Furthermore, we identified novel nephridia genes. Our results identify a transcriptional program and cellular mechanisms for the regeneration of an excretory organ and suggest that metazoan excretory systems are regulated by genetic programs that share a common evolutionary origin.
Collapse
Affiliation(s)
- M Lucila Scimone
- Howard Hughes Medical Institute, Whitehead Institute, and Department of Biology, Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA
| | | | | | | |
Collapse
|
228
|
Longo UG, Loppini M, Berton A, Spiezia F, Maffulli N, Denaro V. Tissue engineered strategies for skeletal muscle injury. Stem Cells Int 2011; 2012:175038. [PMID: 25098362 PMCID: PMC3216349 DOI: 10.1155/2012/175038] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 09/15/2011] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle injuries are common in athletes, occurring with direct and indirect mechanisms and marked residual effects, such as severe long-term pain and physical disability. Current therapy consists of conservative management including RICE protocol (rest, ice, compression and elevation), nonsteroidal anti-inflammatory drugs, and intramuscular corticosteroids. However, current management of muscle injuries often does not provide optimal restoration to preinjury status. New biological therapies, such as injection of platelet-rich plasma and stem-cell-based therapy, are appealing. Although some studies support PRP application in muscle-injury management, reasons for concern persist, and further research is required for a standardized and safe use of PRP in clinical practice. The role of stem cells needs to be confirmed, as studies are still limited and inconsistent. Further research is needed to identify mechanisms involved in muscle regeneration and in survival, proliferation, and differentiation of stem cells.
Collapse
Affiliation(s)
- Umile Giuseppe Longo
- Department of Orthopaedic and Trauma Surgery, Campus Bio-Medico University, Via Alvaro del Portillo 200, Trigoria, 00128 Rome, Italy
- Centro Integrato di Ricerca (CIR) Campus Bio-Medico University, Via Alvaro del Portillo 21, 00128, Rome, Italy
| | - Mattia Loppini
- Department of Orthopaedic and Trauma Surgery, Campus Bio-Medico University, Via Alvaro del Portillo 200, Trigoria, 00128 Rome, Italy
- Centro Integrato di Ricerca (CIR) Campus Bio-Medico University, Via Alvaro del Portillo 21, 00128, Rome, Italy
| | - Alessandra Berton
- Department of Orthopaedic and Trauma Surgery, Campus Bio-Medico University, Via Alvaro del Portillo 200, Trigoria, 00128 Rome, Italy
- Centro Integrato di Ricerca (CIR) Campus Bio-Medico University, Via Alvaro del Portillo 21, 00128, Rome, Italy
| | - Filippo Spiezia
- Department of Orthopaedic and Trauma Surgery, Campus Bio-Medico University, Via Alvaro del Portillo 200, Trigoria, 00128 Rome, Italy
- Centro Integrato di Ricerca (CIR) Campus Bio-Medico University, Via Alvaro del Portillo 21, 00128, Rome, Italy
| | - Nicola Maffulli
- Centre for Sports and Exercise Medicine, Barts and The London School of Medicine and Dentistry, Mile End Hospital, 275 Bancroft Road, London E1 4DG, UK
| | - Vincenzo Denaro
- Department of Orthopaedic and Trauma Surgery, Campus Bio-Medico University, Via Alvaro del Portillo 200, Trigoria, 00128 Rome, Italy
- Centro Integrato di Ricerca (CIR) Campus Bio-Medico University, Via Alvaro del Portillo 21, 00128, Rome, Italy
| |
Collapse
|
229
|
Xue M, Zan LS, Gao L, Wang HB. A novel polymorphism of the myogenin gene is associated with body measurement traits in native Chinese breeds. GENETICS AND MOLECULAR RESEARCH 2011; 10:2721-8. [PMID: 22095598 DOI: 10.4238/2011.november.4.6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Using PCR-SSCP and DNA sequencing technology, we examined the association of single nucleotide polymorphisms (SNPs) in the bovine MyoG gene with body measurement traits in 779 individuals of six native Chinese cattle breeds, namely Luxi, Luxi × Simmental crossbred, Nanyang, Xia'nan, Jiaxian red, and Qinchuan. A novel SNP, T314C, was detected. Allelic frequencies of MyoG-T/C in the six breeds were 0.8308/0.1692, 0.8774/0.1226, 0.8021/0.1979, 0.8209/0.1791, 0.8630/0.1370, 0.8044/0.1956, respectively. Least squares analysis revealed a significant (P < 0.05) association of the MyoG SNP with rump length in four breeds (Luxi, Xia'nan, Jiaxian red, and Qinchuan), with hucklebone width in three breeds (Luxi × Simmental crossbred, Nanyang and Xia'nan), with waist height in two breeds (Luxi × Simmental crossbred and Nanyang) and with body length in the Luxi breed. We conclude that the MyoG SNP has potential as a genetic marker for economically relevant body measurement traits in native Chinese cattle breeds.
Collapse
Affiliation(s)
- M Xue
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, PR China
| | | | | | | |
Collapse
|
230
|
Tao Y, Neppl RL, Huang ZP, Chen J, Tang RH, Cao R, Zhang Y, Jin SW, Wang DZ. The histone methyltransferase Set7/9 promotes myoblast differentiation and myofibril assembly. ACTA ACUST UNITED AC 2011; 194:551-65. [PMID: 21859860 PMCID: PMC3160588 DOI: 10.1083/jcb.201010090] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Set7 associates with the MyoD transcription factor to enhance expression of genes required for muscle differentiation. The molecular events that modulate chromatin structure during skeletal muscle differentiation are still poorly understood. We report in this paper that expression of the H3-K4 histone methyltransferase Set7 is increased when myoblasts differentiate into myotubes and is required for skeletal muscle development, expression of muscle contractile proteins, and myofibril assembly. Knockdown of Set7 or expression of a dominant-negative Set7 mutant impairs skeletal muscle differentiation, accompanied by a decrease in levels of histone monomethylation (H3-K4me1). Set7 directly interacts with MyoD to enhance expression of muscle differentiation genes. Expression of myocyte enhancer factor 2 and genes encoding contractile proteins is decreased in Set7 knockdown myocytes. Furthermore, we demonstrate that Set7 also activates muscle gene expression by precluding Suv39h1-mediated H3-K9 methylation on the promoters of myogenic differentiation genes. Together, our experiments define a biological function for Set7 in muscle differentiation and provide a molecular mechanism by which Set7 modulates myogenic transcription factors during muscle differentiation.
Collapse
Affiliation(s)
- Yazhong Tao
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
231
|
An CI, Dong Y, Hagiwara N. Genome-wide mapping of Sox6 binding sites in skeletal muscle reveals both direct and indirect regulation of muscle terminal differentiation by Sox6. BMC DEVELOPMENTAL BIOLOGY 2011; 11:59. [PMID: 21985497 PMCID: PMC3239296 DOI: 10.1186/1471-213x-11-59] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2011] [Accepted: 10/10/2011] [Indexed: 01/06/2023]
Abstract
Background Sox6 is a multi-faceted transcription factor involved in the terminal differentiation of many different cell types in vertebrates. It has been suggested that in mice as well as in zebrafish Sox6 plays a role in the terminal differentiation of skeletal muscle by suppressing transcription of slow fiber specific genes. In order to understand how Sox6 coordinately regulates the transcription of multiple fiber type specific genes during muscle development, we have performed ChIP-seq analyses to identify Sox6 target genes in mouse fetal myotubes and generated muscle-specific Sox6 knockout (KO) mice to determine the Sox6 null muscle phenotype in adult mice. Results We have identified 1,066 Sox6 binding sites using mouse fetal myotubes. The Sox6 binding sites were found to be associated with slow fiber-specific, cardiac, and embryonic isoform genes that are expressed in the sarcomere as well as transcription factor genes known to play roles in muscle development. The concurrently performed RNA polymerase II (Pol II) ChIP-seq analysis revealed that 84% of the Sox6 peak-associated genes exhibited little to no binding of Pol II, suggesting that the majority of the Sox6 target genes are transcriptionally inactive. These results indicate that Sox6 directly regulates terminal differentiation of muscle by affecting the expression of sarcomere protein genes as well as indirectly through influencing the expression of transcription factors relevant to muscle development. Gene expression profiling of Sox6 KO skeletal and cardiac muscle revealed a significant increase in the expression of the genes associated with Sox6 binding. In the absence of the Sox6 gene, there was dramatic upregulation of slow fiber-specific, cardiac, and embryonic isoform gene expression in Sox6 KO skeletal muscle and fetal isoform gene expression in Sox6 KO cardiac muscle, thus confirming the role Sox6 plays as a transcriptional suppressor in muscle development. Conclusions Our present data indicate that during development, Sox6 functions as a transcriptional suppressor of fiber type-specific and developmental isoform genes to promote functional specification of muscle which is critical for optimum muscle performance and health.
Collapse
Affiliation(s)
- Chung-Il An
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, One Shields Avenue, Davis, California 95616, USA
| | | | | |
Collapse
|
232
|
Seok HY, Tatsuguchi M, Callis TE, He A, Pu WT, Wang DZ. miR-155 inhibits expression of the MEF2A protein to repress skeletal muscle differentiation. J Biol Chem 2011; 286:35339-35346. [PMID: 21868385 DOI: 10.1074/jbc.m111.273276] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
microRNAs (miRNAs) are 21-23-nucleotide non-coding RNAs. It has become more and more evident that this class of small RNAs plays critical roles in the regulation of gene expression at the post-transcriptional level. MEF2A is a member of the MEF2 (myogenic enhancer factor 2) family of transcription factors. Prior report showed that the 3'-untranslated region (3'-UTR) of the Mef2A gene mediated its repression; however, the molecular mechanism underlying this intriguing observation was unknown. Here, we report that MEF2A is repressed by miRNAs. We identify miR-155 as one of the primary miRNAs that significantly represses the expression of MEF2A. We show that knockdown of the Mef2A gene by siRNA impairs myoblast differentiation. Similarly, overexpression of miR-155 leads to the repression of endogenous MEF2A expression and the inhibition of myoblast differentiation. Most importantly, reintroduction of MEF2A in miR-155 overexpressed myoblasts was able to partially rescue the miR-155-induced myoblast differentiation defect. Our data therefore establish miR-155 as an important regulator of MEF2A expression and uncover its function in muscle gene expression and myogenic differentiation.
Collapse
Affiliation(s)
- Hee Young Seok
- Department of Cardiology, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Mariko Tatsuguchi
- Department of Cardiology, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115
| | - Thomas E Callis
- Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Aibin He
- Department of Cardiology, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115
| | - William T Pu
- Department of Cardiology, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115
| | - Da-Zhi Wang
- Department of Cardiology, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, North Carolina 27599.
| |
Collapse
|
233
|
Acharya A, Baek ST, Banfi S, Eskiocak B, Tallquist MD. Efficient inducible Cre-mediated recombination in Tcf21 cell lineages in the heart and kidney. Genesis 2011; 49:870-7. [PMID: 21432986 DOI: 10.1002/dvg.20750] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 12/17/2010] [Accepted: 03/12/2011] [Indexed: 11/10/2022]
Abstract
Tcf21 is a Class II bHLH family member with essential roles in the formation of the lungs, kidneys, gonads, spleen, and heart. Here, we report the utility of a mouse line with targeted insertion of a tamoxifen-inducible Cre recombinase, MerCreMer at the Tcf21 locus. This mouse line will permit the inducible expression of Cre recombinase in Tcf21-expressing cells. Using ROSA26 reporter mice, we show that Cre recombinase is specifically and robustly activated in multiple Tcf21-expressing tissues during embryonic and postnatal development. The expression profile in the kidney is particularly dynamic with the ability to cause recombination in mesangial cells at one time of induction and podocytes at another time. These features make the Tcf21-driven inducible Cre line (Tcf21(iCre) ) a valuable genetic tool for spatiotemporal gene function analysis and lineage tracing of cells in the heart, kidney, cranial muscle, and gonads.
Collapse
Affiliation(s)
- Asha Acharya
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9148, USA
| | | | | | | | | |
Collapse
|
234
|
Scharner J, Zammit PS. The muscle satellite cell at 50: the formative years. Skelet Muscle 2011; 1:28. [PMID: 21849021 PMCID: PMC3177780 DOI: 10.1186/2044-5040-1-28] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Accepted: 08/17/2011] [Indexed: 12/25/2022] Open
Abstract
In February 1961, Alexander Mauro described a cell 'wedged' between the plasma membrane of the muscle fibre and the surrounding basement membrane. He postulated that it could be a dormant myoblast, poised to repair muscle when needed. In the same month, Bernard Katz also reported a cell in a similar location on muscle spindles, suggesting that it was associated with development and growth of intrafusal muscle fibres. Both Mauro and Katz used the term 'satellite cell' in relation to their discoveries. Today, the muscle satellite cell is widely accepted as the resident stem cell of skeletal muscle, supplying myoblasts for growth, homeostasis and repair. Since 2011 marks both the 50th anniversary of the discovery of the satellite cell, and the launch of Skeletal Muscle, it seems an opportune moment to summarise the seminal events in the history of research into muscle regeneration. We start with the 19th-century pioneers who showed that muscle had a regenerative capacity, through to the descriptions from the mid-20th century of the underlying cellular mechanisms. The journey of the satellite cell from electron microscope curio, to its gradual acceptance as a bona fide myoblast precursor, is then charted: work that provided the foundations for our understanding of the role of the satellite cell. Finally, the rapid progress in the age of molecular biology is briefly discussed, and some ongoing debates on satellite cell function highlighted.
Collapse
Affiliation(s)
- Juergen Scharner
- Randall Division of Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK.
| | | |
Collapse
|
235
|
Watanabe S, Hirai H, Asakura Y, Tastad C, Verma M, Keller C, Dutton JR, Asakura A. MyoD gene suppression by Oct4 is required for reprogramming in myoblasts to produce induced pluripotent stem cells. Stem Cells 2011; 29:505-16. [PMID: 21425413 DOI: 10.1002/stem.598] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Expression of the four transcription factors, that is, Oct4, Sox2, cMyc, and Klf4 has been shown to generate induced pluripotent stem cells (iPSCs) from many types of specialized differentiated somatic cells. It remains unclear, however, whether fully committed skeletal muscle progenitor cells (myoblasts) have the potency to undergo reprogramming to develop iPSCs in line with previously reported cases. To test this, we have isolated genetically marked myoblasts derived from satellite cell of adult mouse muscles using the Cre-loxP system (Pax7-CreER:R26R and Myf5-Cre:R26R). On infection with retroviral vectors expressing the four factors, these myoblasts gave rise to myogenic lineage tracer lacZ-positive embryonic stem cell (ESC)-like colonies. These cells expressed ESC-specific genes and were competent to differentiate into all three germ layers and germ cells, indicating the successful generation of myoblast-derived iPSCs. Continuous expression of the MyoD gene, a master transcription factor for skeletal muscle specification, inhibited this reprogramming process in myoblasts. In contrast, reprogramming myoblasts isolated from mice lacking the MyoD gene led to an increase in reprogramming efficiency. Our data also indicated that Oct4 acts as a transcriptional suppressor of MyoD gene expression through its interaction with the upstream enhancer region. Taken together, these results indicate that suppression of MyoD gene expression by Oct4 is required for the initial reprogramming step in the development of iPSCs from myoblasts. This data suggests that the skeletal muscle system provides a well-defined differentiation model to further elaborate on the effects of iPSC reprogramming in somatic cells.
Collapse
Affiliation(s)
- Shuichi Watanabe
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
236
|
Fan H, Cinar MU, Phatsara C, Tesfaye D, Tholen E, Looft C, Schellander K. Molecular mechanism underlying the differential MYF6 expression in postnatal skeletal muscle of Duroc and Pietrain breeds. Gene 2011; 486:8-14. [PMID: 21749918 DOI: 10.1016/j.gene.2011.06.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 06/20/2011] [Accepted: 06/24/2011] [Indexed: 10/18/2022]
Abstract
Among modern western pigs, Duroc (high meat fat ratio) and Pietrain (low meat fat ratio) breeds extensively utilized in commercial pork production differ extremely for their muscle phenotypes. The molecular mechanism, especially the epigenetic mechanism, underlying these breed-specific differences is poorly known. Myogenic factor 6 (MYF6) is the most abundantly expressed myogenic factor in adult muscle. Moreover, MYF6 tends to be expressed more highly in muscle tissue of the lean selection line and is supposed to be one promising candidate gene for growth- and meat quality-related traits in adult pigs. Six months old female Duroc and Pietrain pure breed pigs were used in this study. Protein and mRNA levels of MYF6 in loin eye muscle were determined by Western blotting and quantitative Real-time reverse transcription PCR (qRT-PCR), respectively. The DNA methylation status of the MYF6 5'-regulatory region was determined by bisulfite sequencing PCR (BSP). The global Histone 4 acetylation at lysines 5 (H4K5) and 8 (H4K8) were examined by Western blotting. Pietrain pigs exhibited significant higher expression of MYF6 and hypermethylated E2F1 binding element within MYF6 5'-regulatory region as compared with Duroc pigs. Significant elevation in DNA methyltransferase 1 (DNMT1) expression was observed in Pietrain pigs which are in agreement with hypermethylation of MYF6. Histone acetylation level at neither H4K5 nor H4K8 is significant between two breed pigs. Nevertheless, mRNA and protein expression of E2F1 were significantly elevated in the Pietrain breed. It is thus conceivable that the upregulation of MYF6 transcription in postnatal Pietrain pigs is not associated with cis-activation by epigenetic modification of MYF6 5'-regulatory region, but may be attributed to trans-activation through enriched expression of E2F1.
Collapse
Affiliation(s)
- Huitao Fan
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, Germany.
| | | | | | | | | | | | | |
Collapse
|
237
|
Lee NKL, Skinner JPJ, Zajac JD, MacLean HE. Ornithine decarboxylase is upregulated by the androgen receptor in skeletal muscle and regulates myoblast proliferation. Am J Physiol Endocrinol Metab 2011; 301:E172-9. [PMID: 21505150 DOI: 10.1152/ajpendo.00094.2011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of this study is to determine if the Odc1 gene, which encodes ornithine decarboxylase (ODC), the rate-limiting enzyme in polyamine biosynthesis, is directly regulated by the androgen receptor (AR) in skeletal muscle myoblasts and if Odc1 regulates myoblast proliferation and differentiation. We previously showed that expression of Odc1 is decreased in muscle from AR knockout male mice. In this study, we show in vivo that Odc1 expression is also decreased >60% in muscle from male muscle-specific AR knockout mice. In normal muscle homeostasis, Odc1 expression is regulated by age and sex, reflecting testosterone levels, as muscle of adult male mice expresses high levels of Odc1 compared with age-matched females and younger males. In vitro, expression of Odc1 is 10- and 1.5-fold higher in proliferating mouse C(2)C(12) and human skeletal muscle myoblasts, respectively, than in differentiated myotubes. Dihydrotestosterone increases Odc1 levels 2.7- and 1.6-fold in skeletal muscle cell myoblasts after 12 and 24 h of treatment, respectively. Inhibition of ODC activity in C(2)C(12) myoblasts by α-difluoromethylornithine decreases myoblast number by 40% and 66% following 48 and 72 h of treatment, respectively. In contrast, overexpression of Odc1 in C(2)C(12) myoblasts results in a 27% increase in cell number vs. control when cells are grown under differentiation conditions for 96 h. This prolonged proliferation is associated with delayed differentiation, with reduced expression of the differentiation markers myogenin and Myf6 in Odc1-overexpressing cells. In conclusion, androgens act via the AR to upregulate Odc1 in skeletal muscle myoblasts, and Odc1 promotes myoblast proliferation and delays differentiation.
Collapse
MESH Headings
- Androgens/pharmacology
- Animals
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Embryo, Mammalian
- Female
- Gene Expression Regulation, Enzymologic/drug effects
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiology
- Myoblasts, Skeletal/drug effects
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/physiology
- Ornithine Decarboxylase/genetics
- Ornithine Decarboxylase/metabolism
- Pregnancy
- Receptors, Androgen/metabolism
- Receptors, Androgen/physiology
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Nicole K L Lee
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | | | | | | |
Collapse
|
238
|
Xiang X, Zhao J, Xu G, Li Y, Zhang W. mTOR and the differentiation of mesenchymal stem cells. Acta Biochim Biophys Sin (Shanghai) 2011; 43:501-10. [PMID: 21642276 DOI: 10.1093/abbs/gmr041] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The mammalian target of rapamycin (mTOR), an evolutionarily conserved serine-threonine protein kinase, belongs to the phosphoinositide 3-kinase (PI3K)-related kinase family, which contains a lipid kinase-like domain within their C-terminal region. Recent studies have revealed that mTOR as a critical intracellular molecule can sense the extracellular energy status and regulate the cell growth and proliferation in a variety of cells and tissues. This review summarizes our current understanding about the effects of mTOR on cell differentiation and tissue development, with an emphasis on the lineage determination of mesenchymal stem cells. mTOR can promote adipogenesis in white adipocytes, brown adipocytes, and muscle satellite cells, while rapamycin inhibits the adipogenic function of mTOR. mTOR signaling may function to affect osteoblast proliferation and differentiation, however, rapamycin has been reported to either inhibit or promote osteogenesis. Although the precise mechanism remains unclear, mTOR is indispensable for myogenesis. Depending on the cell type, rapamycin has been reported to inhibit, promote, or have no effect on myogenesis.
Collapse
Affiliation(s)
- Xinxin Xiang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Department of Physiology and Pathophysiology, Health Science Center, Peking University, Beijing, China
| | | | | | | | | |
Collapse
|
239
|
Sambasivan R, Kuratani S, Tajbakhsh S. An eye on the head: the development and evolution of craniofacial muscles. Development 2011; 138:2401-15. [DOI: 10.1242/dev.040972] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Skeletal muscles exert diverse functions, enabling both crushing with great force and movement with exquisite precision. A remarkably distinct repertoire of genes and ontological features characterise this tissue, and recent evidence has shown that skeletal muscles of the head, the craniofacial muscles, are evolutionarily, morphologically and molecularly distinct from those of the trunk. Here, we review the molecular basis of craniofacial muscle development and discuss how this process is different to trunk and limb muscle development. Through evolutionary comparisons of primitive chordates (such as amphioxus) and jawless vertebrates (such as lampreys) with jawed vertebrates, we also provide some clues as to how this dichotomy arose.
Collapse
Affiliation(s)
- Ramkumar Sambasivan
- Institut Pasteur, Stem Cells and Development, Paris, F-75015, France
- CNRS URA 2578, 25 rue du Dr Roux, Paris, F-75015, France
| | - Shigeru Kuratani
- Laboratory for Evolutionary Morphology, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Shahragim Tajbakhsh
- Institut Pasteur, Stem Cells and Development, Paris, F-75015, France
- CNRS URA 2578, 25 rue du Dr Roux, Paris, F-75015, France
| |
Collapse
|
240
|
Klumpp D, Horch RE, Kneser U, Beier JP. Engineering skeletal muscle tissue--new perspectives in vitro and in vivo. J Cell Mol Med 2011; 14:2622-9. [PMID: 21091904 PMCID: PMC4373482 DOI: 10.1111/j.1582-4934.2010.01183.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Muscle tissue engineering (TE) has not yet been clinically applied because of several problems. However, the field of skeletal muscle TE has been developing tremendously and new approaches and techniques have emerged. This review will highlight recent developments in the field of nanotechnology, especially electrospun nanofibre matrices, as well as potential cell sources for muscle TE. Important developments in cardiac muscle TE and clinical studies on Duchenne muscular dystrophy (DMD) will be included to show their implications on skeletal muscle TE.
Collapse
Affiliation(s)
- Dorothee Klumpp
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | |
Collapse
|
241
|
Abstract
There is worldwide enthusiasm for the prospect of some kind of cellular transplant therapy for repair of failing organs. The olfactory mucosa of a patient's nose is easily biopsied to provide a ready source of multipotent cells. In this article we address practical issues pertinent to using olfactory neural stem cells for tissue repair. These cells are emerging as potentially most significant candidates for human tissue repair strategies. Previously we have shown that stem cells from olfactory mucosa are multipotent. As well, we have recently published three potential clinical applications. Their expression of dopaminergic markers in vitro and in a Parkinson's rat transplant model has been demonstrated. Their conversion to chondrogenic phenotype in vitro and in vivo has also been described, as has their transplant into a rat model of cardiac infarction. Here we examine in detail the biology of the olfactory neural stem cell using the rat as our animal model cell source. We establish its presence by examining self-renewal capacity and for phenotypic acquisition in inductive circumstances. We determine its frequency within the cell population and show that our culture system selects for this putative stem cell. Our studies demonstrate that adult olfactory stem cells, when transplanted into an environmental niche different from that of their origin, are able to demonstrate multipotency by acquiring the phenotype of the resident cells. We investigate how immediate the instruction need be. We test the hypothesis that olfactory neurospheres contain stem cells whose capacity for differentiation is triggered by signals of the immediate environmental niche. Significantly, of importance to any tissue regeneration endeavor, stem cell numbers were shown to be enriched by our culture methods. This was confirmed whether measured by sphere-forming capacity or differentiation response rate.
Collapse
Affiliation(s)
- Andrew Wetzig
- Eskitis Institute for Cell and Molecular Therapies, National Centre for Adult Stem Cell Research, Griffith University, Nathan, Queensland, Australia
| | | | | |
Collapse
|
242
|
Lee C, Ma H, Hang JQ, Leveque V, Sklan EH, Elazar M, Klumpp K, Glenn JS. The hepatitis C virus NS5A inhibitor (BMS-790052) alters the subcellular localization of the NS5A non-structural viral protein. Virology 2011; 414:10-8. [PMID: 21513964 DOI: 10.1016/j.virol.2011.03.026] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Revised: 01/25/2011] [Accepted: 03/28/2011] [Indexed: 12/31/2022]
Abstract
The hepatitis C virus (HCV) non-structural (NS) 5A protein plays an essential role in the replication of the viral RNA by the membrane-associated replication complex (RC). Recently, a putative NS5A inhibitor, BMS-790052, exhibited the highest potency of any known anti-HCV compound in inhibiting HCV replication in vitro and showed a promising clinical effect in HCV-infected patients. The precise mechanism of action for this new class of potential anti-HCV therapeutics, however, is still unclear. In order to gain further insight into its mode of action, we sought to test the hypothesis that the antiviral effect of BMS-790052 might be mediated by interfering with the functional assembly of the HCV RC. We observed that BMS-790052 indeed altered the subcellular localization and biochemical fractionation of NS5A. Taken together, our data suggest that NS5A inhibitors such as BMS-790052 can suppress viral genome replication by altering the proper localization of NS5A into functional RCs.
Collapse
Affiliation(s)
- Choongho Lee
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | |
Collapse
|
243
|
Mikami Y, Somei M, Tsuda H. SSH-BM-I, a tryptamine derivative, stimulates mineralization in terminal osteoblast differentiation but inhibits osteogenesis of pre-committed progenitor cells. J Pharmacol Sci 2011; 116:63-72. [PMID: 21487195 DOI: 10.1254/jphs.10329fp] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
SSH-BM-I was synthesized from tryptamine by using a newly developed synthetic method, and it has structural similarity to bromomelatonin. Recently, it had been reported that SSH-BM-I increases osteoblasts in scales of gold fish. However, the effect of SSH-BM-I on osteoblast differentiation in mammalian cells has not yet been examined. Therefore, this study examined the effect of SSH-BM-I on osteoblast differentiation in mesenchymal progenitor-like cells and mature osteoblast-like cells. SSH-BM-I enhanced terminal osteoblast differentiation, as indicated by mineralization, which was accompanied by upregulation of the osteogenic marker genes bone sialoprotein (BSP) and osteocalcin (OC). However, in mesenchymal progenitor ROB-C26 cultures, no mineralized nodules were observed regardless of SSH-BM-I treatment, although BMP-2 was able to induce nodule formation in these cells. Furthermore, BMP-2-induced nodule formation was suppressed by SSH-BM-I treatment in ROB-C26 cultures. We further investigated the impact of the timing and duration of SSH-BM-I treatment on osteoblast differentiation. The effect of SSH-BM-I treatment on osteoblast differentiation of ROB-C26 in the presence of BMP-2 switches from negative to positive sometime between day 6 and 9, because SSH-BM-I treatment enhanced the formation of mineralized nodules when it was started on day 9, but suppressed nodule formation when it was started at day 6 or earlier. These results suggest that the stimulatory effects of SSH-BM-I on the formation of mineralized nodules depend on the degree of cell differentiation.
Collapse
Affiliation(s)
- Yoshikazu Mikami
- Department of Anatomy, Nihon University School of Dentistry, Tokyo 101-8310, Japan.
| | | | | |
Collapse
|
244
|
Zaidi SK, Young DW, Montecino M, van Wijnen AJ, Stein JL, Lian JB, Stein GS. Bookmarking the genome: maintenance of epigenetic information. J Biol Chem 2011; 286:18355-61. [PMID: 21454629 DOI: 10.1074/jbc.r110.197061] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mitotic inheritance of gene function is obligatory to sustain biological control. Emerging evidence suggests that epigenetic mechanisms are linked to transmission of cell fate, lineage commitment, and maintenance of cellular phenotype in progeny cells. Mechanisms of epigenetic memory include gene silencing by DNA methylation, transcriptional regulation by histone modifications, regulation of gene expression by noncoding small RNA molecules, and retention of regulatory machinery on target gene loci for activation and repression. We will focus on the regulatory implications of epigenetic memory for physiological control and for the onset and progression of disease.
Collapse
Affiliation(s)
- Sayyed K Zaidi
- Department of Cell Biology and Cancer Center, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | | | | | | | | | | | |
Collapse
|
245
|
In vitro expression profiling of myostatin, follistatin, decorin and muscle-specific transcription factors in adult caprine contractile myotubes. J Muscle Res Cell Motil 2011; 32:23-30. [PMID: 21416152 DOI: 10.1007/s10974-011-9245-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Accepted: 03/11/2011] [Indexed: 10/18/2022]
Abstract
Skeletal muscle is one of the several adult postmitotic tissues that retain the capacity to regenerate, which relies on a population of quiescent precursors, termed satellite cells. Proliferation and differentiation of myoblasts to form mature myotubes in vitro has been a valuable tool in the characterization of the cellular events during myogenesis, which is a multistep process starting with progenitor cell proliferation, followed by their exit from the cell cycle, differentiation, alignment, and fusion to form multinucleated myotubes. A typical feature during muscle differentiation is the variation in expression of various genes along with myogenic factors. In this experiment, mRNA level of myostatin, follistatin, decorin and three muscle-specific transcription factors in adult caprine contractile myotubes have been studied through quantitative real time PCR. We observed that the expression level of myostatin, decorin, Myf5 and myogenin transcripts were significantly higher in contractile myotubes compared to myoblast monolayer (P < 0.05), and follistatin level was similar in both types of cells, whereas MyoD transcript level was significantly high in monolayer culture which might be due heterogeneity of myoblast population. It is concluded that the information generated would provide the base line information as well as monitoring markers to undertake experiments aimed at modulating muscle growth.
Collapse
|
246
|
Pas MFW, Visscher AH. Genetic regulation of meat production by embryonic muscle formation - a review. J Anim Breed Genet 2011; 111:404-12. [DOI: 10.1111/j.1439-0388.1994.tb00477.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
247
|
Hirai H, Verma M, Watanabe S, Tastad C, Asakura Y, Asakura A. MyoD regulates apoptosis of myoblasts through microRNA-mediated down-regulation of Pax3. ACTA ACUST UNITED AC 2011; 191:347-65. [PMID: 20956382 PMCID: PMC2958479 DOI: 10.1083/jcb.201006025] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Suppression of the myogenic transcription factor MyoD is required for maintenance of muscle stem cells. The molecules that regulate the apoptosis cascade are also involved in differentiation and syncytial fusion in skeletal muscle. MyoD is a myogenic transcription factor that plays essential roles in muscle differentiation. We noticed that MyoD−/− myoblasts display remarkable resistance to apoptosis by down-regulation of miR-1 (microRNA-1) and miR-206 and by up-regulation of Pax3. This resulted in transcriptional activation of antiapoptotic factors Bcl-2 and Bcl-xL. Forced MyoD expression induces up-regulation of miR-1 and miR-206 and down-regulation of Pax3, Bcl-2, and Bcl-xL along with increased apoptosis in MyoD−/− myoblasts. In contrast, MyoD gene knockdown increases cell survival of wild-type myoblasts. The 3′ untranslated region of Pax3 mRNA contains two conserved miR-1/miR-206–binding sites, which are required for targeting of these microRNAs (miRNAs). Therefore, these data suggest that MyoD not only regulates terminal differentiation but also apoptosis through miRNA-mediated down-regulation of Pax3. Finally, MyoD, miR-1, and miR-206 are all down-regulated in quiescent satellite cells, which may be required for maintenance of muscle stem cells.
Collapse
Affiliation(s)
- Hiroyuki Hirai
- Stem Cell Institute, Department of Neurology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | | | | | |
Collapse
|
248
|
Winbanks CE, Wang B, Beyer C, Koh P, White L, Kantharidis P, Gregorevic P. TGF-beta regulates miR-206 and miR-29 to control myogenic differentiation through regulation of HDAC4. J Biol Chem 2011; 286:13805-14. [PMID: 21324893 DOI: 10.1074/jbc.m110.192625] [Citation(s) in RCA: 225] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRs) are emerging as prominent players in the regulation of many biological processes, including myogenic commitment and skeletal muscle formation. Members of the TGF-β family can influence the proliferation and myogenic differentiation of cells, although it is presently not clear what role miRNAs play in the TGF-β-mediated control of myogenic differentiation. Here, we demonstrate in the myogenic C2C12 cell line, and in primary muscle cells, that miR-206 and miR-29-two miRs that act on transcriptional events implicated in muscle differentiation are down-regulated by TGF-β. We further demonstrate that TGF-β treatment of myogenic cells is associated with increased expression of histone deacetylase 4 (HDAC4), a key inhibitor of muscle differentiation that has been identified as a target for regulation by miR-206 and miR-29. We confirmed that increased expression of miR-206 and miR-29 resulted in the translational repression of HDAC4 in the presence or absence of TGF-β via interaction with the HDAC4 3'-untranslated region. Importantly, we found that miR-206 and miR-29 can attenuate the inhibitory actions of TGF-β on myogenic differentiation. Furthermore, we present evidence that the mechanism by which miR-206 and miR-29 can inhibit the TGF-β-mediated up-regulation of HDAC4 is via the inhibition of Smad3 expression, a transducer of TGF-β signaling. These findings identify a novel mechanism of interaction between TGF-β and miR-206 and -29 in the regulation of myogenic differentiation through HDAC4.
Collapse
Affiliation(s)
- Catherine E Winbanks
- Division of Metabolism and Obesity, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 8008, Australia
| | | | | | | | | | | | | |
Collapse
|
249
|
Ciemerych MA, Archacka K, Grabowska I, Przewoźniak M. Cell cycle regulation during proliferation and differentiation of mammalian muscle precursor cells. Results Probl Cell Differ 2011; 53:473-527. [PMID: 21630157 DOI: 10.1007/978-3-642-19065-0_20] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Proliferation and differentiation of muscle precursor cells are intensively studied not only in the developing mouse embryo but also using models of skeletal muscle regeneration or analyzing in vitro cultured cells. These analyses allowed to show the universality of the cell cycle regulation and also uncovered tissue-specific interplay between major cell cycle regulators and factors crucial for the myogenic differentiation. Examination of the events accompanying proliferation and differentiation leading to the formation of functional skeletal muscle fibers allows understanding the molecular basis not only of myogenesis but also of skeletal muscle regeneration. This chapter presents the basis of the cell cycle regulation in proliferating and differentiating muscle precursor cells during development and after muscle injury. It focuses at major cell cycle regulators, myogenic factors, and extracellular environment impacting on the skeletal muscle.
Collapse
Affiliation(s)
- Maria A Ciemerych
- Department of Cytology, Institute of Zoology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland.
| | | | | | | |
Collapse
|
250
|
|