101
|
Jin W, Peng J, Jiang S. The epigenetic regulation of embryonic myogenesis and adult muscle regeneration by histone methylation modification. Biochem Biophys Rep 2016; 6:209-219. [PMID: 28955879 PMCID: PMC5600456 DOI: 10.1016/j.bbrep.2016.04.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 04/14/2016] [Accepted: 04/18/2016] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle formation in vertebrates is derived from the paraxial mesoderm, which develops into myogenic precursor cells and finally differentiates into mature myofibers. This myogenic program involves temporal-spatial molecular events performed by transcription regulators (such as members of the Pax, MRFs and Six families) and signaling pathways (such as Wnts, BMP and Shh signaling). Epigenetic regulation, including histone post-translational modifications is crucial for controlling gene expression through recruitment of various chromatin-modifying enzymes that alter chromatin dynamics during myogenesis. The chromatin modifying enzymes are also recruited at regions of muscle gene regulation, coordinating transcription regulators to influence gene expression. In particular, the reversible methylation status of histone N-terminal tails provides the important regulatory mechanisms in either activation or repression of muscle genes. In this report, we review the recent literatures to deduce mechanisms underlying the epigenetic regulation of gene expression with a focus on histone methylation modification during embryo myogenesis and adult muscle regeneration. Recent results from different histone methylation/demethylation modifications have increased our understanding about the highly intricate layers of epigenetic regulations involved in myogenesis and cross-talk of histone enzymes with the muscle-specific transcriptional machinery. Myogenesis is influenced by regulation of transcription factors, signal pathways and post-transcriptional modifications. Histone methylation modifications as “on/off” switches regulated myogenic lineage commitment and differentiation. The myogenic regulatory factors and histone methylation modifications established dynamic regulatory mechanism.
Collapse
Key Words
- BMP4, bone morphogenic protein 4
- ChIP, chromatin immunoprecipitation
- Epigenetic
- H3K27, methylation of histone H3 lysine 27
- H3K4, methylation of histone H3 lysine 4
- H3K9, methylation of histone H3 lysine 9
- Histone methylation/demethylation modification
- KDMs, lysine demethyltransferases
- LSD1, lysine specific demethyltransferase 1
- MEF2, myocyte enhancer factor 2
- MRFs, myogenic regulatory factors
- Muscle differentiation
- Muscle progenitor cells
- Muscle regeneration
- Myogenesis
- PRC2, polycomb repressive complex 2
- SCs, satellite cells
- Shh, sonic hedgehog
- TSS, transcription start sites
- UTX, ubiquitously transcribed tetratricopeptide repeat, X chromosome
- bHLH, basic helix-loop-helix
- p38 MAPK, p38 mitogen-activated protein kinase
Collapse
Affiliation(s)
- Wei Jin
- Key Laboratory of Pig Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, PR China
| | - Siwen Jiang
- Key Laboratory of Pig Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, PR China.,Key Projects in the Cooperative Innovation Center for Sustainable Pig Production of Wuhan, PR China
| |
Collapse
|
102
|
Wang H, Li X, Liu H, Sun L, Zhang R, Li L, Wangding M, Wang J. Six1 induces protein synthesis signaling expression in duck myoblasts mainly via up-regulation of mTOR. Genet Mol Biol 2016; 39:151-61. [PMID: 27007909 PMCID: PMC4807382 DOI: 10.1590/1678-4685-gmb-2015-0075] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 08/24/2015] [Indexed: 01/05/2023] Open
Abstract
As a critical transcription factor, Six1 plays an important role in the regulation of myogenesis and muscle development. However, little is known about its regulatory mechanism associated with muscular protein synthesis. The objective of this study was to investigate the effects of overexpression ofSix1 on the expression of key protein metabolism-related genes in duck myoblasts. Through an experimental model where duck myoblasts were transfected with a pEGFP-duSix1 construct, we found that overexpression of duckSix1 could enhance cell proliferation activity and increase mRNA expression levels of key genes involved in the PI3K/Akt/mTOR signaling pathway, while the expression of FOXO1, MuRF1and MAFbx was not significantly altered, indicating thatSix1 could promote protein synthesis in myoblasts through up-regulating the expression of several related genes. Additionally, in duck myoblasts treated with LY294002 and rapamycin, the specific inhibitors ofPI3K and mTOR, respectively, the overexpression of Six1 could significantly ameliorate inhibitive effects of these inhibitors on protein synthesis. Especially, the mRNA expression levels of mTOR and S6K1 were observed to undergo a visible change, and a significant increase in protein expression of S6K1 was seen. These data suggested that Six1plays an important role in protein synthesis, which may be mainly due to activation of the mTOR signaling pathway.
Collapse
Affiliation(s)
- Haohan Wang
- Institute of Animal Genetics and Breedings, Sichuan Agricultural University, Ya'an, China
| | - Xinxin Li
- Institute of Animal Genetics and Breedings, Sichuan Agricultural University, Ya'an, China
| | - Hehe Liu
- Institute of Animal Genetics and Breedings, Sichuan Agricultural University, Ya'an, China
| | - Lingli Sun
- Institute of Animal Genetics and Breedings, Sichuan Agricultural University, Ya'an, China
| | - Rongping Zhang
- Institute of Animal Genetics and Breedings, Sichuan Agricultural University, Ya'an, China
| | - Liang Li
- Institute of Animal Genetics and Breedings, Sichuan Agricultural University, Ya'an, China
| | - Mincheng Wangding
- Institute of Animal Genetics and Breedings, Sichuan Agricultural University, Ya'an, China
| | - Jiwen Wang
- Institute of Animal Genetics and Breedings, Sichuan Agricultural University, Ya'an, China
| |
Collapse
|
103
|
Moody SA, Neilson KM, Kenyon KL, Alfandari D, Pignoni F. Using Xenopus to discover new genes involved in branchiootorenal spectrum disorders. Comp Biochem Physiol C Toxicol Pharmacol 2015; 178:16-24. [PMID: 26117063 PMCID: PMC4662879 DOI: 10.1016/j.cbpc.2015.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 12/14/2022]
Abstract
Congenital hearing loss is an important clinical problem because, without early intervention, affected children do not properly acquire language and consequently have difficulties developing social skills. Although most newborns in the US are screened for hearing deficits, even earlier diagnosis can be made with prenatal genetic screening. Genetic screening that identifies the relevant mutated gene can also warn about potential congenital defects in organs not related to hearing. We will discuss efforts to identify new candidate genes that underlie the Branchiootorenal spectrum disorders in which affected children have hearing deficits and are also at risk for kidney defects. Mutations in two genes, SIX1 and EYA1, have been identified in about half of the patients tested. To uncover new candidate genes, we have used the aquatic animal model, Xenopus laevis, to identify genes that are part of the developmental genetic pathway of Six1 during otic and kidney development. We have already identified a large number of potential Six1 transcriptional targets and candidate co-factor proteins that are expressed at the right time and in the correct tissues to interact with Six1 during development. We discuss the advantages of using this system for gene discovery in a human congenital hearing loss syndrome.
Collapse
Affiliation(s)
- Sally A Moody
- Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.
| | - Karen M Neilson
- Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Kristy L Kenyon
- Department of Biology, Hobart and William Smith Colleges, Geneva, NY, USA
| | - Dominique Alfandari
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Francesca Pignoni
- Department of Ophthalmology, Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
104
|
Okumura T, Furuichi K, Higashide T, Sakurai M, Hashimoto SI, Shinozaki Y, Hara A, Iwata Y, Sakai N, Sugiyama K, Kaneko S, Wada T. Association of PAX2 and Other Gene Mutations with the Clinical Manifestations of Renal Coloboma Syndrome. PLoS One 2015; 10:e0142843. [PMID: 26571382 PMCID: PMC4646464 DOI: 10.1371/journal.pone.0142843] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 10/27/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Renal coloboma syndrome (RCS) is characterized by renal anomalies and optic nerve colobomas. PAX2 mutations contribute to RCS. However, approximately half of the patients with RCS have no mutation in PAX2 gene. METHODS To investigate the incidence and effects of mutations of PAX2 and 25 candidate genes, patient genes were screened using next-generation sequence analysis, and candidate mutations were confirmed using Sanger sequencing. The correlation between mutations and clinical manifestation was evaluated. RESULT Thirty patients, including 26 patients (two families of five and two, 19 sporadic cases) with RCS, and 4 optic nerve coloboma only control cases were evaluated in the present study. Six PAX2 mutations in 21 probands [28%; two in family cohorts (n = 5 and n = 2) and in 4 out of 19 patients with sporadic disease] including four novel mutations were confirmed using Sanger sequencing. Moreover, four other sequence variants (CHD7, SALL4, KIF26B, and SIX4) were also confirmed, including a potentially pathogenic novel KIF26B mutation. Kidney function and proteinuria were more severe in patients with PAX2 mutations than in those without the mutation. Moreover, the coloboma score was significantly higher in patients with PAX2 gene mutations. Three out of five patients with PAX2 mutations had focal segmental glomerulosclerosis (FSGS) diagnosed from kidney biopsies. CONCLUSION The results of this study identify several new mutations of PAX2, and sequence variants in four additional genes, including a novel potentially pathogenic mutation in KIF26B, which may play a role in the pathogenesis of RCS.
Collapse
Affiliation(s)
- Toshiya Okumura
- Department of Disease Control, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Japan
| | - Kengo Furuichi
- Department of Disease Control, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Japan
- Division of Blood Purification, Kanazawa University Hospital, Kanazawa, Japan
- * E-mail:
| | - Tomomi Higashide
- Departments of Ophthalmology and Visual Science, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Japan
| | - Mayumi Sakurai
- Departments of Ophthalmology and Visual Science, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Japan
| | - Shin-ichi Hashimoto
- Division of Nephrology, Department of Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Japan
| | - Yasuyuki Shinozaki
- Division of Nephrology, Department of Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Japan
| | - Akinori Hara
- Department of Disease Control, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Japan
| | - Yasunori Iwata
- Division of Nephrology, Department of Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Japan
| | - Norihiko Sakai
- Department of Disease Control, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Japan
- Division of Blood Purification, Kanazawa University Hospital, Kanazawa, Japan
| | - Kazuhisa Sugiyama
- Departments of Ophthalmology and Visual Science, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Japan
| | - Shuichi Kaneko
- Department of Disease Control, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Japan
| | - Takashi Wada
- Division of Nephrology, Department of Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
105
|
Daubas P, Duval N, Bajard L, Langa Vives F, Robert B, Mankoo BS, Buckingham M. Fine-tuning the onset of myogenesis by homeobox proteins that interact with the Myf5 limb enhancer. Biol Open 2015; 4:1614-24. [PMID: 26538636 PMCID: PMC4736032 DOI: 10.1242/bio.014068] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Skeletal myogenesis in vertebrates is initiated at different sites of skeletal muscle formation during development, by activation of specific control elements of the myogenic regulatory genes. In the mouse embryo, Myf5 is the first myogenic determination gene to be expressed and its spatiotemporal regulation requires multiple enhancer sequences, extending over 120 kb upstream of the Mrf4-Myf5 locus. An enhancer, located at −57/−58 kb from Myf5, is responsible for its activation in myogenic cells derived from the hypaxial domain of the somite, that will form limb muscles. Pax3 and Six1/4 transcription factors are essential activators of this enhancer, acting on a 145-bp core element. Myogenic progenitor cells that will form the future muscle masses of the limbs express the factors necessary for Myf5 activation when they delaminate from the hypaxial dermomyotome and migrate into the forelimb bud, however they do not activate Myf5 and the myogenic programme until they have populated the prospective muscle masses. We show that Msx1 and Meox2 homeodomain-containing transcription factors bind in vitro and in vivo to specific sites in the 145-bp element, and are implicated in fine-tuning activation of Myf5 in the forelimb. Msx1, when bound between Pax and Six sites, prevents the binding of these key activators, thus inhibiting transcription of Myf5 and consequent premature myogenic differentiation. Meox2 is required for Myf5 activation at the onset of myogenesis via direct binding to other homeodomain sites in this sequence. Thus, these homeodomain factors, acting in addition to Pax3 and Six1/4, fine-tune the entry of progenitor cells into myogenesis at early stages of forelimb development. Summary: Homeodomain factors Msx1 and Meox2, acting in addition to Pax3 and Six1/4, fine-tune the entry of progenitor cells into myogenesis at early stages of forelimb development via modulation of limb enhancer gene Myf5.
Collapse
Affiliation(s)
- Philippe Daubas
- CNRS URA 2578, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
| | - Nathalie Duval
- CNRS URA 2578, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
| | - Lola Bajard
- CNRS URA 2578, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
| | | | - Benoît Robert
- CNRS URA 2578, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
| | - Baljinder S Mankoo
- King's College London, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Margaret Buckingham
- CNRS URA 2578, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
| |
Collapse
|
106
|
Endo T. Molecular mechanisms of skeletal muscle development, regeneration, and osteogenic conversion. Bone 2015; 80:2-13. [PMID: 26453493 DOI: 10.1016/j.bone.2015.02.028] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 02/18/2015] [Accepted: 02/28/2015] [Indexed: 12/21/2022]
Abstract
Both skeletal muscle and bone are of mesodermal origin and derived from somites during embryonic development. Somites differentiate into the dorsal dermomyotome and the ventral sclerotome, which give rise to skeletal muscle and bone, respectively. Extracellular signaling molecules, such as Wnt and Shh, secreted from the surrounding environment, determine the developmental fate of skeletal muscle. Dermomyotome cells are specified as trunk muscle progenitor cells by transcription factor networks involving Pax3. These progenitor cells delaminate and migrate to form the myotome, where they are determined as myoblasts that differentiate into myotubes or myofibers. The MyoD family of transcription factors plays pivotal roles in myogenic determination and differentiation. Adult skeletal muscle regenerates upon exercise, muscle injury, or degeneration. Satellite cells are muscle-resident stem cells and play essential roles in muscle growth and regeneration. Muscle regeneration recapitulates the process of muscle development in many aspects. In certain muscle diseases, ectopic calcification or heterotopic ossification, as well as fibrosis and adipogenesis, occurs in skeletal muscle. Muscle-resident mesenchymal progenitor cells, which may be derived from vascular endothelial cells, are responsible for the ectopic osteogenesis, fibrogenesis, and adipogenesis. The small GTPase M-Ras is likely to participate in the ectopic calcification and ossification, as well as in osteogenesis during development. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
Affiliation(s)
- Takeshi Endo
- Department of Biology, Graduate School of Science, Chiba University, Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan.
| |
Collapse
|
107
|
Chakroun I, Yang D, Girgis J, Gunasekharan A, Phenix H, Kærn M, Blais A. Genome-wide association between Six4, MyoD, and the histone demethylase Utx during myogenesis. FASEB J 2015; 29:4738-55. [PMID: 26229056 DOI: 10.1096/fj.15-277053] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/20/2015] [Indexed: 01/14/2023]
Abstract
Adult skeletal muscles can regenerate after injury, due to the presence of satellite cells, a quiescent population of myogenic progenitor cells. Once activated, satellite cells repair the muscle damage by undergoing myogenic differentiation. The myogenic regulatory factors (MRFs) coordinate the process of progenitor differentiation in cooperation with other families of transcription factors (TFs). The Six1 and Six4 homeodomain TFs are expressed in developing and adult muscle and Six1 is critical for embryonic and adult myogenesis. However, the lack of a muscle developmental phenotype in Six4-null mice, which has been attributed to compensation by other Six family members, has discouraged further assessment of the role of Six4 during adult muscle regeneration. By employing genome-wide approaches to address the function of Six4 during adult skeletal myogenesis, we have identified a core set of muscle genes coordinately regulated in adult muscle precursors by Six4 and the MRF MyoD. Throughout the genome of differentiating adult myoblasts, the cooperation between Six4 and MyoD is associated with chromatin repressive mark removal by Utx, a demethylase of histone H3 trimethylated at lysine 27. Among the genes coordinately regulated by Six4 and MyoD are several genes critical for proper in vivo muscle regeneration, implicating a role of Six4 in this process. Using in vivo RNA interference of Six4, we expose an uncompensated function of this TF during muscle regeneration. Together, our results reveal a role for Six4 during adult muscle regeneration and suggest a widespread mechanism of cooperation between Six4 and MyoD.
Collapse
Affiliation(s)
- Imane Chakroun
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Dabo Yang
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - John Girgis
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Atchayaa Gunasekharan
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Hilary Phenix
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Mads Kærn
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Alexandre Blais
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
108
|
de Taffin M, Carrier Y, Dubois L, Bataillé L, Painset A, Le Gras S, Jost B, Crozatier M, Vincent A. Genome-Wide Mapping of Collier In Vivo Binding Sites Highlights Its Hierarchical Position in Different Transcription Regulatory Networks. PLoS One 2015. [PMID: 26204530 PMCID: PMC4512700 DOI: 10.1371/journal.pone.0133387] [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] [Indexed: 11/18/2022] Open
Abstract
Collier, the single Drosophila COE (Collier/EBF/Olf-1) transcription factor, is required in several developmental processes, including head patterning and specification of muscle and neuron identity during embryogenesis. To identify direct Collier (Col) targets in different cell types, we used ChIP-seq to map Col binding sites throughout the genome, at mid-embryogenesis. In vivo Col binding peaks were associated to 415 potential direct target genes. Gene Ontology analysis revealed a strong enrichment in proteins with DNA binding and/or transcription-regulatory properties. Characterization of a selection of candidates, using transgenic CRM-reporter assays, identified direct Col targets in dorso-lateral somatic muscles and specific neuron types in the central nervous system. These data brought new evidence that Col direct control of the expression of the transcription regulators apterous and eyes-absent (eya) is critical to specifying neuronal identities. They also showed that cross-regulation between col and eya in muscle progenitor cells is required for specification of muscle identity, revealing a new parallel between the myogenic regulatory networks operating in Drosophila and vertebrates. Col regulation of eya, both in specific muscle and neuronal lineages, may illustrate one mechanism behind the evolutionary diversification of Col biological roles.
Collapse
Affiliation(s)
- Mathilde de Taffin
- Centre de Biologie du Développement, UMR 5547 CNRS Université de Toulouse 3, 118 route de Narbonne, F-31062, Toulouse cedex 09, France
| | - Yannick Carrier
- Centre de Biologie du Développement, UMR 5547 CNRS Université de Toulouse 3, 118 route de Narbonne, F-31062, Toulouse cedex 09, France
| | - Laurence Dubois
- Centre de Biologie du Développement, UMR 5547 CNRS Université de Toulouse 3, 118 route de Narbonne, F-31062, Toulouse cedex 09, France
| | - Laetitia Bataillé
- Centre de Biologie du Développement, UMR 5547 CNRS Université de Toulouse 3, 118 route de Narbonne, F-31062, Toulouse cedex 09, France
| | - Anaïs Painset
- Centre de Biologie du Développement, UMR 5547 CNRS Université de Toulouse 3, 118 route de Narbonne, F-31062, Toulouse cedex 09, France
- Plate-forme bio-informatique Genotoul/MIA-T, INRA, Borde Rouge, 31326, Castanet-Tolosan, France
| | - Stéphanie Le Gras
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 67404, Illkirch, France
| | - Bernard Jost
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 67404, Illkirch, France
| | - Michèle Crozatier
- Centre de Biologie du Développement, UMR 5547 CNRS Université de Toulouse 3, 118 route de Narbonne, F-31062, Toulouse cedex 09, France
| | - Alain Vincent
- Centre de Biologie du Développement, UMR 5547 CNRS Université de Toulouse 3, 118 route de Narbonne, F-31062, Toulouse cedex 09, France
- * E-mail:
| |
Collapse
|
109
|
Khodabukus A, Baar K. The effect of serum origin on tissue engineered skeletal muscle function. J Cell Biochem 2015; 115:2198-207. [PMID: 25146978 DOI: 10.1002/jcb.24938] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 08/15/2014] [Indexed: 02/02/2023]
Abstract
Skeletal muscle phenotype is regulated by a complex interaction between genetic, hormonal, and electrical inputs. However, because of the interrelatedness of these factors in vivo it is difficult to determine the importance of one over the other. Over the last 5 years, we have engineered skeletal muscles in the European Union (EU) and the United States (US) using the same clone of C2C12 cells. Strikingly, the dynamics of contraction of the muscles was dramatically different. Therefore, in this study we sought to determine whether the hormonal milieu (source of fetal bovine serum (FBS)) could alter engineered muscle phenotype. In muscles engineered in serum of US origin time-to-peak tension (2.2-fold), half relaxation (2.6-fold), and fatigue resistance (improved 25%) all showed indications of a shift towards a slower phenotype. Even though there was a dramatic shift in the rate of contraction, myosin heavy chain expression was the same. The contraction speed was instead related to a shift in calcium release/sensitivity proteins (DHPR = 3.1-fold lower, slow CSQ = 3.4-fold higher, and slow TnT = 2.4-fold higher) and calcium uptake proteins (slow SERCA = 1.7-fold higher and parvalbumin = 41-fold lower). These shifts in calcium dynamics were accompanied by a partial shift in metabolic enzymes, but could not be explained by purported regulators of muscle phenotype. These data suggest that hormonal differences in serum of USDA and EU origin cause a shift in calcium handling resulting in a dramatic change in engineered muscle function.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA, 95616, USA
| | | |
Collapse
|
110
|
Zeng J, Shi R, Cai CX, Liu XR, Song YB, Wei M, Ma WL. Increased expression of Six1 correlates with progression and prognosis of prostate cancer. Cancer Cell Int 2015; 15:63. [PMID: 26161040 PMCID: PMC4497425 DOI: 10.1186/s12935-015-0215-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 06/08/2015] [Indexed: 12/28/2022] Open
Abstract
Sineoculis homeobox homolog 1 (Six1), normally a developmentally restricted transcriptional regulator, is frequently dysregulated in mutiple cancers. Increasing evidences show that overexpression of Six1 plays a key role in tumorigenesis. However, the Six1 expression status and its relationship with the clinicopathological characteristics in prostate cancer were unclear. In this study, the mRNA and protein levels of Six1 in prostate cancer tissues and normal prostate tissues were evaluated. The clinicopathological significance of Six1 was investigated by immunohistochemistry (IHC) on a prostate cancer tissue microarray. The cut-off score for high expression of Six1 was determined by the receiver-operating characteristic (ROC) analysis. The correlation between Six1 protein expression and clinicopathological characteristics of prostate cancer was analyzed by Chi-square test. Increased expression of Six1 protein was observed in the majority of prostate cancer, compared with their paired adjacent normal prostate tissues. When Six1 high expression percentage was determined to be above 55 % (area under ROC curve = 0.881, P = 0.000), high expression of Six1 was observed in 55.6 % (80/144) of prostate cancer tissues and low expression of Six1 was observed in all normal prostate tissues by IHC. Increased expression of Six1 in patients was correlated with high histological grade (χ2 = 58.651, P = 0.00), advanced clinical stage (χ2 = 57.330, P = 0.000), high Gleason score (χ2 = 63.480, P = 0.000), high primary tumor grade (χ2 = 57.330, P = 0.000) and positive regional lymph node metastasis (χ2 = 19.294, P = 0.000). Furthermore, univariate and multivariate survival analysis suggested that Six1 was an independent prognostic indicator for overall survival (P < 0.05). This study suggests that Six1 could be served as an additional biomarker in identifying prostate cancer patients at risk of tumor progression, might potentially be used for predicting survival outcome of patients with prostate cancer.
Collapse
Affiliation(s)
- Jun Zeng
- Institute of Genetic Engineering, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, People's Republic of China
| | - Rong Shi
- Institute of Genetic Engineering, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, People's Republic of China
| | - Cui-Xia Cai
- Institute of Genetic Engineering, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, People's Republic of China
| | - Xin-Rui Liu
- Institute of Genetic Engineering, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, People's Republic of China
| | - Yan-Bin Song
- Institute of Genetic Engineering, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, People's Republic of China
| | - Min Wei
- Institute of Genetic Engineering, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, People's Republic of China
| | - Wen-Li Ma
- Institute of Genetic Engineering, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, People's Republic of China
| |
Collapse
|
111
|
Xu J, Xu PX. Eya-six are necessary for survival of nephrogenic cord progenitors and inducing nephric duct development before ureteric bud formation. Dev Dyn 2015; 244:866-73. [PMID: 25903664 DOI: 10.1002/dvdy.24282] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/09/2015] [Accepted: 04/10/2015] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Specification of the metanephric mesenchyme is a central step of kidney development as this mesenchyme promotes nephric duct induction to form a ureteric bud near its caudal end. Before ureteric bud formation, the caudal nephric duct swells to form a pseudostratified epithelial domain that later emerges as the tip of the bud. However, the signals that promote the formation of the transient epithelial domain remain unclear. Here, we investigated the early roles of the mesenchymal factor Six family and its cofactor Eya on the initial induction of nephric duct development. RESULTS The nephrogenic progenitor population is initially present but significantly reduced in mice lacking both Six1 and Six4 and undertakes an abnormal cell death pathway to be completely eliminated by ∼E10.5-E11.0, similar to that observed in Eya1(-/-) embryos. Consequently, the nephric duct fails to be induced to undergo normal proliferation to pseudostratify and form the ureteric bud in Six1(-/-) ;Six4(-/-) or Eya1(-/-) embryos. CONCLUSIONS Our data support a model where Eya-Six may form a complex to regulate nephron progenitor cell development before metanephric specification and are critical mesenchymal factors for inducing nephric duct development.
Collapse
Affiliation(s)
- Jinshu Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pin-Xian Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
112
|
Nogueira JM, Hawrot K, Sharpe C, Noble A, Wood WM, Jorge EC, Goldhamer DJ, Kardon G, Dietrich S. The emergence of Pax7-expressing muscle stem cells during vertebrate head muscle development. Front Aging Neurosci 2015; 7:62. [PMID: 26042028 PMCID: PMC4436886 DOI: 10.3389/fnagi.2015.00062] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 04/10/2015] [Indexed: 12/13/2022] Open
Abstract
Pax7 expressing muscle stem cells accompany all skeletal muscles in the body and in healthy individuals, efficiently repair muscle after injury. Currently, the in vitro manipulation and culture of these cells is still in its infancy, yet muscle stem cells may be the most promising route toward the therapy of muscle diseases such as muscular dystrophies. It is often overlooked that muscular dystrophies affect head and body skeletal muscle differently. Moreover, these muscles develop differently. Specifically, head muscle and its stem cells develop from the non-somitic head mesoderm which also has cardiac competence. To which extent head muscle stem cells retain properties of the early head mesoderm and might even be able to switch between a skeletal muscle and cardiac fate is not known. This is due to the fact that the timing and mechanisms underlying head muscle stem cell development are still obscure. Consequently, it is not clear at which time point one should compare the properties of head mesodermal cells and head muscle stem cells. To shed light on this, we traced the emergence of head muscle stem cells in the key vertebrate models for myogenesis, chicken, mouse, frog and zebrafish, using Pax7 as key marker. Our study reveals a common theme of head muscle stem cell development that is quite different from the trunk. Unlike trunk muscle stem cells, head muscle stem cells do not have a previous history of Pax7 expression, instead Pax7 expression emerges de-novo. The cells develop late, and well after the head mesoderm has committed to myogenesis. We propose that this unique mechanism of muscle stem cell development is a legacy of the evolutionary history of the chordate head mesoderm.
Collapse
Affiliation(s)
- Julia Meireles Nogueira
- School of Pharmacy and Biomedical Sciences, Institute for Biomedical and Biomolecular Science, University of Portsmouth Portsmouth, UK ; Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Katarzyna Hawrot
- School of Pharmacy and Biomedical Sciences, Institute for Biomedical and Biomolecular Science, University of Portsmouth Portsmouth, UK
| | - Colin Sharpe
- School of Biological Sciences, Institute for Biomedical and Biomolecular Science, University of Portsmouth Portsmouth, UK
| | - Anna Noble
- European Xenopus Resource Centre, School of Biological Sciences, University of Portsmouth Portsmouth, UK
| | - William M Wood
- Department of Molecular and Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut Storrs, CT, USA
| | - Erika C Jorge
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - David J Goldhamer
- Department of Molecular and Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut Storrs, CT, USA
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah Salt Lake City, UT, USA
| | - Susanne Dietrich
- School of Pharmacy and Biomedical Sciences, Institute for Biomedical and Biomolecular Science, University of Portsmouth Portsmouth, UK
| |
Collapse
|
113
|
|
114
|
Carrió E, Suelves M. DNA methylation dynamics in muscle development and disease. Front Aging Neurosci 2015; 7:19. [PMID: 25798107 PMCID: PMC4350440 DOI: 10.3389/fnagi.2015.00019] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 02/15/2015] [Indexed: 12/12/2022] Open
Abstract
DNA methylation is an essential epigenetic modification for mammalian development and is crucial for the establishment and maintenance of cellular identity. Traditionally, DNA methylation has been considered as a permanent repressive epigenetic mark. However, the application of genome-wide approaches has allowed the analysis of DNA methylation in different genomic contexts revealing a more dynamic regulation than originally thought, since active DNA methylation and demethylation occur during cellular differentiation and tissue specification. Satellite cells are the primary stem cells in adult skeletal muscle and are responsible for postnatal muscle growth, hypertrophy, and muscle regeneration. This review outlines the published data regarding DNA methylation changes along the skeletal muscle program, in both physiological and pathological conditions, to better understand the epigenetic mechanisms that control myogenesis.
Collapse
Affiliation(s)
- Elvira Carrió
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC) and Health Sciences Research Institute Germans Trias I Pujol (IGTP) Badalona, Spain
| | - Mònica Suelves
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC) and Health Sciences Research Institute Germans Trias I Pujol (IGTP) Badalona, Spain
| |
Collapse
|
115
|
Wotton KR, Schubert FR, Dietrich S. Hypaxial muscle: controversial classification and controversial data? Results Probl Cell Differ 2015; 56:25-48. [PMID: 25344665 DOI: 10.1007/978-3-662-44608-9_2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hypaxial muscle is the anatomical term commonly used when referring to all the ventrally located musculature in the body of vertebrates, including muscles of the body wall and the limbs. Yet these muscles had very humble beginnings when vertebrates evolved from their chordate ancestors, and complex anatomical changes and changes in underlying gene regulatory networks occurred. This review summarises the current knowledge and controversies regarding the development and evolution of hypaxial muscles.
Collapse
Affiliation(s)
- Karl R Wotton
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Dr. Aiguader 88, 08003, Barcelona, Spain
| | | | | |
Collapse
|
116
|
Yan B, Neilson KM, Ranganathan R, Maynard T, Streit A, Moody SA. Microarray identification of novel genes downstream of Six1, a critical factor in cranial placode, somite, and kidney development. Dev Dyn 2014; 244:181-210. [PMID: 25403746 DOI: 10.1002/dvdy.24229] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 11/03/2014] [Accepted: 11/12/2014] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Six1 plays an important role in the development of several vertebrate organs, including cranial sensory placodes, somites, and kidney. Although Six1 mutations cause one form of branchio-otic syndrome (BOS), the responsible gene in many patients has not been identified; genes that act downstream of Six1 are potential BOS candidates. RESULTS We sought to identify novel genes expressed during placode, somite and kidney development by comparing gene expression between control and Six1-expressing ectodermal explants. The expression patterns of 19 of the significantly up-regulated and 11 of the significantly down-regulated genes were assayed from cleavage to larval stages. A total of 28/30 genes are expressed in the otocyst, a structure that is functionally disrupted in BOS, and 26/30 genes are expressed in the nephric mesoderm, a structure that is functionally disrupted in the related branchio-otic-renal (BOR) syndrome. We also identified the chick homologues of five genes and show that they have conserved expression patterns. CONCLUSIONS Of the 30 genes selected for expression analyses, all are expressed at many of the developmental times and appropriate tissues to be regulated by Six1. Many have the potential to play a role in the disruption of hearing and kidney function seen in BOS/BOR patients.
Collapse
Affiliation(s)
- Bo Yan
- Department of Anatomy and Regenerative Biology, The George Washington University, School of Medicine and Health Sciences, Washington, DC
| | | | | | | | | | | |
Collapse
|
117
|
Francetic T, Li Q. Skeletal myogenesis and Myf5 activation. Transcription 2014; 2:109-114. [PMID: 21922054 DOI: 10.4161/trns.2.3.15829] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 04/12/2011] [Accepted: 04/13/2011] [Indexed: 11/19/2022] Open
Abstract
Myogenic regulatory factors (MRFs) are the master regulators of skeletal myogenesis. Among the MRFs, Myf5 is the earliest to be expressed and is regulated by a complex set of enhancers. The expression of Myf5 defines different muscle populations in the somite. Furthermore, Myf5 expression is also found in non-muscle tissues, such as preadipocytes and neurons. Here, we present a current view on the regulation of skeletal myogenesis by transcription factors and cellular signals, with an emphasis on the complexity of transcriptional activation of Myf5. We also discuss Myf5 expression in different populations of myoblasts, preadipocytes and neuronal tissue.
Collapse
Affiliation(s)
- Tanja Francetic
- Department of Cellular and Molecular Medicine; Faculty of Medicine; University of Ottawa; Ottawa, ON Canada
| | | |
Collapse
|
118
|
Kim JA, Shon YH, Lim JO, Yoo JJ, Shin HI, Park EK. MYOD mediates skeletal myogenic differentiation of human amniotic fluid stem cells and regeneration of muscle injury. Stem Cell Res Ther 2014; 4:147. [PMID: 24331373 PMCID: PMC4054934 DOI: 10.1186/scrt358] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 10/18/2013] [Accepted: 12/03/2013] [Indexed: 12/21/2022] Open
Abstract
Introduction Human amniotic fluid stem (hAFS) cells have been shown to differentiate into multiple lineages, including myoblasts. However, molecular mechanisms underlying the myogenic differentiation of hAFS cells and their regenerative potential for muscle injury remain to be elucidated. Methods In order to induce myogenic differentiation of hAFS cells, lentiviruses for MYOD were constructed and transduced into hAFS cells. Formation of myotube-like cells was analyzed by immunocytochemistry, and expression of molecular markers for myoblasts was analyzed by reverse transcription polymerase chain reaction and Western blotting. For in vivo muscle regeneration, MYOD transduced hAFS cells were injected into left tibialis anterior (TA) muscles injured with cardiotoxin, and muscle regeneration was analyzed using hematoxylin and eosin, immunocytochemistry and formation of neuro-muscular junction. Results MYOD expression in hAFS cells successfully induced differentiation into multinucleated myotube-like cells. Consistently, significant expression of myogenic marker genes, such as MYOG, DES, DMD and MYH, was induced by MYOD. Analysis of pre-myogenic factors showed that expression of PAX3, MEOX1 and EYA2 was significantly increased by MYOD. MYOD was phosphorylated and localized in the nucleus. These results suggest that in hAFS cells, MYOD is phosphorylated and localized in the nucleus, thus inducing expression of myogenic factors, resulting in myogenic differentiation of hAFS cells. To test regenerative potential of MYOD-transduced hAFS cells, we transplanted them into injured muscles of immunodeficient BALB/cSlc-nu mice. The results showed a substantial increase in the volume of TA muscle injected with MYOD-hAFS cells. In addition, TA muscle tissue injected with MYOD-hAFS cells has more numbers of neuro-muscular junctions compared to controls, indicating functional restoration of muscle injury by MYOD-hAFS cells. Conclusions Collectively, our data suggest that transduction of hAFS cells with MYOD lentiviruses induces skeletal myogenic differentiation in vitro and morphological and functional regeneration of injured muscle in vivo.
Collapse
|
119
|
Amann R, Wyder S, Slavotinek AM, Trueb B. The FgfrL1 receptor is required for development of slow muscle fibers. Dev Biol 2014; 394:228-41. [DOI: 10.1016/j.ydbio.2014.08.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 08/13/2014] [Accepted: 08/16/2014] [Indexed: 02/03/2023]
|
120
|
Grifone R, Xie X, Bourgeois A, Saquet A, Duprez D, Shi DL. The RNA-binding protein Rbm24 is transiently expressed in myoblasts and is required for myogenic differentiation during vertebrate development. Mech Dev 2014; 134:1-15. [PMID: 25217815 DOI: 10.1016/j.mod.2014.08.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 08/05/2014] [Accepted: 08/22/2014] [Indexed: 12/16/2022]
Abstract
RNA-binding proteins (RBP) contribute to gene regulation through post-transcriptional events. Despite the important roles demonstrated for several RBP in regulating skeletal myogenesis in vitro, very few RBP coding genes have been characterized during skeletal myogenesis in vertebrate embryo. In the present study we report that Rbm24, which encodes the RNA-binding motif protein 24, is required for skeletal muscle differentiation in vivo. We show that Rbm24 transcripts are expressed at all sites of skeletal muscle formation during embryogenesis of different vertebrates, including axial, limb and head muscles. Interestingly, we find that Rbm24 protein starts to accumulate in MyoD-positive myoblasts and is transiently expressed at the onset of muscle cell differentiation. It accumulates in myotomal and limb myogenic cells, but not in Pax3-positive progenitor cells. Rbm24 expression is under the direct regulation by MyoD, as demonstrated by in vivo chromatin immunoprecipitation assay. Using morpholino knockdown approach, we further show that Rbm24 is required for somitic myogenic progenitor cells to differentiate into muscle cells during chick somitic myogenesis. Altogether, these results highlight Rbm24 as a novel key regulator of the myogenic differentiation program during vertebrate development.
Collapse
Affiliation(s)
- Raphaëlle Grifone
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France; CNRS, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France
| | - Xin Xie
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France; CNRS, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France
| | - Adeline Bourgeois
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France; CNRS, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France
| | - Audrey Saquet
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France; CNRS, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France
| | - Delphine Duprez
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France; CNRS, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France
| | - De-Li Shi
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France; CNRS, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France.
| |
Collapse
|
121
|
Wu W, Huang R, Wu Q, Li P, Chen J, Li B, Liu H. The role of Six1 in the genesis of muscle cell and skeletal muscle development. Int J Biol Sci 2014; 10:983-9. [PMID: 25210496 PMCID: PMC4159689 DOI: 10.7150/ijbs.9442] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 06/06/2014] [Indexed: 02/06/2023] Open
Abstract
The sine oculis homeobox 1 (Six1) gene encodes an evolutionarily conserved transcription factor. In the past two decades, much research has indicated that Six1 is a powerful regulator participating in skeletal muscle development. In this review, we summarized the discovery and structural characteristics of Six1 gene, and discussed the functional roles and molecular mechanisms of Six1 in myogenesis and in the formation of skeletal muscle fibers. Finally, we proposed areas of future interest for understanding Six1 gene function.
Collapse
Affiliation(s)
- Wangjun Wu
- 1. Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; ; 2. Huaian Academy of Nanjing Agricultural University, Huaian, Jiangsu, 223001, China
| | - Ruihua Huang
- 1. Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; ; 2. Huaian Academy of Nanjing Agricultural University, Huaian, Jiangsu, 223001, China
| | - Qinghua Wu
- 3. College of Life Science, Yangtze University, Jingzhou, Hubei, 434023, China. ; 4. Center for Basic and Applied Research, Faculty of Informatics and Management, University of Hradec Kradec Kralove, Hradec Kralove, Czech Republic
| | - Pinghua Li
- 1. Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; ; 2. Huaian Academy of Nanjing Agricultural University, Huaian, Jiangsu, 223001, China
| | - Jie Chen
- 1. Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Bojiang Li
- 1. Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Honglin Liu
- 1. Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
122
|
L'honoré A, Commère PH, Ouimette JF, Montarras D, Drouin J, Buckingham M. Redox regulation by Pitx2 and Pitx3 is critical for fetal myogenesis. Dev Cell 2014; 29:392-405. [PMID: 24871946 DOI: 10.1016/j.devcel.2014.04.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 02/21/2014] [Accepted: 04/03/2014] [Indexed: 10/25/2022]
Abstract
During development, major metabolic changes occur as cells become more specialized within a lineage. In the case of skeletal muscle, differentiation is accompanied by a switch from a glycolytic proliferative progenitor state to an oxidative postmitotic differentiated state. Such changes require extensive mitochondrial biogenesis leading to increased reactive oxygen species (ROS) production that needs to be balanced by an antioxidant system. Our analysis of double conditional Pitx2/3 mouse mutants, both in vivo during fetal myogenesis and ex vivo in primary muscle cell cultures, reveals excessive upregulation of ROS levels leading to DNA damage and apoptosis of differentiating cells. This is a consequence of downregulation of Nrf1 and genes for antioxidant enzymes, direct targets of Pitx2/3, leading to decreased expression of antioxidant enzymes, as well as impairment of mitochondrial function. Our analysis identifies Pitx2 and Pitx3 as key regulators of the intracellular redox state preventing DNA damage as cells undergo differentiation.
Collapse
Affiliation(s)
- Aurore L'honoré
- Department of Developmental and Stem Cell Biology, CNRS URA 2578, 28 rue du Dr Roux, 75015 Paris, France.
| | | | - Jean-François Ouimette
- Laboratory of Molecular Genetics, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Didier Montarras
- Department of Developmental and Stem Cell Biology, CNRS URA 2578, 28 rue du Dr Roux, 75015 Paris, France
| | - Jacques Drouin
- Laboratory of Molecular Genetics, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Margaret Buckingham
- Department of Developmental and Stem Cell Biology, CNRS URA 2578, 28 rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
123
|
Sakakibara I, Santolini M, Ferry A, Hakim V, Maire P. Six homeoproteins and a Iinc-RNA at the fast MYH locus lock fast myofiber terminal phenotype. PLoS Genet 2014; 10:e1004386. [PMID: 24852826 PMCID: PMC4031048 DOI: 10.1371/journal.pgen.1004386] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 04/02/2014] [Indexed: 12/18/2022] Open
Abstract
Thousands of long intergenic non-coding RNAs (lincRNAs) are encoded by the mammalian genome. However, the function of most of these lincRNAs has not been identified in vivo. Here, we demonstrate a role for a novel lincRNA, linc-MYH, in adult fast-type myofiber specialization. Fast myosin heavy chain (MYH) genes and linc-MYH share a common enhancer, located in the fast MYH gene locus and regulated by Six1 homeoproteins. linc-MYH in nuclei of fast-type myofibers prevents slow-type and enhances fast-type gene expression. Functional fast-sarcomeric unit formation is achieved by the coordinate expression of fast MYHs and linc-MYH, under the control of a common Six-bound enhancer.
Collapse
Affiliation(s)
- Iori Sakakibara
- INSERM U1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marc Santolini
- Laboratoire de Physique Statistique, CNRS, Université P. et M. Curie, Université D. Diderot, École Normale Supérieure, Paris, France
| | - Arnaud Ferry
- CNRS UMR 8104, Paris, France
- Université Pierre et Marie Curie-Paris 6, Sorbonne Universités, UMR S794, INSERM U974, CNRS UMR7215, Institut de Myologie, Paris, France
| | - Vincent Hakim
- Laboratoire de Physique Statistique, CNRS, Université P. et M. Curie, Université D. Diderot, École Normale Supérieure, Paris, France
| | - Pascal Maire
- INSERM U1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- * E-mail:
| |
Collapse
|
124
|
Krueger AB, Drasin DJ, Lea WA, Patrick AN, Patnaik S, Backos DS, Matheson CJ, Hu X, Barnaeva E, Holliday MJ, Blevins MA, Robin TP, Eisenmesser EZ, Ferrer M, Simeonov A, Southall N, Reigan P, Marugan J, Ford HL, Zhao R. Allosteric inhibitors of the Eya2 phosphatase are selective and inhibit Eya2-mediated cell migration. J Biol Chem 2014; 289:16349-61. [PMID: 24755226 DOI: 10.1074/jbc.m114.566729] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Eya proteins are essential co-activators of the Six family of transcription factors and contain a unique tyrosine phosphatase domain belonging to the haloacid dehalogenase family of phosphatases. The phosphatase activity of Eya is important for the transcription of a subset of Six1-target genes, and also directs cells to the repair rather than apoptosis pathway upon DNA damage. Furthermore, Eya phosphatase activity has been shown to mediate transformation, invasion, migration, and metastasis of breast cancer cells, making it a potential new drug target for breast cancer. We have previously identified a class of N-arylidenebenzohydrazide compounds that specifically inhibit the Eya2 phosphatase. Herein, we demonstrate that these compounds are reversible inhibitors that selectively inhibit the phosphatase activity of Eya2, but not Eya3. Our mutagenesis results suggest that this class of compounds does not bind to the active site and the binding does not require the coordination with Mg(2+). Moreover, these compounds likely bind within a site on the opposite face of the active site, and function as allosteric inhibitors. We also demonstrate that this class of compounds inhibits Eya2 phosphatase-mediated cell migration, setting the foundation for these molecules to be developed into chemical probes for understanding the specific function of the Eya2 phosphatase and to serve as a prototype for the development of Eya2 phosphatase specific anti-cancer drugs.
Collapse
Affiliation(s)
- Aaron B Krueger
- From the Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - David J Drasin
- the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Wendy A Lea
- the National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Aaron N Patrick
- the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Samarjit Patnaik
- the National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Donald S Backos
- the Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado School of Pharmacy, Aurora, Colorado 80045
| | - Christopher J Matheson
- the Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado School of Pharmacy, Aurora, Colorado 80045
| | - Xin Hu
- the National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Elena Barnaeva
- the National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Michael J Holliday
- From the Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Melanie A Blevins
- From the Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Tyler P Robin
- the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Elan Z Eisenmesser
- From the Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Marc Ferrer
- the National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Anton Simeonov
- the National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Noel Southall
- the National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Philip Reigan
- the Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado School of Pharmacy, Aurora, Colorado 80045
| | - Juan Marugan
- the National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Heide L Ford
- the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045,
| | - Rui Zhao
- From the Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045,
| |
Collapse
|
125
|
Gene regulatory networks and transcriptional mechanisms that control myogenesis. Dev Cell 2014; 28:225-38. [PMID: 24525185 DOI: 10.1016/j.devcel.2013.12.020] [Citation(s) in RCA: 439] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 12/24/2013] [Accepted: 12/27/2013] [Indexed: 12/11/2022]
Abstract
We discuss the upstream regulators of myogenesis that lead to the activation of myogenic determination genes and subsequent differentiation, focusing on the mouse model. Key upstream genes, such as Pax3 and Pax7, Six1 and Six4, or Pitx2, participate in gene regulatory networks at different sites of skeletal muscle formation. MicroRNAs also intervene, with emerging evidence for the role of other noncoding RNAs. Myogenic determination and subsequent differentiation depend on members of the MyoD family. We discuss new insights into mechanisms underlying the transcriptional activity of these factors.
Collapse
|
126
|
Rossi G, Messina G. Comparative myogenesis in teleosts and mammals. Cell Mol Life Sci 2014; 71:3081-99. [PMID: 24664432 PMCID: PMC4111864 DOI: 10.1007/s00018-014-1604-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/17/2014] [Accepted: 03/06/2014] [Indexed: 01/02/2023]
Abstract
Skeletal myogenesis has been and is currently under extensive study in both mammals and teleosts, with the latter providing a good model for skeletal myogenesis because of their flexible and conserved genome. Parallel investigations of muscle studies using both these models have strongly accelerated the advances in the field. However, when transferring the knowledge from one model to the other, it is important to take into account both their similarities and differences. The main difficulties in comparing mammals and teleosts arise from their different temporal development. Conserved aspects can be seen for muscle developmental origin and segmentation, and for the presence of multiple myogenic waves. Among the divergences, many fish have an indeterminate growth capacity throughout their entire life span, which is absent in mammals, thus implying different post-natal growth mechanisms. This review covers the current state of the art on myogenesis, with a focus on the most conserved and divergent aspects between mammals and teleosts.
Collapse
Affiliation(s)
- Giuliana Rossi
- Department of Biosciences, University of Milan, 20133, Milan, Italy
| | | |
Collapse
|
127
|
O'Brien JH, Hernandez-Lagunas L, Artinger KB, Ford HL. MicroRNA-30a regulates zebrafish myogenesis through targeting the transcription factor Six1. J Cell Sci 2014; 127:2291-301. [PMID: 24634509 DOI: 10.1242/jcs.143677] [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: 12/17/2022] Open
Abstract
Precise spatiotemporal regulation of the SIX1 homeoprotein is required to coordinate vital tissue development, including myogenesis. Whereas SIX1 is downregulated in most tissues following embryogenesis, it is re-expressed in numerous cancers, including tumors derived from muscle progenitors. Despite crucial roles in development and disease, the upstream regulation of SIX1 expression has remained elusive. Here, we identify the first direct mechanism for Six1 regulation in embryogenesis, through microRNA30a (miR30a)-mediated repression. In zebrafish somites, we show that miR30a and six1a and six1b (hereafter six1a/b) are expressed in an inverse temporal pattern. Overexpression of miR30a leads to a reduction in six1a/b levels, and results in increased apoptosis and altered somite morphology, which phenocopies six1a/b knockdown. Conversely, miR30a inhibition leads to increased Six1 expression and abnormal somite morphology, revealing a role for endogenous miR30a as a muscle-specific miRNA (myomiR). Importantly, restoration of six1a in miR30a-overexpressing embryos restores proper myogenesis. These data demonstrate a new role for miR30a at a key node in the myogenic regulatory gene network through controlling Six1 expression.
Collapse
Affiliation(s)
- Jenean H O'Brien
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Laura Hernandez-Lagunas
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kristin Bruk Artinger
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Heide L Ford
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
128
|
Saint-Jeannet JP, Moody SA. Establishing the pre-placodal region and breaking it into placodes with distinct identities. Dev Biol 2014; 389:13-27. [PMID: 24576539 DOI: 10.1016/j.ydbio.2014.02.011] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 11/17/2022]
Abstract
Specialized sensory organs in the vertebrate head originate from thickenings in the embryonic ectoderm called cranial sensory placodes. These placodes, as well as the neural crest, arise from a zone of ectoderm that borders the neural plate. This zone separates into a precursor field for the neural crest that lies adjacent to the neural plate, and a precursor field for the placodes, called the pre-placodal region (PPR), that lies lateral to the neural crest. The neural crest domain and the PPR are established in response to signaling events mediated by BMPs, FGFs and Wnts, which differentially activate transcription factors in these territories. In the PPR, members of the Six and Eya families, act in part to repress neural crest specific transcription factors, thus solidifying a placode developmental program. Subsequently, in response to environmental cues the PPR is further subdivided into placodal territories with distinct characteristics, each expressing a specific repertoire of transcription factors that provide the necessary information for their progression to mature sensory organs. In this review we summarize recent advances in the characterization of the signaling molecules and transcriptional effectors that regulate PPR specification and its subdivision into placodal domains with distinct identities.
Collapse
Affiliation(s)
- Jean-Pierre Saint-Jeannet
- Department of Basic Science and Craniofacial Biology, New York University, College of Dentistry, 345 East 24th Street, New York City, NY 10010, USA.
| | - Sally A Moody
- Department of Anatomy and Regenerative Biology, The George Washington University, School of Medicine and Health Sciences, 2300 I (eye) Street, NW, Washington, DC 20037, USA.
| |
Collapse
|
129
|
Wu W, Ren Z, Li P, Yu D, Chen J, Huang R, Liu H. Six1: A critical transcription factor in tumorigenesis. Int J Cancer 2014; 136:1245-53. [DOI: 10.1002/ijc.28755] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/15/2014] [Accepted: 01/20/2014] [Indexed: 12/16/2022]
Affiliation(s)
- Wangjun Wu
- Department of Animal Genetics; Breeding and Reproduction; College of Animal Science and Technology; Nanjing Agricultural University; Nanjing China
- Huaian Academy of Nanjing Agricultural University; Huaian Jiangsu China
| | - Zhuqing Ren
- Key Laboratory of Swine Genetics and Breeding; Ministry of Agriculture; Key Lab of Agriculture Animal Genetics; Breeding and Reproduction; Ministry of Education; College of Animal Science; Huazhong Agricultural University; Wuhan Hubei China
| | - Pinghua Li
- Department of Animal Genetics; Breeding and Reproduction; College of Animal Science and Technology; Nanjing Agricultural University; Nanjing China
| | - Debing Yu
- Department of Animal Genetics; Breeding and Reproduction; College of Animal Science and Technology; Nanjing Agricultural University; Nanjing China
| | - Jie Chen
- Department of Animal Genetics; Breeding and Reproduction; College of Animal Science and Technology; Nanjing Agricultural University; Nanjing China
| | - Ruihua Huang
- Department of Animal Genetics; Breeding and Reproduction; College of Animal Science and Technology; Nanjing Agricultural University; Nanjing China
| | - Honglin Liu
- Department of Animal Genetics; Breeding and Reproduction; College of Animal Science and Technology; Nanjing Agricultural University; Nanjing China
| |
Collapse
|
130
|
Hetzler KL, Collins BC, Shanely RA, Sue H, Kostek MC. The homoeobox gene SIX1 alters myosin heavy chain isoform expression in mouse skeletal muscle. Acta Physiol (Oxf) 2014; 210:415-28. [PMID: 24102895 DOI: 10.1111/apha.12168] [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: 04/25/2013] [Revised: 08/21/2013] [Accepted: 09/10/2013] [Indexed: 01/06/2023]
Abstract
AIM Six1 is necessary for the genesis of several tissues, but in adults, it is expressed primarily in skeletal muscle where its function is unclear. Overexpression of Six1 with a cofactor in skeletal muscle causes slow-to-fast fibre-type transition. We sought to characterize the effects of a physiologically relevant Six1 knockdown. METHODS The tibialis anterior (TA) muscles of C57BL/6 mice were electroporated with Six1 knockdown vector (siRNA) or empty vector. Muscles were collected at 2 or 14 days after transfection for Six1 and myosin heavy chain (MHC) expression analysis. C2C12 mouse myoblasts were grown in standard conditions. Cells were cotransfected with MHC promoter vectors and Six1 expression vectors. Cells were harvested after 4 days of differentiation. RESULTS In vivo, the Six1 siRNA caused a decreased expression of Six1,1.8-fold (±0.1, P < 0.05). With decreased Six1, MHC IIB expression decreased 2.7-fold (±0.7, P = 0.04). Proportion of muscle fibres expressing MHC IIB decreased (45.3 ± 4.8% vs. 65.1 ± 7.3% in control group, P = 0.04), and total area expressing MHC IIB decreased with decreased Six1 (59.6 ± 4.3% vs. 75.2 ± 5.4% in control group, P < 0.05). Decreased Six1 increased MHC IIA expression 1.9-fold (±0.3, P = 0.04). In vitro, Six1 overexpression increased promoter activation of MHC IIB 2.9-fold (±0.3, P < 0.01). Six1 knockdown repressed MHC IIB promoter 2.9-fold (±0.1, P < 0.05) and MHC IIX 3.7-fold (±0.08, P < 0.01). CONCLUSION Six1 knockdown caused a fast-to-slow shift in MHC isoform, and this was confirmed by promoter activity of MHC genes. Six1 may ultimately control the contractile and metabolic properties that define muscle fibre phenotype.
Collapse
Affiliation(s)
- K. L. Hetzler
- Department of Exercise Science; University of South Carolina; Columbia SC USA
| | - B. C. Collins
- Department of Exercise Science; University of South Carolina; Columbia SC USA
| | - R. A. Shanely
- Appalachian State University-North Carolina Research Campus Human Performance Laboratory; Appalachian State University; Kannapolis NC USA
| | - H. Sue
- Department of Exercise Science; University of South Carolina; Columbia SC USA
| | - M. C. Kostek
- Department of Exercise Science; University of South Carolina; Columbia SC USA
- Department of Physical Therapy; Duquesne University; Pittsburgh PA USA
| |
Collapse
|
131
|
Novel polymorphisms of SIX4 gene and their association with body measurement traits in Qinchuan cattle. Gene 2014; 539:107-10. [PMID: 24462757 DOI: 10.1016/j.gene.2014.01.042] [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] [Received: 10/11/2013] [Revised: 01/03/2014] [Accepted: 01/13/2014] [Indexed: 11/22/2022]
Abstract
Sine oculis homeobox homolog 4 (SIX4) gene belongs to the sine oculis/SIX gene family, which includes six members in vertebrates. SIX4 gene plays a crucial role in skeletal myogenesis, and its genetic variations or deficiency may cause hypopituitarism, suggesting that SIX4 gene is a potential candidate gene affecting body measurement traits (BMTs) in animals. Herein, the objectives of this study were to identify genetic polymorphisms of bovine SIX4 gene and to analyze potential association between single nucleotide polymorphisms (SNPs) and body measurement traits in Qinchuan cattle. In the present study, we investigated polymorphisms of SIX4 gene in 426 Qinchuan cattle using DNA sequencing and polymerase chain reaction-restriction fragment length polymorphisms. Three novel SNPs were identified within bovine SIX4 gene. Associations between body measurement traits and SIX4 gene polymorphisms were investigated, and significant statistical associations were found between polymorphisms of these three SNPs and body measurement traits (P<0.05). Hence, based on results obtained from this study, we conjectured that SIX4 gene may have potential effects on body measurement traits in Qinchuan cattle population and could be used for marker-assisted selection.
Collapse
|
132
|
Tan WJ, Thike AA, Bay BH, Tan PH. Immunohistochemical expression of homeoproteins Six1 and Pax3 in breast phyllodes tumours correlates with histological grade and clinical outcome. Histopathology 2014; 64:807-17. [DOI: 10.1111/his.12329] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 11/16/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Wai Jin Tan
- Department of Pathology; Singapore General Hospital; Singapore
- Department of Anatomy; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | - Aye Aye Thike
- Department of Pathology; Singapore General Hospital; Singapore
| | - Boon Huat Bay
- Department of Anatomy; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | - Puay Hoon Tan
- Department of Pathology; Singapore General Hospital; Singapore
- Department of Anatomy; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| |
Collapse
|
133
|
Abstract
Since the seminal discovery of the cell-fate regulator Myod, studies in skeletal myogenesis have inspired the search for cell-fate regulators of similar potential in other tissues and organs. It was perplexing that a similar transcription factor for other tissues was not found; however, it was later discovered that combinations of molecular regulators can divert somatic cell fates to other cell types. With the new era of reprogramming to induce pluripotent cells, the myogenesis paradigm can now be viewed under a different light. Here, we provide a short historical perspective and focus on how the regulation of skeletal myogenesis occurs distinctly in different scenarios and anatomical locations. In addition, some interesting features of this tissue underscore the importance of reconsidering the simple-minded view that a single stem cell population emerges after gastrulation to assure tissuegenesis. Notably, a self-renewing long-term Pax7+ myogenic stem cell population emerges during development only after a first wave of terminal differentiation occurs to establish a tissue anlagen in the mouse. How the future stem cell population is selected in this unusual scenario will be discussed. Recently, a wealth of information has emerged from epigenetic and genome-wide studies in myogenic cells. Although key transcription factors such as Pax3, Pax7, and Myod regulate only a small subset of genes, in some cases their genomic distribution and binding are considerably more promiscuous. This apparent nonspecificity can be reconciled in part by the permissivity of the cell for myogenic commitment, and also by new roles for some of these regulators as pioneer transcription factors acting on chromatin state.
Collapse
Affiliation(s)
- Glenda Comai
- Stem Cells and Development, CNRS URA 2578, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, CNRS URA 2578, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France.
| |
Collapse
|
134
|
Borchin B, Chen J, Barberi T. Derivation and FACS-mediated purification of PAX3+/PAX7+ skeletal muscle precursors from human pluripotent stem cells. Stem Cell Reports 2013; 1:620-31. [PMID: 24371814 PMCID: PMC3871395 DOI: 10.1016/j.stemcr.2013.10.007] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 10/15/2013] [Accepted: 10/16/2013] [Indexed: 12/14/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) constitute a promising resource for use in cell-based therapies and a valuable in vitro model for studying early human development and disease. Despite significant advancements in the derivation of specific fates from hPSCs, the generation of skeletal muscle remains challenging and is mostly dependent on transgene expression. Here, we describe a method based on the use of a small-molecule GSK3β inhibitor to derive skeletal muscle from several hPSC lines. We show that early GSK3β inhibition is sufficient to create the conditions necessary for highly effective derivation of muscle cells. Moreover, we developed a strategy for stringent fluorescence-activated cell sorting-based purification of emerging PAX3+/PAX7+ muscle precursors that are able to differentiate in postsort cultures into mature myocytes. This transgene-free, efficient protocol provides an essential tool for producing myogenic cells for in vivo preclinical studies, in vitro screenings, and disease modeling.
Collapse
Affiliation(s)
- Bianca Borchin
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Joseph Chen
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Tiziano Barberi
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
135
|
Figeac N, Serralbo O, Marcelle C, Zammit PS. ErbB3 binding protein-1 (Ebp1) controls proliferation and myogenic differentiation of muscle stem cells. Dev Biol 2013; 386:135-51. [PMID: 24275324 DOI: 10.1016/j.ydbio.2013.11.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 10/21/2013] [Accepted: 11/15/2013] [Indexed: 12/16/2022]
Abstract
Satellite cells are resident stem cells of skeletal muscle, supplying myoblasts for post-natal muscle growth, hypertrophy and repair. Many regulatory networks control satellite cell function, which includes EGF signalling via the ErbB family of receptors. Here we investigated the role of ErbB3 binding protein-1 (Ebp1) in regulation of myogenic stem cell proliferation and differentiation. Ebp1 is a well-conserved DNA/RNA binding protein that is implicated in cell growth, apoptosis and differentiation in many cell types. Of the two main Ebp1 isoforms, only p48 was expressed in satellite cells and C2C12 myoblasts. Although not present in quiescent satellite cells, p48 was strongly induced during activation, remaining at high levels during proliferation and differentiation. While retroviral-mediated over-expression of Ebp1 had only minor effects, siRNA-mediated Ebp1 knockdown inhibited both proliferation and differentiation of satellite cells and C2C12 myoblasts, with a clear failure of myotube formation. Ebp1-knockdown significantly reduced ErbB3 receptor levels, yet over-expression of ErbB3 in Ebp1 knockdown cells did not rescue differentiation. Ebp1 was also expressed by muscle cells during developmental myogenesis in mouse. Since Ebp1 is well-conserved between mouse and chick, we switched to chick to examine its role in muscle formation. In chick embryo, Ebp1 was expressed in the dermomyotome, and myogenic differentiation of muscle progenitors was inhibited by specific Ebp1 down-regulation using shRNA electroporation. These observations demonstrate a conserved function of Ebp1 in the regulation of embryonic muscle progenitors and adult muscle stem cells, which likely operates independently of ErbB3 signaling.
Collapse
Affiliation(s)
- Nicolas Figeac
- King's College London, Randall Division of Cell & Molecular Biophysics, New Hunt's House, Guy's Campus, London SE1 1UL, England, UK
| | - Olivier Serralbo
- EMBL Australia, Australian Regenerative Medicine Institute (ARMI), Monash University, Building 75, Clayton, Victoria 3800, Australia
| | - Christophe Marcelle
- EMBL Australia, Australian Regenerative Medicine Institute (ARMI), Monash University, Building 75, Clayton, Victoria 3800, Australia
| | - Peter S Zammit
- King's College London, Randall Division of Cell & Molecular Biophysics, New Hunt's House, Guy's Campus, London SE1 1UL, England, UK.
| |
Collapse
|
136
|
Core promoter analysis of porcine Six1 gene and its regulation of the promoter activity by CpG methylation. Gene 2013; 529:238-44. [DOI: 10.1016/j.gene.2013.07.102] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Revised: 07/23/2013] [Accepted: 07/27/2013] [Indexed: 11/22/2022]
|
137
|
Abstract
Mammalian skeletal muscles are derived from mesoderm segments flanking the embryonic midline. Upon receiving inductive cues from the adjacent neural tube, lateral plate mesoderm, and surface ectoderm, muscle precursors start to delaminate, migrate to their final destinations and proliferate. Muscle precursor cells become committed to the myogenic fate, become differentiated muscle cells, and fuse to form myofibers. Myofibers then fuse together to form the muscle groups. Muscle precursor cells have the ability to proliferate, and differentiate during development, while a subset remains capable of regeneration and repair of local injuries in adulthood. When the process of muscle development is perturbed such as in muscular dystrophies and injuries, ways to intervene and allow for proper muscle development or repair are the focus of regenerative medicine. Thus, understanding the developmental program of muscle at the genetic, cellular, and molecular levels has become a major focus of skeletal muscle regeneration research in the last few years.
Collapse
|
138
|
Jefferson WN, Chevalier DM, Phelps JY, Cantor AM, Padilla-Banks E, Newbold RR, Archer TK, Kinyamu HK, Williams CJ. Persistently altered epigenetic marks in the mouse uterus after neonatal estrogen exposure. Mol Endocrinol 2013; 27:1666-77. [PMID: 24002655 DOI: 10.1210/me.2013-1211] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Neonatal exposure to diethylstilbestrol (DES) causes permanent alterations in female reproductive tract gene expression, infertility, and uterine cancer in mice. To determine whether epigenetic mechanisms could explain these phenotypes, we first tested whether DES altered uterine expression of chromatin-modifying proteins. DES treatment significantly reduced expression of methylcytosine dioxygenase TET oncogene family, member 1 (TET1) on postnatal day 5; this decrease was correlated with a subtle decrease in DNA 5-hydroxymethylcytosine in adults. There were also significant reductions in histone methyltransferase enhancer of zeste homolog 2 (EZH2), histone lysine acetyltransferase 2A (KAT2A), and histone deacetylases HDAC1, HDAC2, and HDAC3. Uterine chromatin immunoprecipitation was used to analyze the locus-specific association of modified histones with 2 genes, lactoferrin (Ltf) and sine oculis homeobox 1 (Six1), which are permanently upregulated in adults after neonatal DES treatment. Three histone modifications associated with active transcription, histone H3 lysine 9 acetylation (H3K9ac), H3 lysine 4 trimethylation (H3K4me3), and H4 lysine 5 acetylation (H4K5ac) were enriched at specific Ltf promoter regions after DES treatment, but this enrichment was not maintained in adults. H3K9ac, H4K5ac, and H3K4me3 were enriched at Six1 exon 1 immediately after neonatal DES treatment. As adults, DES-treated mice had greater differences in H4K5ac and H3K4me3 occupancy at Six1 exon 1 and new differences in these histone marks at an upstream region. These findings indicate that neonatal DES exposure temporarily alters expression of multiple chromatin-modifying proteins and persistently alters epigenetic marks in the adult uterus at the Six1 locus, suggesting a mechanism for developmental exposures leading to altered reproductive function and increased cancer risk.
Collapse
Affiliation(s)
- Wendy N Jefferson
- National Institute of Environmental Health Sciences, National Institutes of Health, US Department of Health and Human Services, PO Box 12233, MD E4-05, Research Triangle Park, North Carolina 27709.
| | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Fujimoto Y, Tanaka S, Yamaguchi Y, Kobayashi H, Kuroki S, Tachibana M, Shinomura M, Kanai Y, Morohashi KI, Kawakami K, Nishinakamura R. Homeoproteins Six1 and Six4 Regulate Male Sex Determination and Mouse Gonadal Development. Dev Cell 2013; 26:416-30. [DOI: 10.1016/j.devcel.2013.06.018] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 06/11/2013] [Accepted: 06/19/2013] [Indexed: 01/11/2023]
|
140
|
Weimer K, Theobald J, Campbell KS, Esser KA, DiMario JX. Genome-wide expression analysis and EMX2 gene expression in embryonic myoblasts committed to diverse skeletal muscle fiber type fates. Dev Dyn 2013; 242:1001-20. [PMID: 23703830 PMCID: PMC3763492 DOI: 10.1002/dvdy.23988] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 04/24/2013] [Accepted: 05/07/2013] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Primary skeletal muscle fibers form during embryonic development and are characterized as fast or slow fibers based on contractile protein gene expression. Different avian primary muscle fiber types arise from myoblast lineages committed to formation of diverse fiber types. To understand the basis of embryonic muscle fiber type diversity and the distinct myoblast lineages that generate this diversity, gene expression analyses were conducted on differentiated muscle fiber types and their respective myoblast precursor lineages. RESULTS Embryonic fast muscle fibers preferentially expressed 718 genes, and embryonic fast/slow muscle fibers differentially expressed 799 genes. Fast and fast/slow myoblast lineages displayed appreciable diversity in their gene expression profiles, indicating diversity of precursor myoblasts. Several genes, including the transcriptional regulator EMX2, were differentially expressed in both fast/slow myoblasts and muscle fibers vs. fast myoblasts and muscle fibers. EMX2 was localized to nuclei of fast/slow myoblasts and muscle fibers and was not detected in fast lineage cells. Furthermore, EMX2 overexpression and knockdown studies indicated that EMX2 is a positive transcriptional regulator of the slow myosin heavy chain 2 (MyHC2) gene promoter activity in fast/slow muscle fibers. CONCLUSIONS These results indicate the presence of distinct molecular signatures that characterize diverse embryonic myoblast lineages before differentiation.
Collapse
Affiliation(s)
- Kristina Weimer
- Rosalind Franklin University of Medicine and Science, School of Graduate and Postdoctoral Studies, Department of Cell Biology and Anatomy, 3333 Green Bay Road, North Chicago, IL 60064
| | - Jillian Theobald
- Rosalind Franklin University of Medicine and Science, School of Graduate and Postdoctoral Studies, Department of Cell Biology and Anatomy, 3333 Green Bay Road, North Chicago, IL 60064
| | - Kenneth S. Campbell
- Center for Muscle Biology, Department of Physiology, University of Kentucky, Lexington, KY 40536
| | - Karyn A. Esser
- Center for Muscle Biology, Department of Physiology, University of Kentucky, Lexington, KY 40536
| | - Joseph X. DiMario
- Rosalind Franklin University of Medicine and Science, School of Graduate and Postdoctoral Studies, Department of Cell Biology and Anatomy, 3333 Green Bay Road, North Chicago, IL 60064
| |
Collapse
|
141
|
Six1 regulates MyoD expression in adult muscle progenitor cells. PLoS One 2013; 8:e67762. [PMID: 23840772 PMCID: PMC3695946 DOI: 10.1371/journal.pone.0067762] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 05/22/2013] [Indexed: 12/21/2022] Open
Abstract
Quiescent satellite cells are myogenic progenitors that enable regeneration of skeletal muscle. One of the early events of satellite cell activation following myotrauma is the induction of the myogenic regulatory factor MyoD, which eventually induces terminal differentiation and muscle function gene expression. The purpose of this study was to elucidate the mechanism by which MyoD is induced during activation of satellite cells in mouse muscle undergoing regeneration. We show that Six1, a transcription factor essential for embryonic myogenesis, also regulates MyoD expression in muscle progenitor cells. Six1 knock-down by RNA interference leads to decreased expression of MyoD in myoblasts. Chromatin immunoprecipitation assays reveal that Six1 binds the Core Enhancer Region of MyoD. Further, transcriptional reporter assays demonstrate that Core Enhancer Region reporter gene activity in myoblasts and in regenerating muscle depends on the expression of Six1 and on Six1 binding sites. Finally, we provide evidence indicating that Six1 is required for the proper chromatin structure at the Core Enhancer Region, as well as for MyoD binding at its own enhancer. Together, our results reveal that MyoD expression in satellite cells depends on Six1, supporting the idea that Six1 plays an important role in adult myogenesis, in addition to its role in embryonic muscle formation.
Collapse
|
142
|
Xu PX. The EYA-SO/SIX complex in development and disease. Pediatr Nephrol 2013; 28:843-54. [PMID: 22806561 PMCID: PMC6592036 DOI: 10.1007/s00467-012-2246-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 06/10/2012] [Accepted: 06/12/2012] [Indexed: 10/28/2022]
Abstract
Eyes absent (EYA) and Sine oculis (SO/SIX) proteins function as transcriptional activation complexes and play essential roles in organogenesis during embryonic development in regulating cell proliferation and survival and coordination of particular differentiation programs. Mutations of the Eya and So/Six genes cause profound developmental defects in organisms as diverse as flies, frogs, fish, mice, and humans. EYA proteins also possess an intrinsic phosphatase activity, which is essential for normal development. Here, we review crucial roles of EYA and SO/SIX in development and disease in mice and humans.
Collapse
Affiliation(s)
- Pin-Xian Xu
- Department of Genetics and Genomic Sciences and Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|
143
|
Six homeoproteins directly activate Myod expression in the gene regulatory networks that control early myogenesis. PLoS Genet 2013; 9:e1003425. [PMID: 23637613 PMCID: PMC3636133 DOI: 10.1371/journal.pgen.1003425] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 02/14/2013] [Indexed: 01/16/2023] Open
Abstract
In mammals, several genetic pathways have been characterized that govern engagement of multipotent embryonic progenitors into the myogenic program through the control of the key myogenic regulatory gene Myod. Here we demonstrate the involvement of Six homeoproteins. We first targeted into a Pax3 allele a sequence encoding a negative form of Six4 that binds DNA but cannot interact with essential Eya co-factors. The resulting embryos present hypoplasic skeletal muscles and impaired Myod activation in the trunk in the absence of Myf5/Mrf4. At the axial level, we further show that Myod is still expressed in compound Six1/Six4:Pax3 but not in Six1/Six4:Myf5 triple mutant embryos, demonstrating that Six1/4 participates in the Pax3-Myod genetic pathway. Myod expression and head myogenesis is preserved in Six1/Six4:Myf5 triple mutant embryos, illustrating that upstream regulators of Myod in different embryonic territories are distinct. We show that Myod regulatory regions are directly controlled by Six proteins and that, in the absence of Six1 and Six4, Six2 can compensate. The onset of skeletal muscle formation is controlled by complex gene regulatory networks. By manipulation of these genetic pathways in the mouse embryo, we have examined the interplay between genes encoding the transcriptional regulator Pax3; the major myogenic determination proteins Myf5, Mrf4, and Myod; as well as genes encoding homeodomain proteins Six1 and Six4. In the absence of Myf5 and Six1/4, Myod expression is compromised. We demonstrate that key regulatory elements of the Myod gene are directly targeted by Six factors, including Six2, which is unexpectedly upregulated in the absence of Six1 and Six4. This work therefore reveals new aspects of the gene regulatory networks that control myogenesis.
Collapse
|
144
|
Wu W, Ren Z, Zhang L, Liu Y, Li H, Xiong Y. Overexpression of Six1 gene suppresses proliferation and enhances expression of fast-type muscle genes in C2C12 myoblasts. Mol Cell Biochem 2013; 380:23-32. [PMID: 23613228 DOI: 10.1007/s11010-013-1653-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 04/12/2013] [Indexed: 12/15/2022]
Abstract
Sine oculis homeobox 1 (Six1) homeodomain transcription factor is implicated in the genesis of muscle fiber type diversity, but its regulatory mechanisms on the formation of muscle fiber type are still poorly understood. To elucidate the biological roles of Six1 gene in muscle fiber formation, we established C2C12 cell line overexpressing Six1 and determined the effects of forced Six1 expression on muscle-specific genes expression, cell proliferation, and cell cycles. Our results indicated that Six1 overexpression could significantly promote the expression of fast-type muscle genes Atp2a1, Srl, and Mylpf. Furthermore, Six1 overexpressing C2C12 cells displayed a relative lower proliferative potential, and cell cycle analysis showed that Six1 exerted its role in cell cycle primarily through the regulation of G1/S and G2/M phases. In conclusion, Six1 plays an essential role in modulation of the fast-twitch muscle fiber phenotype through up-regulating fast-type muscle genes expression, and it could suppress the proliferation of muscle cells.
Collapse
Affiliation(s)
- Wangjun Wu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | | | | | | | | | | |
Collapse
|
145
|
Direct molecular regulation of the myogenic determination gene Myf5 by Pax3, with modulation by Six1/4 factors, is exemplified by the −111kb-Myf5 enhancer. Dev Biol 2013; 376:236-44. [DOI: 10.1016/j.ydbio.2013.01.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 01/11/2013] [Accepted: 01/25/2013] [Indexed: 11/20/2022]
|
146
|
Singh K, Dilworth FJ. Differential modulation of cell cycle progression distinguishes members of the myogenic regulatory factor family of transcription factors. FEBS J 2013; 280:3991-4003. [DOI: 10.1111/febs.12188] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 02/01/2013] [Accepted: 02/05/2013] [Indexed: 12/29/2022]
Affiliation(s)
- Kulwant Singh
- Sprott Center for Stem Cell Research; Ottawa Hospital Research Institute; ON; Canada
| | | |
Collapse
|
147
|
Nord H, Nygård Skalman L, von Hofsten J. Six1 regulates proliferation of Pax7-positive muscle progenitors in zebrafish. J Cell Sci 2013; 126:1868-80. [PMID: 23444384 DOI: 10.1242/jcs.119917] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In the embryonic zebrafish, skeletal muscle fibres are formed from muscle progenitors in the paraxial mesoderm. The embryonic myotome is mostly constituted of fast-twitch-specific fibres, which are formed from a fast-specific progenitor cell pool. The most lateral fraction of the fast domain in the myotome of zebrafish embryos derives from the Pax7-positive dermomyotome-like cells. In this study, we show that two genes, belonging to the sine oculus class 1 (six1) genes (six1a and six1b), are both essential for the regulation of Pax7(+) cell proliferation and, consequently, in their differentiation during the establishment of the zebrafish dermomyotome. In both six1a and six1b morphant embryos, Pax7(+) cells are initially formed but fail to proliferate, as detected by reduced levels of the proliferation marker phosphohistone3 and reduced brdU incorporation. In congruence, overexpression of six1a or six1b leads to increased Pax7(+) cell number and reduced or alternatively delayed fibre cell differentiation. Bone morphogenetic protein signalling has previously been suggested to inhibit differentiation of Pax7(+) cells in the dermomyotome. Here we show that the remaining Pax7(+) cells in six1a and six1b morphant embryos also have significantly reduced pSmad1/5/8 levels and propose that this leads to a reduced proliferative activity, which may result in a premature differentiation of Pax7(+) cells in the zebrafish dermomyotome. In summary, we show a mechanism for Six1a and Six1b in establishing the Pax7(+) cell derived part of the fast muscle and suggest new important roles for Six1 in the regulation of the Pax7(+) muscle cell population through pSmad1/5/8 signalling.
Collapse
Affiliation(s)
- Hanna Nord
- Umeå Center for Molecular Medicine, UCMM, Umeå University, 901 87 Umeå, Sweden
| | | | | |
Collapse
|
148
|
Le Grand F, Grifone R, Mourikis P, Houbron C, Gigaud C, Pujol J, Maillet M, Pagès G, Rudnicki M, Tajbakhsh S, Maire P. Six1 regulates stem cell repair potential and self-renewal during skeletal muscle regeneration. ACTA ACUST UNITED AC 2013; 198:815-32. [PMID: 22945933 PMCID: PMC3432771 DOI: 10.1083/jcb.201201050] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Six1 in satellite cells is important for muscle regeneration and homeostasis of the stem cell niche by regulating MyoD, Myogenin, and Dusp6-ERK signaling. Satellite cells (SCs) are stem cells that mediate skeletal muscle growth and regeneration. Here, we observe that adult quiescent SCs and their activated descendants expressed the homeodomain transcription factor Six1. Genetic disruption of Six1 specifically in adult SCs impaired myogenic cell differentiation, impaired myofiber repair during regeneration, and perturbed homeostasis of the stem cell niche, as indicated by an increase in SC self-renewal. Six1 regulated the expression of the myogenic regulatory factors MyoD and Myogenin, but not Myf5, which suggests that Six1 acts on divergent genetic networks in the embryo and in the adult. Moreover, we demonstrate that Six1 regulates the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway during regeneration via direct control of Dusp6 transcription. Muscles lacking Dusp6 were able to regenerate properly but showed a marked increase in SC number after regeneration. We conclude that Six1 homeoproteins act as a rheostat system to ensure proper regeneration of the tissue and replenishment of the stem cell pool during the events that follow skeletal muscle trauma.
Collapse
Affiliation(s)
- Fabien Le Grand
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris 75014, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Origin of the Turtle Body Plan: The Folding Theory to Illustrate Turtle-Specific Developmental Repatterning. VERTEBRATE PALEOBIOLOGY AND PALEOANTHROPOLOGY 2013. [DOI: 10.1007/978-94-007-4309-0_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
|
150
|
Abstract
Craniofacial development requires an exquisitely timed and positioned cross-talk between the embryonic cephalic epithelia and mesenchyme. This cross-talk underlies the precise translation of patterning processes and information into distinct, appropriate skeletal morphologies. The molecular and cellular dialogue includes communication via secreted signaling molecules, including Fgf8, and effectors of their interpretation. Herein, we use genetic attenuation of Fgf8 in mice and perform gain-of-function mouse-chick chimeric experiments to demonstrate that significant character states of the frontonasal and optic skeletons are dependent on Fgf8. Notably, we show that the normal orientation and polarity of the nasal capsules and their developing primordia are dependent on Fgf8. We further demonstrate that Fgf8 is required for midfacial integration, and provide evidence for a role for Fgf8 in optic capsular development. Taken together, our data highlight Fgf8 signaling in craniofacial development as a plausible target for evolutionary selective pressures.
Collapse
|