51
|
Clément O, Hemming IA, Gladwyn-Ng IE, Qu Z, Li SS, Piper M, Heng JIT. Rp58 and p27 kip1 coordinate cell cycle exit and neuronal migration within the embryonic mouse cerebral cortex. Neural Dev 2017; 12:8. [PMID: 28506232 PMCID: PMC5433244 DOI: 10.1186/s13064-017-0084-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/01/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND During the development of the mammalian cerebral cortex, newborn postmitotic projection neurons are born from local neural stem cells and must undergo radial migration so as to position themselves appropriately to form functional neural circuits. The zinc finger transcriptional repressor Rp58 (also known as Znf238 or Zbtb18) is critical for coordinating corticogenesis, but its underlying molecular mechanism remains to be better characterised. FINDINGS Here, we demonstrate that the co-expression of Rp58 and the cyclin dependent kinase inhibitor (CDKI) p27kip1 is important for E14.5-born cortical neurons to coordinate cell cycle exit and initiate their radial migration. Notably, we find that the impaired radial positioning of Rp58-deficient cortical neurons within the embryonic (E17.5) mouse cortex, as well as their multipolar to bipolar transition from the intermediate zone to the cortical plate can be restored by forced expression of p27kip1 in concert with suppression of Rnd2, a downstream target gene of Rp58. Furthermore, the restorative effects of p27kip1 and Rnd2 abrogation are reminiscent of suppressing RhoA signalling in Rp58-deficient cells. CONCLUSIONS Our findings demonstrate functional interplay between a transcriptional regulator and a CDKI to mediate neuroprogenitor cell cycle exit, as well as to promote radial migration through a molecular mechanism consistent with suppression of RhoA signalling.
Collapse
Affiliation(s)
- Olivier Clément
- The Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia
- The Centre for Medical Research, University of Western Australia, Perth, WA 6009 Australia
| | - Isabel Anne Hemming
- The Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia
- The Centre for Medical Research, University of Western Australia, Perth, WA 6009 Australia
| | - Ivan Enghian Gladwyn-Ng
- The Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia
- The Centre for Medical Research, University of Western Australia, Perth, WA 6009 Australia
| | - Zhengdong Qu
- EMBL Australia, The Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800 Australia
| | - Shan Shan Li
- EMBL Australia, The Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800 Australia
| | - Michael Piper
- The School of Biomedical Sciences, University of Queensland, Brisbane, 4072 Australia
- Queensland Brain Institute, University of Queensland, Brisbane, 4072 Australia
| | - Julian Ik-Tsen Heng
- The Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia
- The Centre for Medical Research, University of Western Australia, Perth, WA 6009 Australia
- EMBL Australia, The Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800 Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, 6845 Australia
| |
Collapse
|
52
|
Yokoyama S, Furukawa S, Kitada S, Mori M, Saito T, Kawakami K, Belmonte JCI, Kawakami Y, Ito Y, Sato T, Asahara H. Analysis of transcription factors expressed at the anterior mouse limb bud. PLoS One 2017; 12:e0175673. [PMID: 28467430 PMCID: PMC5415108 DOI: 10.1371/journal.pone.0175673] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/29/2017] [Indexed: 12/21/2022] Open
Abstract
Limb bud patterning, outgrowth, and differentiation are precisely regulated in a spatio-temporal manner through integrated networks of transcription factors, signaling molecules, and downstream genes. However, the exact mechanisms that orchestrate morphogenesis of the limb remain to be elucidated. Previously, we have established EMBRYS, a whole-mount in situ hybridization database of transcription factors. Based on the findings from EMBRYS, we focused our expression pattern analysis on a selection of transcription factor genes that exhibit spatially localized and temporally dynamic expression patterns with respect to the anterior-posterior axis in the E9.5–E11.5 limb bud. Among these genes, Irx3 showed a posteriorly expanded expression domain in Shh-/- limb buds and an anteriorly reduced expression domain in Gli3-/- limb buds, suggesting their importance in anterior-posterior patterning. To assess the stepwise EMBRYS-based screening system for anterior regulators, we generated Irx3 transgenic mice in which Irx3 was expressed in the entire limb mesenchyme under the Prrx1 regulatory element. The Irx3 gain-of-function model displayed complex phenotypes in the autopods, including digit loss, radial flexion, and fusion of the metacarpal bones, suggesting that Irx3 may contribute to the regulation of limb patterning, especially in the autopods. Our results demonstrate that gene expression analysis based on EMBRYS could contribute to the identification of genes that play a role in patterning of the limb mesenchyme.
Collapse
Affiliation(s)
- Shigetoshi Yokoyama
- Department of Systems Biomedicine, National Institute of Child Health and Development, Setagaya, Tokyo, Japan
| | - Soichi Furukawa
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| | - Shoya Kitada
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| | - Masaki Mori
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| | - Takeshi Saito
- Department of Systems Biomedicine, National Institute of Child Health and Development, Setagaya, Tokyo, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Shizuoka, Japan
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Yasuhiko Kawakami
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Yoshiaki Ito
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| | - Tempei Sato
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| | - Hiroshi Asahara
- Department of Systems Biomedicine, National Institute of Child Health and Development, Setagaya, Tokyo, Japan
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
- Department of Experimental Medicine, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
53
|
Abstract
CRISPR/Cas9 genome editing technology has taken the research world by storm since its use in eukaryotes was first proposed in 2012. Publications describing advances in technology and new applications have continued at an unrelenting pace since that time. In this Review, we discuss the application of CRISPR/Cas9 for creating gene mutations - the application that initiated the current avalanche of interest - and new developments that have largely answered initial concerns about its specificity and ability to introduce new gene sequences. We discuss the new, diverse and rapidly growing adaptations of the CRISPR/Cas9 technique that enable activation, repression, multiplexing and gene screening. These developments have enabled researchers to create sophisticated tools for dissecting the function and inter-relatedness of genes, as well as noncoding regions of the genome, and to identify gene networks and noncoding regions that promote disease or confer disease susceptibility. These approaches are beginning to be used to interrogate complex and multilayered biological systems and to produce complex animal models of disease. CRISPR/Cas9 technology has enabled the application of new therapeutic approaches to treating disease in animal models, some of which are beginning to be seen in the first human clinical trials. We discuss the direct application of these techniques to rheumatic diseases, which are currently limited but are sure to increase rapidly in the near future.
Collapse
Affiliation(s)
- Gary J Gibson
- Henry Ford Hospital, Bone and Joint Center, 6135 Woodward Avenue, Detroit, Michigan 48202, USA
| | - Maozhou Yang
- Henry Ford Hospital, Bone and Joint Center, 6135 Woodward Avenue, Detroit, Michigan 48202, USA
| |
Collapse
|
54
|
Identification of targets of tumor suppressor microRNA-34a using a reporter library system. Proc Natl Acad Sci U S A 2017; 114:3927-3932. [PMID: 28356515 DOI: 10.1073/pnas.1620019114] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
miRNAs play critical roles in various biological processes by targeting specific mRNAs. Current approaches to identifying miRNA targets are insufficient for elucidation of a miRNA regulatory network. Here, we created a cell-based screening system using a luciferase reporter library composed of 4,891 full-length cDNAs, each of which was integrated into the 3' UTR of a luciferase gene. Using this reporter library system, we conducted a screening for targets of miR-34a, a tumor-suppressor miRNA. We identified both previously characterized and previously uncharacterized targets. miR-34a overexpression in MDA-MB-231 breast cancer cells repressed the expression of these previously unrecognized targets. Among these targets, GFRA3 is crucial for MDA-MB-231 cell growth, and its expression correlated with the overall survival of patients with breast cancer. Furthermore, GFRA3 was found to be directly regulated by miR-34a via its coding region. These data show that this system is useful for elucidating miRNA functions and networks.
Collapse
|
55
|
Su Z, Zheng X, Zhang X, Wang Y, Zhu S, Lu F, Qu J, Hou L. Sox10 regulates skin melanocyte proliferation by activating the DNA replication licensing factor MCM5. J Dermatol Sci 2017; 85:216-225. [PMID: 27955842 DOI: 10.1016/j.jdermsci.2016.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/21/2016] [Accepted: 12/02/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND The control of cell proliferation is a fundamental aspect of tissue formation in development and regeneration. A cell type that illustrates this point particularly well is the neural crest-derived melanocyte, the pigment cell of vertebrates, as melanocytes can be followed easily during development and their pigment is directly visible in the integument of the adult. In mammals, melanocytes undergo physiological cycles of loss and proliferative regeneration during the hair cycle, and their proliferation is also critical during wound healing, repigmentation of depigmented lesions, and in melanoma formation and progression. Hence, a thorough analysis of the molecular parameters controlling melanocyte proliferation is crucial for our understanding of the physiology of this cell type both in health and disease. OBJECTIVE SOX10 is a critical regulator in melanocytes and melanoma cells, but its specific role in their proliferation is far from clear. In this study we analyze the role of SOX10 in regulating mammalian melanocyte proliferation in a mouse model. METHODS The role of SOX10 in melanoblast proliferation was analyzed in Sox10/+ mice by co-staining for melanocyte-specific markers and cell proliferation. In vitro, the role of SOX10 was studied by manipulating its levels using RNAi and analyzing the effects on DNA synthesis and cell growth and on gene expression at the RNA and protein levels. RESULTS Reduction of Sox10 gene dose led to a reduction in the number of melanoblasts. Knockdown of Sox10 in melanocytes led to inhibition of cell proliferation and a decrease in the expression of the minichromosome maintenance complex component 5 (MCM5). In fact, SOX10 directly activated MCM5 transcription by binding to conserved SOX10 consensus DNA sequences in the MCM5 promoter. Furthermore, the defect in cell proliferation could be rescued partially by overexpression of MCM5 in Sox10 knockdown melanocytes. CONCLUSION The results suggest that the SOX10-MCM5 axis plays an important role in controlling melanocyte proliferation. Our findings provide novel insights into the regulatory mechanisms of melanocyte proliferation and may have implications for our understanding of the roles of SOX10 and MCM5 in abnormal melanocyte proliferation disorders such as cutaneous melanoma.
Collapse
Affiliation(s)
- Zhongyuan Su
- Labratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325003, China; State Key Laboratory Cultivation Base and Key Laboratory of Vision Science of Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology, Wenzhou, 325003, China
| | - Xiaozi Zheng
- Labratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325003, China
| | - Xiaobo Zhang
- Labratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325003, China
| | - Yipin Wang
- Labratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325003, China
| | - Shanpu Zhu
- Labratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325003, China
| | - Fan Lu
- State Key Laboratory Cultivation Base and Key Laboratory of Vision Science of Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology, Wenzhou, 325003, China
| | - Jia Qu
- State Key Laboratory Cultivation Base and Key Laboratory of Vision Science of Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology, Wenzhou, 325003, China
| | - Ling Hou
- Labratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325003, China; State Key Laboratory Cultivation Base and Key Laboratory of Vision Science of Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology, Wenzhou, 325003, China.
| |
Collapse
|
56
|
Murphy KE, Shylo NA, Alexander KA, Churchill AJ, Copperman C, García-García MJ. The Transcriptional Repressive Activity of KRAB Zinc Finger Proteins Does Not Correlate with Their Ability to Recruit TRIM28. PLoS One 2016; 11:e0163555. [PMID: 27658112 PMCID: PMC5033580 DOI: 10.1371/journal.pone.0163555] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 08/23/2016] [Indexed: 12/27/2022] Open
Abstract
KRAB domain Zinc finger proteins are one of the most abundant families of transcriptional regulators in higher vertebrates. The prevailing view is that KRAB domain proteins function as potent transcriptional repressors by recruiting TRIM28 and promoting heterochromatin spreading. However, the extent to which all KRAB domain proteins are TRIM28-dependent transcriptional repressors is currently unclear. Our studies on mouse ZFP568 revealed that TRIM28 recruitment by KRAB domain proteins is not sufficient to warrant transcriptional repressive activity. By using luciferase reporter assays and yeast two-hybrid experiments, we tested the ability of ZFP568 and other mouse KRAB domain proteins to repress transcription and bind TRIM28. We found that some mouse KRAB domain proteins are poor transcriptional repressors despite their ability to recruit TRIM28, while others showed strong KRAB-dependent transcriptional repression, but no TRIM28 binding. Together, our results show that the transcriptional repressive activity of KRAB-ZNF proteins does not correlate with their ability to recruit TRIM28, and provide evidence that KRAB domains can regulate transcription in a TRIM28-independent fashion. Our findings challenge the current understanding of the molecular mechanisms used by KRAB domain proteins to control gene expression and highlight that a high percentage of KRAB domain proteins in the mouse genome differ from the consensus KRAB sequence at amino acid residues that are critical for TRIM28 binding and/or repressive activity.
Collapse
Affiliation(s)
- Kristin E. Murphy
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, United States of America
| | - Natalia A. Shylo
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, United States of America
| | - Katherine A. Alexander
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, United States of America
| | - Angela J. Churchill
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, United States of America
| | - Cecilia Copperman
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, United States of America
| | - María J. García-García
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, United States of America
- * E-mail:
| |
Collapse
|
57
|
Mohawk promotes the maintenance and regeneration of the outer annulus fibrosus of intervertebral discs. Nat Commun 2016; 7:12503. [PMID: 27527664 PMCID: PMC4990710 DOI: 10.1038/ncomms12503] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 07/07/2016] [Indexed: 01/07/2023] Open
Abstract
The main pathogenesis of intervertebral disc (IVD) herniation involves disruption of the annulus fibrosus (AF) caused by ageing or excessive mechanical stress and the resulting prolapse of the nucleus pulposus. Owing to the avascular nature of the IVD and lack of understanding the mechanisms that maintain the IVD, current therapies do not lead to tissue regeneration. Here we show that homeobox protein Mohawk (Mkx) is a key transcription factor that regulates AF development, maintenance and regeneration. Mkx is mainly expressed in the outer AF (OAF) of humans and mice. In Mkx−/− mice, the OAF displays a deficiency of multiple tendon/ligament-related genes, a smaller OAF collagen fibril diameter and a more rapid progression of IVD degeneration compared with the wild type. Mesenchymal stem cells overexpressing Mkx promote functional AF regeneration in a mouse AF defect model, with abundant collagen fibril formation. Our results indicate a therapeutic strategy for AF regeneration. Homeobox protein Mohwak (Mkx) is involved in tendon and ligament development. Here the authors show that Mkx in the outer annulus fibrosus of the intervertebral disc plays a role in maintenance of the IVD, showing that stem cells overexpressing Mkx enhance therapeutic IVD regeneration in mice.
Collapse
|
58
|
Dnmt3a Regulates Proliferation of Muscle Satellite Cells via p57Kip2. PLoS Genet 2016; 12:e1006167. [PMID: 27415617 PMCID: PMC4944932 DOI: 10.1371/journal.pgen.1006167] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 06/13/2016] [Indexed: 11/18/2022] Open
Abstract
Cell differentiation status is defined by the gene expression profile, which is coordinately controlled by epigenetic mechanisms. Cell type-specific DNA methylation patterns are established by chromatin modifiers including de novo DNA methyltransferases, such as Dnmt3a and Dnmt3b. Since the discovery of the myogenic master gene MyoD, myogenic differentiation has been utilized as a model system to study tissue differentiation. Although knowledge about myogenic gene networks is accumulating, there is only a limited understanding of how DNA methylation controls the myogenic gene program. With an aim to elucidate the role of DNA methylation in muscle development and regeneration, we investigate the consequences of mutating Dnmt3a in muscle precursor cells in mice. Pax3 promoter-driven Dnmt3a-conditional knockout (cKO) mice exhibit decreased organ mass in the skeletal muscles, and attenuated regeneration after cardiotoxin-induced muscle injury. In addition, Dnmt3a-null satellite cells (SCs) exhibit a striking loss of proliferation in culture. Transcriptome analysis reveals dysregulated expression of p57Kip2, a member of the Cip/Kip family of cyclin-dependent kinase inhibitors (CDKIs), in the Dnmt3a-KO SCs. Moreover, RNAi-mediated depletion of p57Kip2 replenishes the proliferation activity of the SCs, thus establishing a role for the Dnmt3a-p57Kip2 axis in the regulation of SC proliferation. Consistent with these findings, Dnmt3a-cKO muscles exhibit fewer Pax7+ SCs, which show increased expression of p57Kip2 protein. Thus, Dnmt3a is found to maintain muscle homeostasis by epigenetically regulating the proliferation of SCs through p57Kip2. How muscle homeostasis is maintained is not completely elucidated yet. Epigenetic disorders such as Beckwith-Wiedemann syndrome, which causes hypergrowth of skeletal muscles and rhabdomyosarcoma, indicate that epigenetic regulations such as DNA methylation, contribute to this homeostasis control. DNA methylation is mediated by DNA methyltransferases, such as Dnmt3a and Dnmt3b, which are de novo DNA methyltransferases. The role of DNA methylation in somatic stem cells is not completely understood, although it has been shown to be indispensable in differentiation of primordial germ cells and embryonic stem cells. In this report, we investigated the role of Dnmt3a in muscle satellite cells by analyzing Dnmt3a-conditional knockout (cKO) mice in which Dnmt3a loci are deleted utilizing Cre-recombinase driven by Pax7 or Pax3 promoters that are specifically activated in the muscle precursor lineage. The loss of Dnmt3a in cKO mice causes decreased muscle mass and significantly impaired muscle regeneration. Moreover, Dnmt3a loss also results in a striking loss of proliferation of SCs, which is caused by mis-expression of a cyclin-dependent kinase inhibitor, p57Kip2. Therefore, our findings suggest that DNA methylation plays an essential role in muscle homeostasis.
Collapse
|
59
|
Gene targeting of the transcription factor Mohawk in rats causes heterotopic ossification of Achilles tendon via failed tenogenesis. Proc Natl Acad Sci U S A 2016; 113:7840-5. [PMID: 27370800 DOI: 10.1073/pnas.1522054113] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Cell-based or pharmacological approaches for promoting tendon repair are currently not available because the molecular mechanisms of tendon development and healing are not well understood. Although analysis of knockout mice provides many critical insights, small animals such as mice have some limitations. In particular, precise physiological examination for mechanical load and the ability to obtain a sufficient number of primary tendon cells for molecular biology studies are challenging using mice. Here, we generated Mohawk (Mkx)(-/-) rats by using CRISPR/Cas9, which showed not only systemic hypoplasia of tendons similar to Mkx(-/-) mice, but also earlier heterotopic ossification of the Achilles tendon compared with Mkx(-/-) mice. Analysis of tendon-derived cells (TDCs) revealed that Mkx deficiency accelerated chondrogenic and osteogenic differentiation, whereas Mkx overexpression suppressed chondrogenic, osteogenic, and adipogenic differentiation. Furthermore, mechanical stretch stimulation of Mkx(-/-) TDCs led to chondrogenic differentiation, whereas the same stimulation in Mkx(+/+) TDCs led to formation of tenocytes. ChIP-seq of Mkx overexpressing TDCs revealed significant peaks in tenogenic-related genes, such as collagen type (Col)1a1 and Col3a1, and chondrogenic differentiation-related genes, such as SRY-box (Sox)5, Sox6, and Sox9 Our results demonstrate that Mkx has a dual role, including accelerating tendon differentiation and preventing chondrogenic/osteogenic differentiation. This molecular network of Mkx provides a basis for tendon physiology and tissue engineering.
Collapse
|
60
|
Clarkson MD. Representation of anatomy in online atlases and databases: a survey and collection of patterns for interface design. BMC DEVELOPMENTAL BIOLOGY 2016; 16:18. [PMID: 27206491 PMCID: PMC4875762 DOI: 10.1186/s12861-016-0116-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/09/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND A large number of online atlases and databases have been developed to mange the rapidly growing amount of data describing embryogenesis. As these community resources continue to evolve, it is important to understand how representations of anatomy can facilitate the sharing and integration of data. In addition, attention to the design of the interfaces is critical to make online resources useful and usable. RESULTS I first present a survey of online atlases and gene expression resources for model organisms, with a focus on methods of semantic and spatial representation of anatomy. A total of 14 anatomical atlases and 21 gene expression resources are included. This survey demonstrates how choices in semantic representation, in the form of ontologies, can enhance interface search functions and provide links between relevant information. This survey also reviews methods for spatially representing anatomy in online resources. I then provide a collection of patterns for interface design based on the atlases and databases surveyed. These patterns include methods for displaying graphics, integrating semantic and spatial representations, organizing information, and querying databases to find genes expressed in anatomical structures. CONCLUSIONS This collection of patterns for interface design will assist biologists and software developers in planning the interfaces of new atlases and databases or enhancing existing ones. They also show the benefits of standardizing semantic and spatial representations of anatomy by demonstrating how interfaces can use standardization to provide enhanced functionality.
Collapse
Affiliation(s)
- Melissa D Clarkson
- Department of Biological Structure, School of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
61
|
Tanaka M. Fins into limbs: Autopod acquisition and anterior elements reduction by modifying gene networks involving 5’Hox , Gli3 , and Shh. Dev Biol 2016; 413:1-7. [DOI: 10.1016/j.ydbio.2016.03.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/04/2016] [Accepted: 03/04/2016] [Indexed: 11/25/2022]
|
62
|
Gurdziel K, Vogt KR, Walton KD, Schneider GK, Gumucio DL. Transcriptome of the inner circular smooth muscle of the developing mouse intestine: Evidence for regulation of visceral smooth muscle genes by the hedgehog target gene, cJun. Dev Dyn 2016; 245:614-26. [PMID: 26930384 DOI: 10.1002/dvdy.24399] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/29/2016] [Accepted: 02/16/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Digestion is facilitated by coordinated contractions of the intestinal muscularis externa, a bilayered smooth muscle structure that is composed of inner circular muscles (ICM) and outer longitudinal muscles (OLM). We performed transcriptome analysis of intestinal mesenchyme tissue at E14.5, when the ICM, but not the OLM, is present, to investigate the transcriptional program of the ICM. RESULTS We identified 3967 genes enriched in E14.5 intestinal mesenchyme. The gene expression profiles were clustered and annotated to known muscle genes, identifying a muscle-enriched subcluster. Using publically available in situ data, 127 genes were verified as expressed in ICM. Examination of the promoter and regulatory regions for these co-expressed genes revealed enrichment for cJUN transcription factor binding sites, and cJUN protein was enriched in ICM. cJUN ChIP-seq, performed at E14.5, revealed that cJUN regulatory regions contain characteristics of muscle enhancers. Finally, we show that cJun is a target of Hedgehog (Hh), a signaling pathway known to be important in smooth muscle development, and identify a cJun genomic enhancer that is responsive to Hh. CONCLUSIONS This work provides the first transcriptional catalog for the developing ICM and suggests that cJun regulates gene expression in the ICM downstream of Hh signaling. Developmental Dynamics 245:614-626, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Katherine Gurdziel
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109
| | - Kyle R Vogt
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109
| | - Katherine D Walton
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109
| | - Gary K Schneider
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109
| | - Deborah L Gumucio
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109
| |
Collapse
|
63
|
Motohashi T, Watanabe N, Nishioka M, Nakatake Y, Yulan P, Mochizuki H, Kawamura Y, Ko MSH, Goshima N, Kunisada T. Gene array analysis of neural crest cells identifies transcription factors necessary for direct conversion of embryonic fibroblasts into neural crest cells. Biol Open 2016; 5:311-22. [PMID: 26873953 PMCID: PMC4810742 DOI: 10.1242/bio.015735] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neural crest cells (NC cells) are multipotent cells that emerge from the edge of the neural folds and migrate throughout the developing embryo. Although the gene regulatory network for generation of NC cells has been elucidated in detail, it has not been revealed which of the factors in the network are pivotal to directing NC identity. In this study we analyzed the gene expression profile of a pure NC subpopulation isolated from Sox10-IRES-Venus mice and investigated whether these genes played a key role in the direct conversion of Sox10-IRES-Venus mouse embryonic fibroblasts (MEFs) into NC cells. The comparative molecular profiles of NC cells and neural tube cells in 9.5-day embryos revealed genes including transcription factors selectively expressed in developing trunk NC cells. Among 25 NC cell-specific transcription factor genes tested, SOX10 and SOX9 were capable of converting MEFs into SOX10-positive (SOX10+) cells. The SOX10+ cells were then shown to differentiate into neurons, glial cells, smooth muscle cells, adipocytes and osteoblasts. These SOX10+ cells also showed limited self-renewal ability, suggesting that SOX10 and SOX9 directly converted MEFs into NC cells. Conversely, the remaining transcription factors, including well-known NC cell specifiers, were unable to convert MEFs into SOX10+ NC cells. These results suggest that SOX10 and SOX9 are the key factors necessary for the direct conversion of MEFs into NC cells. Summary: In this study, we identified the transcription factors specifically expressed in developing neural crest cells, and showed that SOX10 and SOX9 directly converted fibroblasts into neural crest cells.
Collapse
Affiliation(s)
- Tsutomu Motohashi
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan Japan Science and Technology Agency (JST), Core Research for Evolutional Science and Technology (CREST), Tokyo 102-0076, Japan
| | - Natsuki Watanabe
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Masahiro Nishioka
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Yuhki Nakatake
- Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD 21224, USA Department of Systems Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Piao Yulan
- Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Hiromi Mochizuki
- Japan Biological Informatics Consortium (JBiC), Tokyo 135-8073, Japan
| | | | - Minoru S H Ko
- Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD 21224, USA Department of Systems Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Naoki Goshima
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tokyo 135-0064, Japan
| | - Takahiro Kunisada
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan Japan Science and Technology Agency (JST), Core Research for Evolutional Science and Technology (CREST), Tokyo 102-0076, Japan
| |
Collapse
|
64
|
Davignon L, Chauveau C, Julien C, Dill C, Duband-Goulet I, Cabet E, Buendia B, Lilienbaum A, Rendu J, Minot MC, Guichet A, Allamand V, Vadrot N, Fauré J, Odent S, Lazaro L, Leroy JP, Marcorelles P, Dubourg O, Ferreiro A. The transcription coactivator ASC-1 is a regulator of skeletal myogenesis, and its deficiency causes a novel form of congenital muscle disease. Hum Mol Genet 2016; 25:1559-73. [PMID: 27008887 DOI: 10.1093/hmg/ddw033] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 02/04/2016] [Indexed: 01/17/2023] Open
Abstract
Despite recent progress in the genetic characterization of congenital muscle diseases, the genes responsible for a significant proportion of cases remain unknown. We analysed two branches of a large consanguineous family in which four patients presented with a severe new phenotype, clinically marked by neonatal-onset muscle weakness predominantly involving axial muscles, life-threatening respiratory failure, skin abnormalities and joint hyperlaxity without contractures. Muscle biopsies showed the unreported association of multi-minicores, caps and dystrophic lesions. Genome-wide linkage analysis followed by gene and exome sequencing in patients identified a homozygous nonsense mutation in TRIP4 encoding Activating Signal Cointegrator-1 (ASC-1), a poorly characterized transcription coactivator never associated with muscle or with human inherited disease. This mutation resulted in TRIP4 mRNA decay to around 10% of control levels and absence of detectable protein in patient cells. ASC-1 levels were higher in axial than in limb muscles in mouse, and increased during differentiation in C2C12 myogenic cells. Depletion of ASC-1 in cultured muscle cells from a patient and in Trip4 knocked-down C2C12 led to a significant reduction in myotube diameter ex vivo and in vitro, without changes in fusion index or markers of initial myogenic differentiation. This work reports the first TRIP4 mutation and defines a novel form of congenital muscle disease, expanding their histological, clinical and molecular spectrum. We establish the importance of ASC-1 in human skeletal muscle, identify transcriptional co-regulation as novel pathophysiological pathway, define ASC-1 as a regulator of late myogenic differentiation and suggest defects in myotube growth as a novel myopathic mechanism.
Collapse
Affiliation(s)
- Laurianne Davignon
- Pathophysiology of Striated Muscles Laboratory, Unit of Functional and Adaptive Biology (BFA), University Paris Diderot, Sorbonne Paris Cité, BFA, UMR CNRS 8251, 75250 Paris Cedex 13, France, Inserm U787, Myology Group, Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, 75013 Paris, France, UPMC, UMR787, 75013 Paris, France
| | - Claire Chauveau
- Inserm U787, Myology Group, Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, 75013 Paris, France, UPMC, UMR787, 75013 Paris, France
| | - Cédric Julien
- Inserm U787, Myology Group, Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, 75013 Paris, France, UPMC, UMR787, 75013 Paris, France
| | - Corinne Dill
- Pathophysiology of Striated Muscles Laboratory, Unit of Functional and Adaptive Biology (BFA), University Paris Diderot, Sorbonne Paris Cité, BFA, UMR CNRS 8251, 75250 Paris Cedex 13, France
| | - Isabelle Duband-Goulet
- Pathophysiology of Striated Muscles Laboratory, Unit of Functional and Adaptive Biology (BFA), University Paris Diderot, Sorbonne Paris Cité, BFA, UMR CNRS 8251, 75250 Paris Cedex 13, France
| | - Eva Cabet
- Pathophysiology of Striated Muscles Laboratory, Unit of Functional and Adaptive Biology (BFA), University Paris Diderot, Sorbonne Paris Cité, BFA, UMR CNRS 8251, 75250 Paris Cedex 13, France
| | - Brigitte Buendia
- Pathophysiology of Striated Muscles Laboratory, Unit of Functional and Adaptive Biology (BFA), University Paris Diderot, Sorbonne Paris Cité, BFA, UMR CNRS 8251, 75250 Paris Cedex 13, France
| | - Alain Lilienbaum
- Pathophysiology of Striated Muscles Laboratory, Unit of Functional and Adaptive Biology (BFA), University Paris Diderot, Sorbonne Paris Cité, BFA, UMR CNRS 8251, 75250 Paris Cedex 13, France
| | - John Rendu
- Université Grenoble Alpes, Université Joseph Fourier, 38041 Grenoble, France, Biochimie Génétique et Moléculaire, CHRU de Grenoble, 38700 Grenoble, France, INSERM U386, Equipe Muscle et Pathologies, Grenoble Institut des Neurosciences, 38700 Grenoble, France
| | | | - Agnès Guichet
- CHU Angers, Service de génétique médicale, 49100 Angers, France
| | - Valérie Allamand
- UPMC, Inserm UMRS974, CNRS FRE3617, Center for Research in Myology, 75013 Paris, France
| | - Nathalie Vadrot
- Pathophysiology of Striated Muscles Laboratory, Unit of Functional and Adaptive Biology (BFA), University Paris Diderot, Sorbonne Paris Cité, BFA, UMR CNRS 8251, 75250 Paris Cedex 13, France
| | - Julien Fauré
- Université Grenoble Alpes, Université Joseph Fourier, 38041 Grenoble, France, Biochimie Génétique et Moléculaire, CHRU de Grenoble, 38700 Grenoble, France, INSERM U386, Equipe Muscle et Pathologies, Grenoble Institut des Neurosciences, 38700 Grenoble, France
| | - Sylvie Odent
- Pôle Neurosciences, Service de Neurologie, CHU de Rennes, 35033 Rennes, France
| | - Leïla Lazaro
- Service de Pédiatrie, Centre Hospitalier de la Côte Basque, 64109 Bayonne, France
| | - Jean Paul Leroy
- Laboratoire d'Anatomo-Pathologie, CHU de Brest, 29609 Brest, France
| | - Pascale Marcorelles
- Laboratoire d'Anatomo-Pathologie, CHU de Brest, 29609 Brest, France, EA 4685 Laboratoire de Neuroscience de Brest, Université Bretagne Occidentale, 29200 Brest, France
| | - Odile Dubourg
- Inserm U787, Myology Group, Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, 75013 Paris, France, UPMC, UMR787, 75013 Paris, France, AP-HP, Laboratoire de Neuropathologie, Groupe Hospitalier Pitié-Salpêtrière, 75013 Paris, France and
| | - Ana Ferreiro
- Pathophysiology of Striated Muscles Laboratory, Unit of Functional and Adaptive Biology (BFA), University Paris Diderot, Sorbonne Paris Cité, BFA, UMR CNRS 8251, 75250 Paris Cedex 13, France, Inserm U787, Myology Group, Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, 75013 Paris, France, UPMC, UMR787, 75013 Paris, France, AP-HP, Centre de Référence Maladies Neuromusculaires Paris-Est, Groupe Hospitalier Pitié-Salpêtrière, 75013 Paris, France
| |
Collapse
|
65
|
|
66
|
Jin K, Xiao D, Andersen B, Xiang M. Lmo4 and Other LIM domain only factors are necessary and sufficient for multiple retinal cell type development. Dev Neurobiol 2015; 76:900-15. [PMID: 26579872 DOI: 10.1002/dneu.22365] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 10/01/2015] [Accepted: 11/12/2015] [Indexed: 12/13/2022]
Abstract
Understanding the molecular basis by which distinct cell types are specified is a central issue in retinogenesis and retinal disease development. Here we examined the role of LIM domain only 4 (Lmo4) in retinal development using both gain-of-function and loss-of-function approaches. By immunostaining, Lmo4 was found to be expressed in mouse retina from E10.5 to mature stages. Retroviral delivery of Lmo4 into retinal progenitor cells could promote the amacrine, bipolar and Müller cell fates at the expense of photoreceptors. It also inhibited the fate of early-born retinal ganglion cells. Using a dominant-negative form of Lmo4 which suppresses transcriptional activities of all LIM domain only factors, we demonstrated that LIM domain only factors are both necessary and sufficient for promoting amacrine and bipolar cell development, but not for the differentiation of ganglion, horizontal, Müller, or photoreceptor cells. Taken together, our study uncovers multiple roles of Lmo4 during retinal development and demonstrates the importance of LIM domain only factors in ensuring proper retinal cell specification and differentiation. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 900-915, 2016.
Collapse
Affiliation(s)
- Kangxin Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China.,Center for Advanced Biotechnology and Medicine and Department of Pediatrics, Rutgers University-Robert Wood Johnson Medical School, Piscataway, New Jersey, 08854
| | - Dongchang Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Bogi Andersen
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, Irvine, California, 92697-4030.,Department of Biological Chemistry, Division of Endocrinology and Metabolism, University of California, Irvine, California, 92697-4030
| | - Mengqing Xiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China.,Center for Advanced Biotechnology and Medicine and Department of Pediatrics, Rutgers University-Robert Wood Johnson Medical School, Piscataway, New Jersey, 08854
| |
Collapse
|
67
|
Seki R, Kitajima K, Matsubara H, Suzuki T, Saito D, Yokoyama H, Tamura K. AP-2β is a transcriptional regulator for determination of digit length in tetrapods. Dev Biol 2015; 407:75-89. [DOI: 10.1016/j.ydbio.2015.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 08/06/2015] [Accepted: 08/07/2015] [Indexed: 10/23/2022]
|
68
|
Onimaru K, Kuraku S, Takagi W, Hyodo S, Sharpe J, Tanaka M. A shift in anterior-posterior positional information underlies the fin-to-limb evolution. eLife 2015; 4. [PMID: 26283004 PMCID: PMC4538735 DOI: 10.7554/elife.07048] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 07/15/2015] [Indexed: 02/07/2023] Open
Abstract
The pectoral fins of ancestral fishes had multiple proximal elements connected to their pectoral girdles. During the fin-to-limb transition, anterior proximal elements were lost and only the most posterior one remained as the humerus. Thus, we hypothesised that an evolutionary alteration occurred in the anterior–posterior (AP) patterning system of limb buds. In this study, we examined the pectoral fin development of catshark (Scyliorhinus canicula) and revealed that the AP positional values in fin buds are shifted more posteriorly than mouse limb buds. Furthermore, examination of Gli3 function and regulation shows that catshark fins lack a specific AP patterning mechanism, which restricts its expression to an anterior domain in tetrapods. Finally, experimental perturbation of AP patterning in catshark fin buds results in an expansion of posterior values and loss of anterior skeletal elements. Together, these results suggest that a key genetic event of the fin-to-limb transformation was alteration of the AP patterning network. DOI:http://dx.doi.org/10.7554/eLife.07048.001 Humans, mice, and other animals with four limbs belong to a group of land-dwelling animals known as the tetrapods. This group of animals evolved from ancient fish and one crucial adaptation to life on land involved the modification of fins to form limbs. The front pair of limbs (the ‘arms’) evolved from the ‘pectoral’ fins of the ancient fish. These fins contain numerous bones that fan out from a set of bones called the pectoral girdle. However, most of the bones nearer the front side (the thumb side in the human limb) were lost in the ancestors of tetrapods as they moved onto land. Only the bone nearest the back remained as the ‘humerus’, which forms the upper part of the limb (i.e., the upper arm of humans). In the embryos of mice and other animals, the limbs develop from structures called limb buds. For the limb to develop properly, the cells in the limb bud need to receive specific instructions that depend on their position in the bud. A protein called Gli3R provides cells with information about their position along the ‘anterior–posterior’ (or thumb-to-little finger) axis of the bud. This protein regulates several genes that are involved in limb development, and this results in different genes being expressed in cells along the anterior–posterior axis. For example, Alx4 is only expressed in a small area at the anterior end of the bud, while Hand2 expression is found in a large area towards the posterior part. Gli3R is also found in a fish called the catshark, but it is not clear how it controls the formation of fins. Onimaru et al. show that the pattern of gene expression in the catshark fin bud is different to that of the mouse limb bud. For example, Alx4 is expressed in a larger area of the fin bud that extends further towards the posterior, while Hand2 is only found in a much smaller area at the posterior end of the bud. The experiments also suggest that Gli3R is active in a much larger area of the fin bud than in the limb bud. Next, Onimaru et al. used a drug on the catshark embryos to increase the activity of another protein that can inhibit Gli3R. The fin buds of these shark had anterior shift in several gene expression domains, and the fins that formed were missing several anterior bones and had only a single bone connected to the pectoral girdle. Onimaru et al.'s findings suggest that during the evolution of the tetrapods, there may have been a shift in the anterior–posterior patterning of the fin bud to form a limb. An important area for future work will be to use genome-wide studies to study the fin/limb buds of other species. DOI:http://dx.doi.org/10.7554/eLife.07048.002
Collapse
Affiliation(s)
- Koh Onimaru
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | - Shigehiro Kuraku
- Phyloinformatics Unit, RIKEN Center for Life Science Technologies, Kobe, Japan
| | - Wataru Takagi
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Chiba, Japan
| | - Susumu Hyodo
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Chiba, Japan
| | - James Sharpe
- EMBL-CRG Systems Biology Research Unit, Centre for Genomic Regulation, Universitat Pompeu Fabra, Barcelona, Spain
| | - Mikiko Tanaka
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
69
|
Lidral AC, Liu H, Bullard SA, Bonde G, Machida J, Visel A, Uribe LMM, Li X, Amendt B, Cornell RA. A single nucleotide polymorphism associated with isolated cleft lip and palate, thyroid cancer and hypothyroidism alters the activity of an oral epithelium and thyroid enhancer near FOXE1. Hum Mol Genet 2015; 24:3895-907. [PMID: 25652407 PMCID: PMC4476440 DOI: 10.1093/hmg/ddv047] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 02/02/2015] [Indexed: 12/11/2022] Open
Abstract
Three common diseases, isolated cleft lip and cleft palate (CLP), hypothyroidism and thyroid cancer all map to the FOXE1 locus, but causative variants have yet to be identified. In patients with CLP, the frequency of coding mutations in FOXE1 fails to account for the risk attributable to this locus, suggesting that the common risk alleles reside in nearby regulatory elements. Using a combination of zebrafish and mouse transgenesis, we screened 15 conserved non-coding sequences for enhancer activity, identifying three that regulate expression in a tissue specific pattern consistent with endogenous foxe1 expression. These three, located -82.4, -67.7 and +22.6 kb from the FOXE1 start codon, are all active in the oral epithelium or branchial arches. The -67.7 and +22.6 kb elements are also active in the developing heart, and the -67.7 kb element uniquely directs expression in the developing thyroid. Within the -67.7 kb element is the SNP rs7850258 that is associated with all three diseases. Quantitative reporter assays in oral epithelial and thyroid cell lines show that the rs7850258 allele (G) associated with CLP and hypothyroidism has significantly greater enhancer activity than the allele associated with thyroid cancer (A). Moreover, consistent with predicted transcription factor binding differences, the -67.7 kb element containing rs7850258 allele G is significantly more responsive to both MYC and ARNT than allele A. By demonstrating that this common non-coding variant alters FOXE1 expression, we have identified at least in part the functional basis for the genetic risk of these seemingly disparate disorders.
Collapse
Affiliation(s)
| | - Huan Liu
- Dows Research Institute, State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | | | - Greg Bonde
- Department of Anatomy, University of Iowa, Iowa City, IA, USA
| | - Junichiro Machida
- Department of Oral and Maxillofacial Surgery, Toyota Memorial Hospital, Toyota City, Aichi, Japan
| | - Axel Visel
- Genomics Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA and Department of Energy Joint Genome Institute, Walnut Creek, CA, USA
| | | | - Xiao Li
- Department of Anatomy, University of Iowa, Iowa City, IA, USA
| | - Brad Amendt
- Department of Anatomy, University of Iowa, Iowa City, IA, USA
| | | |
Collapse
|
70
|
Jung ES, Sim YJ, Jeong HS, Kim SJ, Yun YJ, Song JH, Jeon SH, Choe C, Park KT, Kim CH, Kim KS. Jmjd2C increases MyoD transcriptional activity through inhibiting G9a-dependent MyoD degradation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:1081-94. [PMID: 26149774 DOI: 10.1016/j.bbagrm.2015.07.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/17/2015] [Accepted: 07/02/2015] [Indexed: 01/05/2023]
Abstract
Skeletal muscle cell differentiation requires a family of proteins called myogenic regulatory factors (MRFs) to which MyoD belongs. The activity of MyoD is under epigenetic regulation, however, the molecular mechanism by which histone KMTs and KDMs regulate MyoD transcriptional activity through methylation remains to be determined. Here we provide evidence for a unique regulatory mechanism of MyoD transcriptional activity through demethylation by Jmjd2C demethylase whose level increases during muscle differentiation. G9a decreases MyoD stability via methylation-dependent MyoD ubiquitination. Jmjd2C directly associates with MyoD in vitro and in vivo to demethylate and stabilize MyoD. The hypo-methylated MyoD due to Jmjd2C is significantly more stable than hyper-methylated MyoD by G9a. Cul4/Ddb1/Dcaf1 pathway is essential for the G9a-mediated MyoD degradation in myoblasts. By the stabilization of MyoD, Jmjd2C increases myogenic conversion of mouse embryonic fibroblasts and MyoD transcriptional activity with erasing repressive H3K9me3 level at the promoter of MyoD target genes. Collectively, Jmjd2C increases MyoD transcriptional activity to facilitate skeletal muscle differentiation by increasing MyoD stability through inhibiting G9a-dependent MyoD degradation.
Collapse
Affiliation(s)
- Eun-Shil Jung
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Republic of Korea
| | - Ye-Ji Sim
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Republic of Korea
| | - Hoe-Su Jeong
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Republic of Korea
| | - Su-Jin Kim
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Republic of Korea
| | - Ye-Jin Yun
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Republic of Korea
| | - Joo-Hoon Song
- Bio Focus Co., Ltd., Gyeonggi-do 437-753, Republic of Korea
| | - Su-Hee Jeon
- Department of Biological & Environmental Science, Dongguk University, Seoul 100-175, Republic of Korea
| | - Chungyoul Choe
- Samsung Biomedical Research Institute, School of Medicine, Sungkyunkwan University, Seoul 135-710, Republic of Korea
| | - Kyung-Tae Park
- Center for Cancer Research, Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Chang-Hoon Kim
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Republic of Korea.
| | - Kye-Seong Kim
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Republic of Korea.
| |
Collapse
|
71
|
Oralová V, Matalová E, Janečková E, Drobná Krejčí E, Knopfová L, Šnajdr P, Tucker AS, Veselá I, Šmarda J, Buchtová M. Role of c-Myb in chondrogenesis. Bone 2015; 76:97-106. [PMID: 25845979 DOI: 10.1016/j.bone.2015.02.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 02/20/2015] [Accepted: 02/21/2015] [Indexed: 12/22/2022]
Abstract
The Myb locus encodes the c-Myb transcription factor involved in controlling a broad variety of cellular processes. Recently, it has been shown that c-Myb may play a specific role in hard tissue formation; however, all of these results were gathered from an analysis of intramembranous ossification. To investigate a possible role of c-Myb in endochondral ossification, we carried out our study on the long bones of mouse limbs during embryonic development. Firstly, the c-myb expression pattern was analyzed by in situ hybridization during endochondral ossification of long bones. c-myb positive areas were found in proliferating as well as hypertrophic zones of the growth plate. At early embryonic stages, localized expression was also observed in the perichondrium and interdigital areas. The c-Myb protein was found in proliferating chondrocytes and in the perichondrium of the forelimb bones (E14.5-E17.5). Furthermore, protein was detected in pre-hypertrophic as well as hypertrophic chondrocytes. Gain-of-function and loss-of-function approaches were used to test the effect of altered c-myb expression on chondrogenesis in micromass cultures established from forelimb buds of mouse embryos. A loss-of-function approach using c-myb specific siRNA decreased nodule formation, as well as downregulated the level of Sox9 expression, a major marker of chondrogenesis. Transient c-myb overexpression markedly increased the formation of cartilage nodules and the production of extracellular matrix as detected by intense staining with Alcian blue. Moreover, the expression of early chondrogenic genes such as Sox9, Col2a1 and activity of a Col2-LUC reporter were increased in the cells overexpressing c-myb while late chondrogenic markers such as Col10a1 and Mmp13 were not significantly changed or were downregulated. Taken together, the results of this study demonstrate that the c-Myb transcription factor is involved in the regulation and promotion of endochondral bone formation.
Collapse
Affiliation(s)
- V Oralová
- Institute of Animal Physiology and Genetics CAS, v.v.i., Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
| | - E Matalová
- Institute of Animal Physiology and Genetics CAS, v.v.i., Brno, Czech Republic; Department of Physiology, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| | - E Janečková
- Institute of Animal Physiology and Genetics CAS, v.v.i., Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - E Drobná Krejčí
- Institute of Anatomy, Charles University, Prague, Czech Republic
| | - L Knopfová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - P Šnajdr
- Institute of Anatomy, Charles University, Prague, Czech Republic
| | - A S Tucker
- Department of Craniofacial Development and Stem Cell Biology, King's College London, London, UK
| | - I Veselá
- Department of Anatomy, Histology and Embryology, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| | - J Šmarda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - M Buchtová
- Institute of Animal Physiology and Genetics CAS, v.v.i., Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| |
Collapse
|
72
|
Analysis of tandem E-box motifs within human Complement receptor 2 (CR2/CD21) promoter reveals cell specific roles for RP58, E2A, USF and localized chromatin accessibility. Int J Biochem Cell Biol 2015; 64:107-19. [DOI: 10.1016/j.biocel.2015.03.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/03/2015] [Accepted: 03/18/2015] [Indexed: 02/06/2023]
|
73
|
Zhang P, Liang X, Shan T, Jiang Q, Deng C, Zheng R, Kuang S. mTOR is necessary for proper satellite cell activity and skeletal muscle regeneration. Biochem Biophys Res Commun 2015; 463:102-8. [PMID: 25998386 DOI: 10.1016/j.bbrc.2015.05.032] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 05/09/2015] [Indexed: 12/14/2022]
Abstract
The serine/threonine kinase mammalian target of rapamycin (mTOR) is a key regulator of protein synthesis, cell proliferation and energy metabolism. As constitutive deletion of Mtor gene results in embryonic lethality, the function of mTOR in muscle stem cells (satellite cells) and skeletal muscle regeneration remains to be determined. In this study, we established a satellite cell specific Mtor conditional knockout (cKO) mouse model by crossing Pax7(CreER) and Mtor(flox/flox) mice. Skeletal muscle regeneration after injury was severely compromised in the absence of Mtor, indicated by increased number of necrotic myofibers infiltrated by Evans blue dye, and reduced number and size of regenerated myofibers in the Mtor cKO mice compared to wild type (WT) littermates. To dissect the cellular mechanism, we analyzed satellite cell-derived primary myoblasts grown on single myofibers or adhered to culture plates. The Mtor cKO myoblasts exhibited defective proliferation and differentiation kinetics when compared to myoblasts derived from WT littermates. At the mRNA and protein levels, the Mtor cKO myoblasts expressed lower levels of key myogenic determinant genes Pax7, Myf5, Myod, Myog than did the WT myoblasts. These results suggest that mTOR is essential for satellite cell function and skeletal muscle regeneration through controlling the expression of myogenic genes.
Collapse
Affiliation(s)
- Pengpeng Zhang
- Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Xinrong Liang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Tizhong Shan
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Qinyang Jiang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Changyan Deng
- Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Rong Zheng
- Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
74
|
Heng JIT, Qu Z, Ohtaka-Maruyama C, Okado H, Kasai M, Castro D, Guillemot F, Tan SS. The zinc finger transcription factor RP58 negatively regulates Rnd2 for the control of neuronal migration during cerebral cortical development. Cereb Cortex 2015; 25:806-16. [PMID: 24084125 DOI: 10.1093/cercor/bht277] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
The zinc finger transcription factor RP58 (also known as ZNF238) regulates neurogenesis of the mouse neocortex and cerebellum (Okado et al. 2009; Xiang et al. 2011; Baubet et al. 2012; Ohtaka-Maruyama et al. 2013), but its mechanism of action remains unclear. In this study, we report a cell-autonomous function for RP58 during the differentiation of embryonic cortical projection neurons via its activities as a transcriptional repressor. Disruption of RP58 expression alters the differentiation of immature neurons and impairs their migration and positioning within the mouse cerebral cortex. Loss of RP58 within the embryonic cortex also leads to elevated mRNA for Rnd2, a member of the Rnd family of atypical RhoA-like GTPase proteins important for cortical neuron migration (Heng et al. 2008). Mechanistically, RP58 represses transcription of Rnd2 via binding to a 3'-regulatory enhancer in a sequence-specific fashion. Using reporter assays, we found that RP58 repression of Rnd2 is competed by proneural basic helix-loop-helix transcriptional activators. Finally, our rescue experiments revealed that negative regulation of Rnd2 by RP58 was important for cortical cell migration in vivo. Taken together, these studies demonstrate that RP58 is a key player in the transcriptional control of cell migration in the developing cerebral cortex.
Collapse
Affiliation(s)
- Julian Ik-Tsen Heng
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia Florey Institute of Neuroscience and Mental Health, Genetics Lane, The University of Melbourne, Parkville, Melbourne, VIC 3010, Australia
| | - Zhengdong Qu
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia Florey Institute of Neuroscience and Mental Health, Genetics Lane, The University of Melbourne, Parkville, Melbourne, VIC 3010, Australia
| | - Chiaki Ohtaka-Maruyama
- Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8506, Japan
| | - Haruo Okado
- Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8506, Japan
| | - Masataka Kasai
- Center for Stem Cell and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan and
| | - Diogo Castro
- Division of Molecular Neurobiology, MRC National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - François Guillemot
- Division of Molecular Neurobiology, MRC National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Seong-Seng Tan
- Florey Institute of Neuroscience and Mental Health, Genetics Lane, The University of Melbourne, Parkville, Melbourne, VIC 3010, Australia
| |
Collapse
|
75
|
Elwell JA, Lovato TL, Adams MM, Baca EM, Lee T, Cripps RM. The myogenic repressor gene Holes in muscles is a direct transcriptional target of Twist and Tinman in the Drosophila embryonic mesoderm. Dev Biol 2015; 400:266-76. [PMID: 25704510 DOI: 10.1016/j.ydbio.2015.02.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/14/2015] [Accepted: 02/10/2015] [Indexed: 11/19/2022]
Abstract
Understanding the regulatory circuitry controlling myogenesis is critical to understanding developmental mechanisms and developmentally-derived diseases. We analyzed the transcriptional regulation of a Drosophila myogenic repressor gene, Holes in muscles (Him). Previously, Him was shown to inhibit Myocyte enhancer factor-2 (MEF2) activity, and is expressed in myoblasts but not differentiating myotubes. We demonstrate that different phases of Him embryonic expression arises through the actions of different enhancers, and we characterize the enhancer required for its early mesoderm expression. This Him early mesoderm enhancer contains two conserved binding sites for the basic helix-loop-helix regulator Twist, and one binding site for the NK homeodomain protein Tinman. The sites for both proteins are required for enhancer activity in early embryos. Twist and Tinman activate the enhancer in tissue culture assays, and ectopic expression of either factor is sufficient to direct ectopic expression of a Him-lacZ reporter, or of the endogenous Him gene. Moreover, sustained expression of twist in the mesoderm up-regulates mesodermal Him expression in late embryos. Our findings provide a model to define mechanistically how Twist can both promotes myogenesis through direct activation of Mef2, and can place a brake on myogenesis, through direct activation of Him.
Collapse
Affiliation(s)
- Jennifer A Elwell
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - TyAnna L Lovato
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Melanie M Adams
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Erica M Baca
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Thai Lee
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Richard M Cripps
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA.
| |
Collapse
|
76
|
Hoxa2 selectively enhances Meis binding to change a branchial arch ground state. Dev Cell 2015; 32:265-77. [PMID: 25640223 PMCID: PMC4333904 DOI: 10.1016/j.devcel.2014.12.024] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/18/2014] [Accepted: 12/23/2014] [Indexed: 02/07/2023]
Abstract
Hox transcription factors (TFs) are essential for vertebrate development, but how these evolutionary conserved proteins function in vivo remains unclear. Because Hox proteins have notoriously low binding specificity, they are believed to bind with cofactors, mainly homeodomain TFs Pbx and Meis, to select their specific targets. We mapped binding of Meis, Pbx, and Hoxa2 in the branchial arches, a series of segments in the developing vertebrate head. Meis occupancy is largely similar in Hox-positive and -negative arches. Hoxa2, which specifies second arch (IIBA) identity, recognizes a subset of Meis prebound sites that contain Hox motifs. Importantly, at these sites Meis binding is strongly increased. This enhanced Meis binding coincides with active enhancers, which are linked to genes highly expressed in the IIBA and regulated by Hoxa2. These findings show that Hoxa2 operates as a tissue-specific cofactor, enhancing Meis binding to specific sites that provide the IIBA with its anatomical identity. Meis provides a ground state that is common to all the branchial arches Hoxa2 recognizes Meis prebound sites in the second arch that contain Hox motifs Hoxa2 enhances Meis binding, which coincides with active enhancers, at these sites Hoxa2 modulates the ground-state binding of Meis to instruct second arch identity
Collapse
|
77
|
Shekaran A, Shoemaker JT, Kavanaugh TE, Lin AS, LaPlaca MC, Fan Y, Guldberg RE, García AJ. The effect of conditional inactivation of beta 1 integrins using twist 2 Cre, Osterix Cre and osteocalcin Cre lines on skeletal phenotype. Bone 2014; 68:131-41. [PMID: 25183373 PMCID: PMC4189988 DOI: 10.1016/j.bone.2014.08.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 08/13/2014] [Accepted: 08/16/2014] [Indexed: 11/27/2022]
Abstract
Skeletal development and growth are complex processes regulated by multiple microenvironmental cues, including integrin-ECM interactions. The β1 sub-family of integrins is the largest integrin sub-family and constitutes the main integrin binding partners of collagen I, the major ECM component of bone. As complete β1 integrin knockout results in embryonic lethality, studies of β1 integrin function in vivo rely on tissue-specific gene deletions. While multiple in vitro studies indicate that β1 integrins are crucial regulators of osteogenesis and mineralization, in vivo osteoblast-specific perturbations of β1 integrins have resulted in mild and sometimes contradictory skeletal phenotypes. To further investigate the role of β1 integrins on skeletal phenotype, we used the Twist2-Cre, Osterix-Cre and osteocalcin-Cre lines to generate conditional β1 integrin deletions, where Cre is expressed primarily in mesenchymal condensation, pre-osteoblast, and mature osteoblast lineage cells respectively within these lines. Mice with Twist2-specific β1 integrin disruption were smaller, had impaired skeletal development, especially in the craniofacial and vertebral tissues at E19.5, and did not survive beyond birth. Osterix-specific β1 integrin deficiency resulted in viable mice which were normal at birth but displayed early defects in calvarial ossification, incisor eruption and growth as well as femoral bone mineral density, structure, and mechanical properties. Although these defects persisted into adulthood, they became milder with age. Finally, a lack of β1 integrins in mature osteoblasts and osteocytes resulted in minor alterations to femur structure but had no effect on mineral density, biomechanics or fracture healing. Taken together, our data indicate that β1 integrin expression in early mesenchymal condensations play an important role in skeletal ossification, while β1 integrin-ECM interactions in pre-osteoblast, odontoblast- and hypertrophic chondryocyte-lineage cells regulate incisor eruption and perinatal bone formation in both intramembranously and endochondrally formed bones in young, rapidly growing mice. In contrast, the osteocalcin-specific β1 integrin deletion had only minor effects on skeletal phenotype.
Collapse
Affiliation(s)
- Asha Shekaran
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332, USA
| | - James T Shoemaker
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Taylor E Kavanaugh
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Angela S Lin
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332, USA; School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA 30332, USA
| | - Michelle C LaPlaca
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Yuhong Fan
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332, USA; School of Biology, Georgia Institute of Technology, 310 Ferst Drive, Atlanta, GA 30332, USA
| | - Robert E Guldberg
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332, USA; School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA 30332, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332, USA; School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA 30332, USA.
| |
Collapse
|
78
|
Miyata K, Miyata T, Nakabayashi K, Okamura K, Naito M, Kawai T, Takada S, Kato K, Miyamoto S, Hata K, Asahara H. DNA methylation analysis of human myoblasts during in vitro myogenic differentiation: de novo methylation of promoters of muscle-related genes and its involvement in transcriptional down-regulation. Hum Mol Genet 2014; 24:410-23. [PMID: 25190712 DOI: 10.1093/hmg/ddu457] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Although DNA methylation is considered to play an important role during myogenic differentiation, chronological alterations in DNA methylation and gene expression patterns in this process have been poorly understood. Using the Infinium HumanMethylation450 BeadChip array, we obtained a chronological profile of the genome-wide DNA methylation status in a human myoblast differentiation model, where myoblasts were cultured in low-serum medium to stimulate myogenic differentiation. As the differentiation of the myoblasts proceeded, their global DNA methylation level increased and their methylation patterns became more distinct from those of mesenchymal stem cells. Gene ontology analysis revealed that genes whose promoter region was hypermethylated upon myoblast differentiation were highly significantly enriched with muscle-related terms such as 'muscle contraction' and 'muscle system process'. Sequence motif analysis identified 8-bp motifs somewhat similar to the binding motifs of ID4 and ZNF238 to be most significantly enriched in hypermethylated promoter regions. ID4 and ZNF238 have been shown to be critical transcriptional regulators of muscle-related genes during myogenic differentiation. An integrated analysis of DNA methylation and gene expression profiles revealed that de novo DNA methylation of non-CpG island (CGI) promoters was more often associated with transcriptional down-regulation than that of CGI promoters. These results strongly suggest the existence of an epigenetic mechanism in which DNA methylation modulates the functions of key transcriptional factors to coordinately regulate muscle-related genes during myogenic differentiation.
Collapse
Affiliation(s)
- Kohei Miyata
- Department of Systems BioMedicine and Department of Obstetrics and Gynecology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | - Tomoko Miyata
- Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kazuhiko Nakabayashi
- Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | | | - Masashi Naito
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan and
| | - Tomoko Kawai
- Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | | | - Kiyoko Kato
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Shingo Miyamoto
- Department of Obstetrics and Gynecology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | - Kenichiro Hata
- Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Hiroshi Asahara
- Department of Systems BioMedicine and Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan and Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
79
|
Suda N, Itoh T, Nakato R, Shirakawa D, Bando M, Katou Y, Kataoka K, Shirahige K, Tickle C, Tanaka M. Dimeric combinations of MafB, cFos and cJun control the apoptosis-survival balance in limb morphogenesis. Development 2014; 141:2885-94. [DOI: 10.1242/dev.099150] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Apoptosis is an important mechanism for sculpting morphology. However, the molecular cascades that control apoptosis in developing limb buds remain largely unclear. Here, we show that MafB was specifically expressed in apoptotic regions of chick limb buds, and MafB/cFos heterodimers repressed apoptosis, whereas MafB/cJun heterodimers promoted apoptosis for sculpting the shape of the limbs. Chromatin immunoprecipitation sequencing in chick limb buds identified potential target genes and regulatory elements controlled by Maf and Jun. Functional analyses revealed that expression of p63 and p73, key components known to arrest the cell cycle, was directly activated by MafB and cJun. Our data suggest that dimeric combinations of MafB, cFos and cJun in developing chick limb buds control the number of apoptotic cells, and that MafB/cJun heterodimers lead to apoptosis via activation of p63 and p73.
Collapse
Affiliation(s)
- Natsuno Suda
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B-17, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Takehiko Itoh
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B-34, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Ryuichiro Nakato
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Daisuke Shirakawa
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B-17, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Masashige Bando
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Yuki Katou
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Kohsuke Kataoka
- Graduate School of Medical Life Sciences, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Katsuhiko Shirahige
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Cheryll Tickle
- Department of Biology and Biochemistry, University of Bath, Claverton Down Road, Bath BA2 7AY, UK
| | - Mikiko Tanaka
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B-17, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| |
Collapse
|
80
|
Patterson ES, Waller LE, Kroll KL. Geminin loss causes neural tube defects through disrupted progenitor specification and neuronal differentiation. Dev Biol 2014; 393:44-56. [PMID: 24995796 DOI: 10.1016/j.ydbio.2014.06.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 06/17/2014] [Accepted: 06/18/2014] [Indexed: 01/13/2023]
Abstract
Geminin is a nucleoprotein that can directly bind chromatin regulatory complexes to modulate gene expression during development. Geminin knockout mouse embryos are preimplantation lethal by the 32-cell stage, precluding in vivo study of Geminin's role in neural development. Therefore, here we used a conditional Geminin allele in combination with several Cre-driver lines to define an essential role for Geminin during mammalian neural tube (NT) formation and patterning. Geminin was required in the NT within a critical developmental time window (embryonic day 8.5-10.5), when NT patterning and closure occurs. Geminin excision at these stages resulted in strongly diminished expression of genes that mark and promote dorsal NT identities and decreased differentiation of ventral motor neurons, resulting in completely penetrant NT defects, while excision after embryonic day 10.5 did not result in NT defects. When Geminin was deleted specifically in the spinal NT, both NT defects and axial skeleton defects were observed, but neither defect occurred when Geminin was excised in paraxial mesenchyme, indicating a tissue autonomous requirement for Geminin in developing neuroectoderm. Despite a potential role for Geminin in cell cycle control, we found no evidence of proliferation defects or altered apoptosis. Comparisons of gene expression in the NT of Geminin mutant versus wild-type siblings at embryonic day 10.5 revealed decreased expression of key regulators of neurogenesis, including neurogenic bHLH transcription factors and dorsal interneuron progenitor markers. Together, these data demonstrate a requirement for Geminin for NT patterning and neuronal differentiation during mammalian neurulation in vivo.
Collapse
Affiliation(s)
- Ethan S Patterson
- Department of Developmental Biology, Washington University School of Medicine, 320 McDonnell Sciences Building, Campus Box 8103, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Laura E Waller
- Department of Developmental Biology, Washington University School of Medicine, 320 McDonnell Sciences Building, Campus Box 8103, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Kristen L Kroll
- Department of Developmental Biology, Washington University School of Medicine, 320 McDonnell Sciences Building, Campus Box 8103, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
81
|
The Mohawk homeobox transcription factor regulates the differentiation of tendons and volar plates. J Orthop Sci 2014; 19:172-80. [PMID: 24166359 PMCID: PMC3943675 DOI: 10.1007/s00776-013-0485-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 10/08/2013] [Indexed: 12/27/2022]
Abstract
BACKGROUND Mohawk (Mkx) is a homeodomain-containing transcription factor that is expressed in various mesoderm-derived tissues, particularly in developing tendons. In this study, we investigate the exact expression pattern and functions of Mkx in forelimbs. METHODS We analyzed the forelimbs of Mkx knockout mice [from embryonic day (E) 18.5 to postnatal day (P) 28 weeks] by using knocked-in Venus signals, Masson trichrome staining, and hematoxylin and eosin (H&E) staining. RESULTS We detected Venus signals in forelimb tendons, pulleys, and volar plates (VPs) in P21 mice. In-depth histological analysis showed that compared to the wild-type mice, the Mkx knockout mice showed significant hypoplasia in the flexor digitorum profundus tendons from E18.5. The VPs and pulleys appeared normal until P0; however, by P14, they became increasingly thicker in Mkx-null mice compared to wild-type mice. The fiber alignment was particularly disrupted in VPs of Mkx-null mice. CONCLUSIONS These results suggest that Mkx is an important regulator of the differentiation of VPs and pulleys, as well as of tendon differentiation.
Collapse
|
82
|
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
|
83
|
Richardson L, Venkataraman S, Stevenson P, Yang Y, Moss J, Graham L, Burton N, Hill B, Rao J, Baldock RA, Armit C. EMAGE mouse embryo spatial gene expression database: 2014 update. Nucleic Acids Res 2013; 42:D835-44. [PMID: 24265223 PMCID: PMC3965061 DOI: 10.1093/nar/gkt1155] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
EMAGE (http://www.emouseatlas.org/emage/) is a freely available database of in situ gene expression patterns that allows users to perform online queries of mouse developmental gene expression. EMAGE is unique in providing both text-based descriptions of gene expression plus spatial maps of gene expression patterns. This mapping allows spatial queries to be accomplished alongside more traditional text-based queries. Here, we describe our recent progress in spatial mapping and data integration. EMAGE has developed a method of spatially mapping 3D embryo images captured using optical projection tomography, and through the use of an IIP3D viewer allows users to view arbitrary sections of raw and mapped 3D image data in the context of a web browser. EMAGE now includes enhancer data, and we have spatially mapped images from a comprehensive screen of transgenic reporter mice that detail the expression of mouse non-coding genomic DNA fragments with enhancer activity. We have integrated the eMouseAtlas anatomical atlas and the EMAGE database so that a user of the atlas can query the EMAGE database easily. In addition, we have extended the atlas framework to enable EMAGE to spatially cross-index EMBRYS whole mount in situ hybridization data. We additionally report on recent developments to the EMAGE web interface, including new query and analysis capabilities.
Collapse
Affiliation(s)
- Lorna Richardson
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital EH4 2XU, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Attanasio C, Nord AS, Zhu Y, Blow MJ, Li Z, Liberton DK, Morrison H, Plajzer-Frick I, Holt A, Hosseini R, Phouanenavong S, Akiyama JA, Shoukry M, Afzal V, Rubin EM, FitzPatrick DR, Ren B, Hallgrímsson B, Pennacchio LA, Visel A. Fine tuning of craniofacial morphology by distant-acting enhancers. Science 2013; 342:1241006. [PMID: 24159046 PMCID: PMC3991470 DOI: 10.1126/science.1241006] [Citation(s) in RCA: 182] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The shape of the human face and skull is largely genetically determined. However, the genomic basis of craniofacial morphology is incompletely understood and hypothesized to involve protein-coding genes, as well as gene regulatory sequences. We used a combination of epigenomic profiling, in vivo characterization of candidate enhancer sequences in transgenic mice, and targeted deletion experiments to examine the role of distant-acting enhancers in craniofacial development. We identified complex regulatory landscapes consisting of enhancers that drive spatially complex developmental expression patterns. Analysis of mouse lines in which individual craniofacial enhancers had been deleted revealed significant alterations of craniofacial shape, demonstrating the functional importance of enhancers in defining face and skull morphology. These results demonstrate that enhancers are involved in craniofacial development and suggest that enhancer sequence variation contributes to the diversity of human facial morphology.
Collapse
Affiliation(s)
- Catia Attanasio
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720,
USA
| | - Alex S. Nord
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720,
USA
| | - Yiwen Zhu
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720,
USA
| | | | - Zirong Li
- Ludwig Institute for Cancer Research, and Department of
Cellular and Molecular Medicine, University of California, San Diego School of
Medicine, 9500 Gilman Drive, La Jolla, CA
| | - Denise K. Liberton
- Dept. of Cell Biology & Anatomy, McCaig Bone and
Joint Institute and the Alberta Children's Hospital Research Institute,
University of Calgary, Canada
| | - Harris Morrison
- MRC Human Genetics Unit, MRC Institute for Genetic and
Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | | | - Amy Holt
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720,
USA
| | - Roya Hosseini
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720,
USA
| | | | | | - Malak Shoukry
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720,
USA
| | - Veena Afzal
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720,
USA
| | - Edward M. Rubin
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720,
USA
- DOE Joint Genome Institute, Walnut Creek, CA
| | - David R. FitzPatrick
- MRC Human Genetics Unit, MRC Institute for Genetic and
Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Bing Ren
- Ludwig Institute for Cancer Research, and Department of
Cellular and Molecular Medicine, University of California, San Diego School of
Medicine, 9500 Gilman Drive, La Jolla, CA
| | - Benedikt Hallgrímsson
- Dept. of Cell Biology & Anatomy, McCaig Bone and
Joint Institute and the Alberta Children's Hospital Research Institute,
University of Calgary, Canada
| | - Len A. Pennacchio
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720,
USA
- DOE Joint Genome Institute, Walnut Creek, CA
| | - Axel Visel
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720,
USA
- DOE Joint Genome Institute, Walnut Creek, CA
| |
Collapse
|
85
|
Shimizu H, Kubo A, Uchibe K, Hashimoto M, Yokoyama S, Takada S, Mitsuoka K, Asahara H. The AERO system: a 3D-like approach for recording gene expression patterns in the whole mouse embryo. PLoS One 2013; 8:e75754. [PMID: 24146773 PMCID: PMC3797748 DOI: 10.1371/journal.pone.0075754] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 08/20/2013] [Indexed: 12/30/2022] Open
Abstract
We have recently constructed a web-based database of gene expression in the mouse whole embryo, EMBRYS (http://embrys.jp/embrys/html/MainMenu.html). To allow examination of gene expression patterns to the fullest extent possible, this database provides both photo images and annotation data. However, since embryos develop via an intricate process of morphogenesis, it would be of great value to track embryonic gene expression from a three dimensional perspective. In fact, several methods have been developed to achieve this goal, but highly laborious procedures and specific operational skills are generally required. We utilized a novel microscopic technique that enables the easy capture of rotational, 3D-like images of the whole embryo. In this method, a rotary head equipped with two mirrors that are designed to obtain an image tilted at 45 degrees to the microscope stage captures serial images at 2-degree intervals. By a simple operation, 180 images are automatically collected. These 2D images obtained at multiple angles are then used to reconstruct 3D-like images, termed AERO images. By means of this system, over 800 AERO images of 191 gene expression patterns were captured. These images can be easily rotated on the computer screen using the EMBRYS database so that researchers can view an entire embryo by a virtual viewing on a computer screen in an unbiased or non-predetermined manner. The advantages afforded by this approach make it especially useful for generating data viewed in public databases.
Collapse
Affiliation(s)
- Hirohito Shimizu
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Atsushi Kubo
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenta Uchibe
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Megumi Hashimoto
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Shigetoshi Yokoyama
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Shuji Takada
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kazuhiko Mitsuoka
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hiroshi Asahara
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail:
| |
Collapse
|
86
|
Tassy O, Pourquié O. Manteia, a predictive data mining system for vertebrate genes and its applications to human genetic diseases. Nucleic Acids Res 2013; 42:D882-91. [PMID: 24038354 PMCID: PMC3964984 DOI: 10.1093/nar/gkt807] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The function of genes is often evolutionarily conserved, and comparing the annotation of ortholog genes in different model organisms has proved to be a powerful predictive tool to identify the function of human genes. Here, we describe Manteia, a resource available online at http://manteia.igbmc.fr. Manteia allows the comparison of embryological, expression, molecular and etiological data from human, mouse, chicken and zebrafish simultaneously to identify new functional and structural correlations and gene-disease associations. Manteia is particularly useful for the analysis of gene lists produced by high-throughput techniques such as microarrays or proteomics. Data can be easily analyzed statistically to characterize the function of groups of genes and to correlate the different aspects of their annotation. Sophisticated querying tools provide unlimited ways to merge the information contained in Manteia along with the possibility of introducing custom user-designed biological questions into the system. This allows for example to connect all the animal experimental results and annotations to the human genome, and take advantage of data not available for human to look for candidate genes responsible for genetic disorders. Here, we demonstrate the predictive and analytical power of the system to predict candidate genes responsible for human genetic diseases.
Collapse
Affiliation(s)
- Olivier Tassy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch. F-67400, France, Stowers Institute for Medical Research, Kansas City, MO 64110, USA and Howard Hughes Medical Institute, Kansas City, MO 64110, USA
| | | |
Collapse
|
87
|
The snail family gene snai3 is not essential for embryogenesis in mice. PLoS One 2013; 8:e65344. [PMID: 23762348 PMCID: PMC3675094 DOI: 10.1371/journal.pone.0065344] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 04/25/2013] [Indexed: 12/29/2022] Open
Abstract
The Snail gene family encodes zinc finger-containing transcriptional repressor proteins. Three members of the Snail gene family have been described in mammals, encoded by the Snai1, Snai2, and Snai3 genes. The function of the Snai1 and Snai2 genes have been studied extensively during both vertebrate embryogenesis and tumor progression and metastasis, and play critically important roles during these processes. However, little is known about the function of the Snai3 gene and protein. We describe here generation and analysis of Snai3 conditional and null mutant mice. We also generated an EYFP-tagged Snai3 null allele that accurately reflects endogenous Snai3 gene expression, with the highest levels of expression detected in thymus and skeletal muscle. Snai3 null mutant homozygous mice are viable and fertile, and exhibit no obvious phenotypic defects. These results demonstrate that Snai3 gene function is not essential for embryogenesis in mice.
Collapse
|
88
|
Cheung VCK, DeBoer C, Hanson E, Tunesi M, D'Onofrio M, Arisi I, Brandi R, Cattaneo A, Goosens KA. Gene expression changes in the motor cortex mediating motor skill learning. PLoS One 2013; 8:e61496. [PMID: 23637843 PMCID: PMC3634858 DOI: 10.1371/journal.pone.0061496] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 03/08/2013] [Indexed: 12/11/2022] Open
Abstract
The primary motor cortex (M1) supports motor skill learning, yet little is known about the genes that contribute to motor cortical plasticity. Such knowledge could identify candidate molecules whose targeting might enable a new understanding of motor cortical functions, and provide new drug targets for the treatment of diseases which impair motor function, such as ischemic stroke. Here, we assess changes in the motor-cortical transcriptome across different stages of motor skill acquisition. Adult rats were trained on a gradually acquired appetitive reach and grasp task that required different strategies for successful pellet retrieval, or a sham version of the task in which the rats received pellet reward without needing to develop the reach and grasp skill. Tissue was harvested from the forelimb motor-cortical area either before training commenced, prior to the initial rise in task performance, or at peak performance. Differential classes of gene expression were observed at the time point immediately preceding motor task improvement. Functional clustering revealed that gene expression changes were related to the synapse, development, intracellular signaling, and the fibroblast growth factor (FGF) family, with many modulated genes known to regulate synaptic plasticity, synaptogenesis, and cytoskeletal dynamics. The modulated expression of synaptic genes likely reflects ongoing network reorganization from commencement of training till the point of task improvement, suggesting that motor performance improves only after sufficient modifications in the cortical circuitry have accumulated. The regulated FGF-related genes may together contribute to M1 remodeling through their roles in synaptic growth and maturation.
Collapse
Affiliation(s)
- Vincent C. K. Cheung
- McGovern Institute for Brain Research, and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- * E-mail: (VCKC); (KAG)
| | - Caroline DeBoer
- McGovern Institute for Brain Research, and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Elizabeth Hanson
- McGovern Institute for Brain Research, and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Marta Tunesi
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta’, Politecnico di Milano, Milan, Italy
| | - Mara D'Onofrio
- European Brain Research Institute ‘Rita Levi-Montalcini’, Rome, Italy
| | - Ivan Arisi
- European Brain Research Institute ‘Rita Levi-Montalcini’, Rome, Italy
| | - Rossella Brandi
- European Brain Research Institute ‘Rita Levi-Montalcini’, Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute ‘Rita Levi-Montalcini’, Rome, Italy
| | - Ki A. Goosens
- McGovern Institute for Brain Research, and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- * E-mail: (VCKC); (KAG)
| |
Collapse
|
89
|
Chen CK, Symmons O, Uslu VV, Tsujimura T, Ruf S, Smedley D, Spitz F. TRACER: a resource to study the regulatory architecture of the mouse genome. BMC Genomics 2013; 14:215. [PMID: 23547943 PMCID: PMC3618316 DOI: 10.1186/1471-2164-14-215] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 03/22/2013] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Mammalian genes are regulated through the action of multiple regulatory elements, often distributed across large regions. The mechanisms that control the integration of these diverse inputs into specific gene expression patterns are still poorly understood. New approaches enabling the dissection of these mechanisms in vivo are needed. RESULTS Here, we describe TRACER (http://tracerdatabase.embl.de), a resource that centralizes information from a large on-going functional exploration of the mouse genome with different transposon-associated regulatory sensors. Hundreds of insertions have been mapped to specific genomic positions, and their corresponding regulatory potential has been documented by analysis of the expression of the reporter sensor gene in mouse embryos. The data can be easily accessed and provides information on the regulatory activities present in a large number of genomic regions, notably in gene-poor intervals that have been associated with human diseases. CONCLUSIONS TRACER data enables comparisons with the expression pattern of neighbouring genes, activity of surrounding regulatory elements or with other genomic features, revealing the underlying regulatory architecture of these loci. TRACER mouse lines can also be requested for in vivo transposition and chromosomal engineering, to analyse further regions of interest.
Collapse
Affiliation(s)
- Chao-Kung Chen
- European Bioinformatics Institute - European Molecular Biology Laboratory, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | | | | | | | | | | | | |
Collapse
|
90
|
Ohtaka-Maruyama C, Hirai S, Miwa A, Heng JIT, Shitara H, Ishii R, Taya C, Kawano H, Kasai M, Nakajima K, Okado H. RP58 regulates the multipolar-bipolar transition of newborn neurons in the developing cerebral cortex. Cell Rep 2013; 3:458-71. [PMID: 23395638 DOI: 10.1016/j.celrep.2013.01.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 11/16/2012] [Accepted: 01/14/2013] [Indexed: 01/03/2023] Open
Abstract
Accumulating evidence suggests that many brain diseases are associated with defects in neuronal migration, suggesting that this step of neurogenesis is critical for brain organization. However, the molecular mechanisms underlying neuronal migration remain largely unknown. Here, we identified the zinc-finger transcriptional repressor RP58 as a key regulator of neuronal migration via multipolar-to-bipolar transition. RP58(-/-) neurons exhibited severe defects in the formation of leading processes and never shifted to the locomotion mode. Cre-mediated deletion of RP58 using in utero electroporation in RP58(flox/flox) mice revealed that RP58 functions in cell-autonomous multipolar-to-bipolar transition, independent of cell-cycle exit. Finally, we found that RP58 represses Ngn2 transcription to regulate the Ngn2-Rnd2 pathway; Ngn2 knockdown rescued migration defects of the RP58(-/-) neurons. Our findings highlight the critical role of RP58 in multipolar-to-bipolar transition via suppression of the Ngn2-Rnd2 pathway in the developing cerebral cortex.
Collapse
Affiliation(s)
- Chiaki Ohtaka-Maruyama
- Department of Brain Development and Neural Regeneration, Neural Development Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Lamplot JD, Denduluri S, Liu X, Wang J, Yin L, Li R, Shui W, Zhang H, Wang N, Nan G, Angeles J, Shi LL, Haydon RC, Luu HH, Ho S, He TC. Major Signaling Pathways Regulating the Proliferation and Differentiation of Mesenchymal Stem Cells. ESSENTIALS OF MESENCHYMAL STEM CELL BIOLOGY AND ITS CLINICAL TRANSLATION 2013:75-100. [DOI: 10.1007/978-94-007-6716-4_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
92
|
Shin M, Ohte S, Fukuda T, Sasanuma H, Yoneyama K, Kokabu S, Miyamoto A, Tsukamoto S, Hohjoh H, Jimi E, Katagiri T. Identification of a novel bone morphogenetic protein (BMP)-inducible transcript, BMP-inducible transcript-1, by utilizing the conserved BMP-responsive elements in the Id genes. J Bone Miner Metab 2013; 31:34-43. [PMID: 22976053 DOI: 10.1007/s00774-012-0381-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 07/25/2012] [Indexed: 01/15/2023]
Abstract
Bone morphogenetic proteins (BMPs) inhibit myogenesis and induce osteoblastic differentiation in myoblasts. They also induce the transcription of several common genes, such as Id1, Id2 and Id3, in various cell types. We have reported that a GC-rich element in the Id1 gene functions as a BMP-responsive element (BRE) that is regulated by Smads. In this study, we analyzed and identified BREs in the 5'-flanking regions of the mouse Id2 and Id3 genes. The core GGCGCC sequence was conserved among the BREs in the Id1, Id2 and Id3 genes and was essential for the response to BMP signaling via Smads. We found a novel BRE on mouse chromosome 13 at position 47,723,740-47,723,768 by searching for conserved sequences containing the Id1 BRE. This potential BRE was found in the 5'-flanking region of a novel gene that produces a non-coding transcript, termed BMP-inducible transcript-1 (BIT-1), and this element regulated the expression of this gene in response to BMP signaling. We found that BIT-1 is expressed in BMP target tissues such as the testis, brain, kidney and cartilage. These findings suggest that the transcriptional induction of the Ids, BIT-1 and additional novel genes containing the conserved BRE sequence may play an important role in the regulation of the differentiation and/or function of target cells in response to BMPs.
Collapse
Affiliation(s)
- Masashi Shin
- Division of Pathophysiology, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Asahara H. A systems approach reveals that the musculoskeletal tissues development and homeostasis network. Arthritis Res Ther 2012. [PMCID: PMC3332495 DOI: 10.1186/ar3583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
94
|
Farina NH, Hausburg M, Betta ND, Pulliam C, Srivastava D, Cornelison D, Olwin BB. A role for RNA post-transcriptional regulation in satellite cell activation. Skelet Muscle 2012; 2:21. [PMID: 23046558 PMCID: PMC3563611 DOI: 10.1186/2044-5040-2-21] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 08/15/2012] [Indexed: 12/18/2022] Open
Abstract
Background Satellite cells are resident skeletal muscle stem cells responsible for muscle maintenance and repair. In resting muscle, satellite cells are maintained in a quiescent state. Satellite cell activation induces the myogenic commitment factor, MyoD, and cell cycle entry to facilitate transition to a population of proliferating myoblasts that eventually exit the cycle and regenerate muscle tissue. The molecular mechanism involved in the transition of a quiescent satellite cell to a transit-amplifying myoblast is poorly understood. Methods Satellite cells isolated by FACS from uninjured skeletal muscle and 12 h post-muscle injury from wild type and Syndecan-4 null mice were probed using Affymetrix 430v2 gene chips and analyzed by Spotfiretm and Ingenuity Pathway analysis to identify gene expression changes and networks associated with satellite cell activation, respectively. Additional analyses of target genes identify miRNAs exhibiting dynamic changes in expression during satellite cell activation. The function of the miRNAs was assessed using miRIDIAN hairpin inhibitors. Results An unbiased gene expression screen identified over 4,000 genes differentially expressed in satellite cells in vivo within 12 h following muscle damage and more than 50% of these decrease dramatically. RNA binding proteins and genes involved in post-transcriptional regulation were significantly over-represented whereas splicing factors were preferentially downregulated and mRNA stability genes preferentially upregulated. Furthermore, six computationally identified miRNAs demonstrated novel expression through muscle regeneration and in satellite cells. Three of the six miRNAs were found to regulate satellite cell fate. Conclusions The quiescent satellite cell is actively maintained in a state poised to activate in response to external signals. Satellite cell activation appears to be regulated by post-transcriptional gene regulation.
Collapse
Affiliation(s)
- Nicholas H Farina
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO, 80309, USA.
| | | | | | | | | | | | | |
Collapse
|
95
|
A systems approach and skeletal myogenesis. Comp Funct Genomics 2012; 2012:759407. [PMID: 22991503 PMCID: PMC3443578 DOI: 10.1155/2012/759407] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 05/27/2012] [Accepted: 06/28/2012] [Indexed: 12/22/2022] Open
Abstract
Skeletal myogenesis depends on the strict regulation of the expression of various gene subsets. Therefore, the understanding of genome wide gene regulation is imperative for elucidation of skeletal myogenesis. In recent years, systems approach has contributed to the understanding of various biological processes. Our group recently revealed the critical genome network of skeletal myogenesis by using a novel systems approach combined with whole-mount in situ hybridization (WISH) database, high-throughput screening, and microarray analysis. In this paper, we introduce our systems approach for understanding the myogenesis regulatory network and describe the advantages of systems approach.
Collapse
|
96
|
Geffers L, Herrmann B, Eichele G. Web-based digital gene expression atlases for the mouse. Mamm Genome 2012; 23:525-38. [PMID: 22936000 DOI: 10.1007/s00335-012-9413-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 07/04/2012] [Indexed: 11/26/2022]
Abstract
Over the past 15 years the publicly available mouse gene expression data determined by in situ hybridization have dramatically increased in scope and spatiotemporal resolution. As a consequence of resources and tools available in the post-genomic era, full transcriptomes in the mouse brain and in the mouse embryo can be studied. Here we introduce and discuss seven current databases (MAMEP, EMBRYS, GenePaint, EURExpress, EuReGene, BGEM, and GENSAT) that grant access to large collections of expression data in mouse. We review the experimental focus, coverage, data assessment, and annotation for each of these databases and the implementation of analytic tools and links to other relevant databases. We provide a user-oriented summary of how to interrogate each database.
Collapse
Affiliation(s)
- Lars Geffers
- Department of Genes and Behavior, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany.
| | | | | |
Collapse
|
97
|
Uchibe K, Shimizu H, Yokoyama S, Kuboki T, Asahara H. Identification of novel transcription-regulating genes expressed during murine molar development. Dev Dyn 2012; 241:1217-26. [PMID: 22639370 DOI: 10.1002/dvdy.23808] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The mechanism of tooth development is a complex process regulated by numerous genes including transcription factors, growth factors, and other intra- and extracellular molecules. Especially, transcription factors play a central role in gene expression, regulating a wide spectrum of biological processes including organogenesis. Substantial evidence has been demonstrated by a number of studies using genetically engineered animal models. However, detailed molecular mechanisms of tooth development have not been completely elucidated, partially because numerous genes that play essential roles in tooth development remain unidentified. RESULTS In this study, we conducted an expression-based screening using gene expression database and in situ hybridization assays. Based on the gene expression database "EMBRYS," 207 out of 1,520 genes were expressed in the maxillary and/or mandibular processes and thus were selected for further analysis by section in situ hybridization. Among these candidates, 28 genes were newly identified as potential factors associated with tooth development by in situ hybridization assays with frontal sections of embryonic day 13.5 and 14.5 mouse embryos. The expression patterns were also examined at embryonic day 16.5 and 18.5. CONCLUSIONS These results will contribute to elucidating the mechanisms of tooth development and to improving the technology for regeneration of tooth.
Collapse
Affiliation(s)
- Kenta Uchibe
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Setagaya, Japan
| | | | | | | | | |
Collapse
|
98
|
Abstract
A dual role of transforming growth factor β (TGF-β), to both suppress and promote tumor progression and metastasis, has been well established, but its molecular basis has remained elusive. In this review, we focus on Smad proteins, which are central mediators of the signal transduction of TGF-β family members. We describe current knowledge of cell-type-specific binding patterns of Smad proteins and mechanisms of transcriptional regulation, obtained from recent studies on genome-wide binding sites of Smad molecules. We also discuss potential application of the genome-wide analyses for cancer research, which will allow clarification of the complex mechanisms occurring during cancer progression, and the identification of potential biomarkers for future cancer diagnosis, prognosis and therapy.
Collapse
Affiliation(s)
- M Morikawa
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | | | | | | |
Collapse
|
99
|
Macquarrie KL, Yao Z, Young JM, Cao Y, Tapscott SJ. miR-206 integrates multiple components of differentiation pathways to control the transition from growth to differentiation in rhabdomyosarcoma cells. Skelet Muscle 2012; 2:7. [PMID: 22541669 PMCID: PMC3417070 DOI: 10.1186/2044-5040-2-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 04/29/2012] [Indexed: 01/02/2023] Open
Abstract
Background Similar to replicating myoblasts, many rhabdomyosarcoma cells express the myogenic determination gene MyoD. In contrast to myoblasts, rhabdomyosarcoma cells do not make the transition from a regulative growth phase to terminal differentiation. Previously we demonstrated that the forced expression of MyoD with its E-protein dimerization partner was sufficient to induce differentiation and suppress multiple growth-promoting genes, suggesting that the dimer was targeting a switch that regulated the transition from growth to differentiation. Our data also suggested that a balance between various inhibitory transcription factors and MyoD activity kept rhabdomyosarcomas trapped in a proliferative state. Methods Potential myogenic co-factors were tested for their ability to drive differentiation in rhabdomyosarcoma cell culture models, and their relation to MyoD activity determined through molecular biological experiments. Results Modulation of the transcription factors RUNX1 and ZNF238 can induce differentiation in rhabdomyosarcoma cells and their activity is integrated, at least in part, through the activation of miR-206, which acts as a genetic switch to transition the cell from a proliferative growth phase to differentiation. The inhibitory transcription factor MSC also plays a role in controlling miR-206, appearing to function by occluding a binding site for MyoD in the miR-206 promoter. Conclusions These findings support a network model composed of coupled regulatory circuits with miR-206 functioning as a switch regulating the transition from one stable state (growth) to another (differentiation).
Collapse
Affiliation(s)
- Kyle L Macquarrie
- Human Biology Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, C3-168, Seattle, WA, 98109, USA.
| | | | | | | | | |
Collapse
|
100
|
Saccone V, Puri PL. Epigenetic regulation of skeletal myogenesis. Organogenesis 2012; 6:48-53. [PMID: 20592865 DOI: 10.4161/org.6.1.11293] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2010] [Revised: 01/24/2010] [Accepted: 01/25/2010] [Indexed: 12/31/2022] Open
Abstract
During embryogenesis a timely and coordinated expression of different subsets of genes drives the formation of skeletal muscles in response to developmental cues. In this review, we will summarize the most recent advances on the "epigenetic network" that promotes the transcription of selective groups of genes in muscle progenitors, through the concerted action of chromatin-associated complexes that modify histone tails and microRNAs (miRNAs). These epigenetic players cooperate to establish focal domains of euchromatin, which facilitates gene transcription, and large portions of heterochromatin, which precludes inappropriate gene expression. We also discuss the analogies and differences in the transcriptional and the epigenetic networks driving developmental and adult myogenesis. The elucidation of the epigenetic basis controlling skeletal myogenesis during development and adult life will facilitate experimental strategies toward generating muscle stem cells, either by reprogramming embryonic stem cells or by inducing pluripotency in adult skeletal muscles. During embryogenesis a timely and coordinated expression of different subsets of genes drives the formation of skeletal muscles in response to developmental cues. In this review, we will summarize the most recent advances on the "epigenetic network" that promotes the transcription of selective groups of genes in muscle progenitors, through the concerted action of chromatin-associated complexes that modify histone tails and microRNAs (miRNAs). These epigenetic players cooperate to establish focal domains of euchromatin, which facilitates gene transcription, and large portions of heterochromatin, which precludes inappropriate gene expression. We also discuss the analogies and differences in the transcriptional and the epigenetic networks driving developmental and adult myogenesis. The elucidation of the epigenetic basis controlling skeletal myogenesis during development and adult life will facilitate experimental strategies toward generating muscle stem cells, either by reprogramming embryonic stem cells or by inducing pluripotency in adult skeletal muscles.
Collapse
Affiliation(s)
- Valentina Saccone
- Istituto Dulbecco Telethon, IR CCS Santa Lucia Foundation and European Brain Research Institute, Rome, Italy
| | | |
Collapse
|