201
|
PAK1 and CtBP1 Regulate the Coupling of Neuronal Activity to Muscle Chromatin and Gene Expression. Mol Cell Biol 2015; 35:4110-20. [PMID: 26416879 DOI: 10.1128/mcb.00354-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 09/01/2015] [Indexed: 11/20/2022] Open
Abstract
Acetylcholine receptor (AChR) expression in innervated muscle is limited to the synaptic region. Neuron-induced electrical activity participates in this compartmentalization by promoting the repression of AChR expression in the extrasynaptic regions. Here, we show that the corepressor CtBP1 (C-terminal binding protein 1) is present on the myogenin promoter together with repressive histone marks. shRNA-mediated downregulation of CtBP1 expression is sufficient to derepress myogenin and AChR expression in innervated muscle. Upon denervation, CtBP1 is displaced from the myogenin promoter and relocates to the cytoplasm, while repressive histone marks are replaced by activating ones concomitantly to the activation of myogenin expression. We also observed that upon denervation the p21-activated kinase 1 (PAK1) expression is upregulated, suggesting that phosphorylation by PAK1 may be involved in the relocation of CtBP1. Indeed, preventing CtBP1 Ser158 phosphorylation induces CtBP1 accumulation in the nuclei and abrogates the activation of myogenin and AChR expression. Altogether, these findings reveal a molecular mechanism to account for the coordinated control of chromatin modifications and muscle gene expression by presynaptic neurons via a PAK1/CtBP1 pathway.
Collapse
|
202
|
Association of lamin A/C with muscle gene-specific promoters in myoblasts. Biochem Biophys Rep 2015; 4:76-82. [PMID: 29124189 PMCID: PMC5668900 DOI: 10.1016/j.bbrep.2015.08.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 08/10/2015] [Accepted: 08/26/2015] [Indexed: 11/20/2022] Open
Abstract
The A-type and B-type lamins form a filamentous meshwork underneath the inner nuclear membrane called the nuclear lamina, which is an important component of nuclear architecture in metazoan cells. The lamina interacts with large, mostly repressive chromatin domains at the nuclear periphery. In addition, genome–lamina interactions also involve dynamic association of lamin A/C with gene promoters in adipocytes. Mutations in the human lamin A gene cause a spectrum of hereditary diseases called the laminopathies which affect muscle, cardiac and adipose tissues. Since most mutations in lamin A/C affect skeletal muscle, we investigated lamin–chromatin interactions at promoters of muscle specific genes in both muscle and non-muscle cell lines by ChIP-qPCR. We observed that lamin A/C was specifically associated with promoter regions of muscle genes in myoblasts but not in fibroblasts. Lamin A/C dissociated from the promoter regions of the differentiation specific MyoD, myogenin and muscle creatine kinase genes when myoblasts were induced to differentiate. In the promoter regions of the myogenin and MyoD genes, the binding of lamin A/C in myoblasts inversely correlated with the active histone mark, H3K4me3. Lamin A/C binding on muscle genes was reduced and differentiation potential was enhanced on treatment of myoblasts with a histone deacetylase inhibitor. These findings suggest a role for lamina–chromatin interactions in muscle differentiation and have important implications for the pathological mechanisms of striated muscle associated laminopathies. Lamina–chromatin interactions are important for nuclear processes. We show lamin A/C binding to promoters of muscle genes in myoblasts. Lamin A/C binding is reduced upon myoblast differentiation. Lamin A/C binding inversely correlates with active histone marks on muscle genes. Our findings suggest that lamin A/C binding to promoters is cell-type specific.
Collapse
|
203
|
Soibam B, Benham A, Kim J, Weng KC, Yang L, Xu X, Robertson M, Azares A, Cooney AJ, Schwartz RJ, Liu Y. Genome-Wide Identification of MESP1 Targets Demonstrates Primary Regulation Over Mesendoderm Gene Activity. Stem Cells 2015. [PMID: 26205879 DOI: 10.1002/stem.2111] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
MESP1 is considered the first sign of the nascent cardiac mesoderm and plays a critical role in the appearance of cardiac progenitors, while exhibiting a transient expression in the developing embryo. We profiled the transcriptome of a pure population of differentiating MESP1-marked cells and found that they chiefly contribute to the mesendoderm lineage. High-throughput sequencing of endogenous MESP1-bound DNA revealed that MESP1 preferentially binds to two variants of E-box sequences and activates critical mesendoderm modulators, including Eomes, Gata4, Wnt5a, Wnt5b, Mixl1, T, Gsc, and Wnt3. These mesendoderm markers were enriched in the MESP1 marked population before the appearance of cardiac progenitors and myocytes. Further, MESP1-binding is globally associated with H(3)K(27) acetylation, supporting a novel pivotal role of it in regulating target gene epigenetics. Therefore, MESP1, the pioneer cardiac factor, primarily directs the appearance of mesendoderm, the intermediary of the earliest progenitors of mesoderm and endoderm organogenesis.
Collapse
Affiliation(s)
- Benjamin Soibam
- Texas Heart Institute, Texas Medical Center, Houston, Texas, USA.,Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Ashley Benham
- Texas Heart Institute, Texas Medical Center, Houston, Texas, USA
| | - Jong Kim
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Kuo-Chan Weng
- The Institute of Biosciences and Technology, Texas A & M University Health Science Center, Houston, Texas, USA
| | - Litao Yang
- Texas Heart Institute, Texas Medical Center, Houston, Texas, USA
| | - Xueping Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | | | - Alon Azares
- Texas Heart Institute, Texas Medical Center, Houston, Texas, USA
| | - Austin J Cooney
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Robert J Schwartz
- Texas Heart Institute, Texas Medical Center, Houston, Texas, USA.,Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Yu Liu
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| |
Collapse
|
204
|
Chandra S, Terragni J, Zhang G, Pradhan S, Haushka S, Johnston D, Baribault C, Lacey M, Ehrlich M. Tissue-specific epigenetics in gene neighborhoods: myogenic transcription factor genes. Hum Mol Genet 2015; 24:4660-73. [PMID: 26041816 PMCID: PMC4512632 DOI: 10.1093/hmg/ddv198] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/18/2015] [Accepted: 05/26/2015] [Indexed: 12/15/2022] Open
Abstract
Myogenic regulatory factor (MRF) genes, MYOD1, MYOG, MYF6 and MYF5, are critical for the skeletal muscle lineage. Here, we used various epigenome profiles from human myoblasts (Mb), myotubes (Mt), muscle and diverse non-muscle samples to elucidate the involvement of multigene neighborhoods in the regulation of MRF genes. We found more far-distal enhancer chromatin associated with MRF genes in Mb and Mt than previously reported from studies in mice. For the MYF5/MYF6 gene-pair, regions of Mb-associated enhancer chromatin were located throughout the adjacent 236-kb PTPRQ gene even though Mb expressed negligible amounts of PTPRQ mRNA. Some enhancer chromatin regions inside PTPRQ in Mb were also seen in PTPRQ mRNA-expressing non-myogenic cells. This suggests dual-purpose PTPRQ enhancers that upregulate expression of PTPRQ in non-myogenic cells and MYF5/MYF6 in myogenic cells. In contrast, the myogenic enhancer chromatin regions distal to MYOD1 were intergenic and up to 19 kb long. Two of them contain small, known MYOD1 enhancers, and one displayed an unusually high level of 5-hydroxymethylcytosine in a quantitative DNA hydroxymethylation assay. Unexpectedly, three regions of MYOD1-distal enhancer chromatin in Mb and Mt overlapped enhancer chromatin in umbilical vein endothelial cells, which might upregulate a distant gene (PIK3C2A). Lastly, genes surrounding MYOG were preferentially transcribed in Mt, like MYOG itself, and exhibited nearby myogenic enhancer chromatin. These neighboring chromatin regions may be enhancers acting in concert to regulate myogenic expression of multiple adjacent genes. Our findings reveal the very different and complex organization of gene neighborhoods containing closely related transcription factor genes.
Collapse
Affiliation(s)
- Sruti Chandra
- Program in Human Genetics, Tulane Cancer Center, and Center for Bioinformatics and Genomics, Tulane Health Sciences Center, New Orleans, LA 70122, USA
| | | | | | | | - Stephen Haushka
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA, and
| | - Douglas Johnston
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, LA 70112, USA
| | - Carl Baribault
- Tulane Cancer Center and Department of Mathematics, Tulane Health Sciences Center and Tulane University, New Orleans, LA 70122, USA
| | - Michelle Lacey
- Tulane Cancer Center and Department of Mathematics, Tulane Health Sciences Center and Tulane University, New Orleans, LA 70122, USA
| | - Melanie Ehrlich
- Program in Human Genetics, Tulane Cancer Center, and Center for Bioinformatics and Genomics, Tulane Health Sciences Center, New Orleans, LA 70122, USA,
| |
Collapse
|
205
|
Umansky KB, Gruenbaum-Cohen Y, Tsoory M, Feldmesser E, Goldenberg D, Brenner O, Groner Y. Runx1 Transcription Factor Is Required for Myoblasts Proliferation during Muscle Regeneration. PLoS Genet 2015; 11:e1005457. [PMID: 26275053 PMCID: PMC4537234 DOI: 10.1371/journal.pgen.1005457] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/21/2015] [Indexed: 01/09/2023] Open
Abstract
Following myonecrosis, muscle satellite cells proliferate, differentiate and fuse, creating new myofibers. The Runx1 transcription factor is not expressed in naïve developing muscle or in adult muscle tissue. However, it is highly expressed in muscles exposed to myopathic damage yet, the role of Runx1 in muscle regeneration is completely unknown. Our study of Runx1 function in the muscle’s response to myonecrosis reveals that this transcription factor is activated and cooperates with the MyoD and AP-1/c-Jun transcription factors to drive the transcription program of muscle regeneration. Mice lacking dystrophin and muscle Runx1 (mdx-/Runx1f/f), exhibit impaired muscle regeneration leading to age-dependent muscle waste, gradual decrease in motor capabilities and a shortened lifespan. Runx1-deficient primary myoblasts are arrested at cell cycle G1 and consequently differentiate. Such premature differentiation disrupts the myoblasts’ normal proliferation/differentiation balance, reduces the number and size of regenerating myofibers and impairs muscle regeneration. Our combined Runx1-dependent gene expression, ChIP-seq, ATAC-seq and histone H3K4me1/H3K27ac modification analyses revealed a subset of Runx1-regulated genes that are co-occupied by MyoD and c-Jun in mdx-/Runx1f/f muscle. The data provide unique insights into the transcriptional program driving muscle regeneration and implicate Runx1 as an important participant in the pathology of muscle wasting diseases. In response to muscle injury, the muscle initiates a repair process that calls for the proliferation of muscle stem cells, which differentiate and fuse to create the myofibers that regenerate the tissue. Maintaining the balance between myoblast proliferation and differentiation is crucial for proper regeneration, with disruption leading to impaired regeneration characteristic of muscle-wasting diseases. Our study highlights the important role the Runx1 transcription factor plays in muscle regeneration and in regulating the balance between muscle stem cell proliferation and differentiation. While not expressed in healthy muscle tissue, Runx1 level significantly increases in response to various types of muscle damage. This aligns with our finding that mice lacking Runx1 in their muscles suffer from impaired muscle regeneration. Their muscles contained a significantly low number of regenerating myofibers, which were also relatively smaller in size, resulting in loss of muscle mass and motor capabilities. Our results indicate that Runx1 regulates muscle regeneration by preventing premature differentiation of proliferating myoblasts, thereby facilitating the buildup of the myoblast pool required for proper regeneration. Through genome-wide gene-expression analysis we identify a set of Runx1-regulated genes responsible for muscle regeneration thereby implicating Runx1 in the pathology of muscle wasting diseases such as Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Kfir Baruch Umansky
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Yael Gruenbaum-Cohen
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, The Weizmann Institute of Science, Rehovot, Israel
| | - Ester Feldmesser
- Grand Israel National Center for Personalized Medicine (INCPM), The Weizmann Institute of Science, Rehovot, Israel
| | - Dalia Goldenberg
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Ori Brenner
- Department of Veterinary Resources, The Weizmann Institute of Science, Rehovot, Israel
| | - Yoram Groner
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
206
|
Albini S, Coutinho Toto P, Dall'Agnese A, Malecova B, Cenciarelli C, Felsani A, Caruso M, Bultman SJ, Puri PL. Brahma is required for cell cycle arrest and late muscle gene expression during skeletal myogenesis. EMBO Rep 2015; 16:1037-1050. [PMID: 26136374 PMCID: PMC4552495 DOI: 10.15252/embr.201540159] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 05/21/2015] [Accepted: 05/25/2015] [Indexed: 02/03/2023] Open
Abstract
Although the two catalytic subunits of the SWI/SNF chromatin-remodeling complex--Brahma (Brm) and Brg1--are almost invariably co-expressed, their mutually exclusive incorporation into distinct SWI/SNF complexes predicts that Brg1- and Brm-based SWI/SNF complexes execute specific functions. Here, we show that Brg1 and Brm have distinct functions at discrete stages of muscle differentiation. While Brg1 is required for the activation of muscle gene transcription at early stages of differentiation, Brm is required for Ccnd1 repression and cell cycle arrest prior to the activation of muscle genes. Ccnd1 knockdown rescues the ability to exit the cell cycle in Brm-deficient myoblasts, but does not recover terminal differentiation, revealing a previously unrecognized role of Brm in the activation of late muscle gene expression independent from the control of cell cycle. Consistently, Brm null mice displayed impaired muscle regeneration after injury, with aberrant proliferation of satellite cells and delayed formation of new myofibers. These data reveal stage-specific roles of Brm during skeletal myogenesis, via formation of repressive and activatory SWI/SNF complexes.
Collapse
Affiliation(s)
- Sonia Albini
- Sanford-Burnham Institute for Medical Research, La Jolla, CA, USA
| | | | | | - Barbora Malecova
- Sanford-Burnham Institute for Medical Research, La Jolla, CA, USA
| | | | - Armando Felsani
- CNR-Istituto di Biologia Cellulare e Neurobiologia Fondazione Santa Lucia, Rome, Italy
| | - Maurizia Caruso
- CNR-Istituto di Biologia Cellulare e Neurobiologia Fondazione Santa Lucia, Rome, Italy
| | - Scott J Bultman
- Department of Genetics, Lineberger Comprehensive Cancer Center University of North Carolina, Chapel Hill, NC, USA
| | - Pier Lorenzo Puri
- Sanford-Burnham Institute for Medical Research, La Jolla, CA, USA IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
207
|
Chakroun I, Yang D, Girgis J, Gunasekharan A, Phenix H, Kærn M, Blais A. Genome-wide association between Six4, MyoD, and the histone demethylase Utx during myogenesis. FASEB J 2015; 29:4738-55. [PMID: 26229056 DOI: 10.1096/fj.15-277053] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/20/2015] [Indexed: 01/14/2023]
Abstract
Adult skeletal muscles can regenerate after injury, due to the presence of satellite cells, a quiescent population of myogenic progenitor cells. Once activated, satellite cells repair the muscle damage by undergoing myogenic differentiation. The myogenic regulatory factors (MRFs) coordinate the process of progenitor differentiation in cooperation with other families of transcription factors (TFs). The Six1 and Six4 homeodomain TFs are expressed in developing and adult muscle and Six1 is critical for embryonic and adult myogenesis. However, the lack of a muscle developmental phenotype in Six4-null mice, which has been attributed to compensation by other Six family members, has discouraged further assessment of the role of Six4 during adult muscle regeneration. By employing genome-wide approaches to address the function of Six4 during adult skeletal myogenesis, we have identified a core set of muscle genes coordinately regulated in adult muscle precursors by Six4 and the MRF MyoD. Throughout the genome of differentiating adult myoblasts, the cooperation between Six4 and MyoD is associated with chromatin repressive mark removal by Utx, a demethylase of histone H3 trimethylated at lysine 27. Among the genes coordinately regulated by Six4 and MyoD are several genes critical for proper in vivo muscle regeneration, implicating a role of Six4 in this process. Using in vivo RNA interference of Six4, we expose an uncompensated function of this TF during muscle regeneration. Together, our results reveal a role for Six4 during adult muscle regeneration and suggest a widespread mechanism of cooperation between Six4 and MyoD.
Collapse
Affiliation(s)
- Imane Chakroun
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Dabo Yang
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - John Girgis
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Atchayaa Gunasekharan
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Hilary Phenix
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Mads Kærn
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Alexandre Blais
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
208
|
Abstract
Transcription factors (TFs) are multidomain proteins that play a critical role in orchestrating stem cell differentiation, but several limitations hinder the full potential of TF-based gene regulation. Here we report a unique strategy to emulate TFs and differentiate stem cells in a nonviral approach using an artificial, nanoparticle-based transcription factor called NanoScript. The NanoScript platform consists of a gold nanoparticle functionalized with small molecules that mimic the various domains of TFs. As a result, NanoScript mimics the function and structure of TF proteins. Specifically, NanoScript was designed to regulate muscle cell differentiation by targeting myogenic regulatory factors (MRFs), which play an important role in inducing myogenesis. This NanoScript-MRF is stable in physiological environments, localizes within the nucleus, induces differentiation of adipose-derived mesenchymal stem cells into mature muscle cells in 7 days, and is naturally excreted from induced muscle cells. As such, NanoScript represents a safe and powerful tool for applications requiring gene manipulation.
Collapse
Affiliation(s)
- Sahishnu Patel
- Department of Chemistry and Chemical Biology, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Perry T. Yin
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Hiroshi Sugiyama
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8501, Japan
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8501, Japan
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, The State University of New Jersey, Piscataway, New Jersey 08854, United States
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
209
|
|
210
|
Abstract
The 4D organization of the interphase nucleus, or the 4D Nucleome (4DN), reflects a dynamical interaction between 3D genome structure and function and its relationship to phenotype. We present initial analyses of the human 4DN, capturing genome-wide structure using chromosome conformation capture and 3D imaging, and function using RNA-sequencing. We introduce a quantitative index that measures underlying topological stability of a genomic region. Our results show that structural features of genomic regions correlate with function with surprising persistence over time. Furthermore, constructing genome-wide gene-level contact maps aided in identifying gene pairs with high potential for coregulation and colocalization in a manner consistent with expression via transcription factories. We additionally use 2D phase planes to visualize patterns in 4DN data. Finally, we evaluated gene pairs within a circadian gene module using 3D imaging, and found periodicity in the movement of clock circadian regulator and period circadian clock 2 relative to each other that followed a circadian rhythm and entrained with their expression.
Collapse
|
211
|
White MA. Understanding how cis-regulatory function is encoded in DNA sequence using massively parallel reporter assays and designed sequences. Genomics 2015; 106:165-170. [PMID: 26072432 DOI: 10.1016/j.ygeno.2015.06.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 05/09/2015] [Accepted: 06/08/2015] [Indexed: 01/07/2023]
Abstract
Genome-scale methods have identified thousands of candidate cis-regulatory elements (CREs), but methods to directly assay the regulatory function of these elements on a comparably large scale have not been available. The inability to directly test and perturb the regulatory activity of large numbers of DNA sequences has hindered efforts to discover how cis-regulatory function is encoded in genomic sequence. Recently developed massively parallel reporter gene assays combine next generation sequencing with high-throughput oligonucleotide synthesis to offer the capacity to test and mutationally perturb thousands of specifically chosen or designed cis-regulatory sequences in a single experiment. These assays are the basis of recent studies that include large-scale functional validation of genomic CREs, exhaustive mutational analyses of individual regulatory sequences, and tests of large libraries of synthetic CREs. The results demonstrate how massively parallel reporter assays with libraries of designed sequences provide the statistical power required to address previously intractable questions about cis-regulatory function.
Collapse
Affiliation(s)
- Michael A White
- Center for Genome Sciences and Systems Biology, Department of Genetics, Washington University in St. Louis School of Medicine, St. Louis, MO 63108, USA.
| |
Collapse
|
212
|
Gibson TM, Gersbach CA. Single-molecule analysis of myocyte differentiation reveals bimodal lineage commitment. Integr Biol (Camb) 2015; 7:663-71. [PMID: 25953198 PMCID: PMC4461500 DOI: 10.1039/c5ib00057b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Cell differentiation is the foundation for tissue development and regeneration, disease modeling, and cell-based therapies. Although the differentiation of cell populations has been extensively studied in many systems, much less is known about the distribution of decision making of single cells within these populations. To characterize the differentiation of single skeletal muscle cells, we used single-molecule mRNA fluorescence in situ hybridization (smFISH) to precisely quantify the expression levels of the master myogenic regulatory factors MyoD and myogenin in individual myoblasts. We identified distinct cell states characterized by the number of myogenin transcripts expressed by a cell, with myoblasts stochastically transitioning to a myogenin-high state during differentiation. We also used MyoD overexpression to force the transdifferentiation of C3H10T1/2 cells into an induced myoblast phenotype. These reprogrammed cells revealed the presence of a critical threshold of MyoD expression required to initiate myogenin expression. These results provide quantitative single-molecule data to support the model of switch-like cell decision making and lineage specification.
Collapse
Affiliation(s)
- Tyler M Gibson
- Department of Biomedical Engineering, Duke University, Room 136 Hudson Hall, Box 90281, Durham, North Carolina 27708, USA.
| | | |
Collapse
|
213
|
Blais A. Myogenesis in the Genomics Era. J Mol Biol 2015; 427:2023-38. [DOI: 10.1016/j.jmb.2015.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/04/2015] [Accepted: 02/05/2015] [Indexed: 01/06/2023]
|
214
|
Abstract
Despite improvement in clinical treatment of childhood cancer, it remains the leading cause of disease-related mortality in children with survivors often suffering from treatment-related toxicity and premature death. Because childhood cancer is vastly different from cancer in adults, a thorough understanding of the underlying molecular mechanisms specific to childhood cancer is essential. Although childhood cancer contains much fewer mutations, a subset of cancer subtypes has a higher frequency of mutations in gene encoding epigenetic regulators. Thus, in this review, we will focus on epigenetic deregulations in childhood cancers, the use of genome-wide analysis for cancer subtype classification, prediction of clinical outcomes and the influence of folate on epigenetic mechanisms.
Collapse
Affiliation(s)
- Teresa T Yiu
- Department of Pediatrics, Texas Children's Cancer Center, 6701 Fannin St, Ste 1400, Houston, TX 77030, USA.,Dan L Duncan Cancer Center, 1 Baylor Plaza 450A, Houston, TX 77030, USA
| | - Wei Li
- Dan L Duncan Cancer Center, 1 Baylor Plaza 450A, Houston, TX 77030, USA.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
215
|
Wang E, Kawaoka S, Roe JS, Shi J, Hohmann AF, Xu Y, Bhagwat AS, Suzuki Y, Kinney JB, Vakoc CR. The transcriptional cofactor TRIM33 prevents apoptosis in B lymphoblastic leukemia by deactivating a single enhancer. eLife 2015; 4:e06377. [PMID: 25919951 PMCID: PMC4409649 DOI: 10.7554/elife.06377] [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: 01/07/2015] [Accepted: 03/30/2015] [Indexed: 11/13/2022] Open
Abstract
Most mammalian transcription factors (TFs) and cofactors occupy thousands of genomic sites and modulate the expression of large gene networks to implement their biological functions. In this study, we describe an exception to this paradigm. TRIM33 is identified here as a lineage dependency in B cell neoplasms and is shown to perform this essential function by associating with a single cis element. ChIP-seq analysis of TRIM33 in murine B cell leukemia revealed a preferential association with two lineage-specific enhancers that harbor an exceptional density of motifs recognized by the PU.1 TF. TRIM33 is recruited to these elements by PU.1, yet acts to antagonize PU.1 function. One of the PU.1/TRIM33 co-occupied enhancers is upstream of the pro-apoptotic gene Bim, and deleting this enhancer renders TRIM33 dispensable for leukemia cell survival. These findings reveal an essential role for TRIM33 in preventing apoptosis in B lymphoblastic leukemia by interfering with enhancer-mediated Bim activation. DOI:http://dx.doi.org/10.7554/eLife.06377.001 The DNA inside every cell in a human body is the same, and yet the activities that occur within different types of cells can vary greatly. White blood cells, for example, are different from skin cells or liver cells because different genes are active in each type of cell. Molecules called transcription factors and transcriptional cofactors associate with specific DNA sequences to control the activity of nearby genes. It is common for a single transcription factor or cofactor to bind to thousands of sites across the DNA of any cell. In humans, our immune systems protect us against infectious diseases and from malfunctioning cells that could become cancerous. White blood cells called B cells provide part of this immune defense. These cells help to identify invading bacteria and viruses, and can also develop into memory cells that help the immune system to rapidly recognize, respond to and eliminate a disease if it is re-encountered. Immature B cells—also known as B lymphoblasts—mature within bone marrow. If any problem occurs in a cell as it matures, that cell is usually programmed to self-destruct in a process called apoptosis. If these cells are not destroyed, they can accumulate in the bone marrow and prevent the production of other immune cells. This leads to a type of cancer called acute lymphoblastic leukemia. Wang et al. now reveal that TRIM33—a protein that B-lymphoid leukemia cells need to survive—is a transcriptional cofactor that prevents apoptosis. Furthermore, unlike other known transcription factors and cofactors in mammals, TRIM33 binds to an exceedingly small number of sites across the DNA of B cells. In fact, the cancer cell's dependency on the protein is due to TRIM33 associating with just a single binding site. The role of TRIM33 in B cell leukemia also has potential therapeutic implications. Although it is found in cells throughout the body, Wang et al. found that inhibiting TRIM33 in mice resulted in lower numbers of B cells being produced, but did not affect other tissues. Developing drugs that prevent TRIM33 from working could therefore provide new options for treating leukemia. DOI:http://dx.doi.org/10.7554/eLife.06377.002
Collapse
Affiliation(s)
- Eric Wang
- Cold Spring Harbor Laboratory, New York, United States
| | | | - Jae-Seok Roe
- Cold Spring Harbor Laboratory, New York, United States
| | - Junwei Shi
- Cold Spring Harbor Laboratory, New York, United States
| | | | - Yali Xu
- Cold Spring Harbor Laboratory, New York, United States
| | | | - Yutaka Suzuki
- Department of Medical Genome Sciences, University of Tokyo, Kashiwa, Japan
| | | | | |
Collapse
|
216
|
Dogan N, Wu W, Morrissey CS, Chen KB, Stonestrom A, Long M, Keller CA, Cheng Y, Jain D, Visel A, Pennacchio LA, Weiss MJ, Blobel GA, Hardison RC. Occupancy by key transcription factors is a more accurate predictor of enhancer activity than histone modifications or chromatin accessibility. Epigenetics Chromatin 2015; 8:16. [PMID: 25984238 PMCID: PMC4432502 DOI: 10.1186/s13072-015-0009-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 04/02/2015] [Indexed: 12/12/2022] Open
Abstract
Background Regulated gene expression controls organismal development, and variation in regulatory patterns has been implicated in complex traits. Thus accurate prediction of enhancers is important for further understanding of these processes. Genome-wide measurement of epigenetic features, such as histone modifications and occupancy by transcription factors, is improving enhancer predictions, but the contribution of these features to prediction accuracy is not known. Given the importance of the hematopoietic transcription factor TAL1 for erythroid gene activation, we predicted candidate enhancers based on genomic occupancy by TAL1 and measured their activity. Contributions of multiple features to enhancer prediction were evaluated based on the results of these and other studies. Results TAL1-bound DNA segments were active enhancers at a high rate both in transient transfections of cultured cells (39 of 79, or 56%) and transgenic mice (43 of 66, or 65%). The level of binding signal for TAL1 or GATA1 did not help distinguish TAL1-bound DNA segments as active versus inactive enhancers, nor did the density of regulation-related histone modifications. A meta-analysis of results from this and other studies (273 tested predicted enhancers) showed that the presence of TAL1, GATA1, EP300, SMAD1, H3K4 methylation, H3K27ac, and CAGE tags at DNase hypersensitive sites gave the most accurate predictors of enhancer activity, with a success rate over 80% and a median threefold increase in activity. Chromatin accessibility assays and the histone modifications H3K4me1 and H3K27ac were sensitive for finding enhancers, but they have high false positive rates unless transcription factor occupancy is also included. Conclusions Occupancy by key transcription factors such as TAL1, GATA1, SMAD1, and EP300, along with evidence of transcription, improves the accuracy of enhancer predictions based on epigenetic features. Electronic supplementary material The online version of this article (doi:10.1186/s13072-015-0009-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nergiz Dogan
- Center for Comparative Genomics and Bioinformatics, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, 304 Wartik Laboratory, University Park, PA 16802 USA
| | - Weisheng Wu
- Center for Comparative Genomics and Bioinformatics, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, 304 Wartik Laboratory, University Park, PA 16802 USA ; Bioinformatics Core, Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109-2218 USA
| | - Christapher S Morrissey
- Center for Comparative Genomics and Bioinformatics, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, 304 Wartik Laboratory, University Park, PA 16802 USA
| | - Kuan-Bei Chen
- Center for Comparative Genomics and Bioinformatics, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, 304 Wartik Laboratory, University Park, PA 16802 USA
| | - Aaron Stonestrom
- Division of Hematology, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104 USA ; Perelman School of Medicine at the University of Pennsylvania, 415 Curie Boulevard, Philadelphia, PA 19104 USA
| | - Maria Long
- Center for Comparative Genomics and Bioinformatics, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, 304 Wartik Laboratory, University Park, PA 16802 USA
| | - Cheryl A Keller
- Center for Comparative Genomics and Bioinformatics, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, 304 Wartik Laboratory, University Park, PA 16802 USA
| | - Yong Cheng
- Department of Genetics, Mail Stop-5120, Stanford University, Stanford, CA 94305 USA
| | - Deepti Jain
- Center for Comparative Genomics and Bioinformatics, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, 304 Wartik Laboratory, University Park, PA 16802 USA
| | - Axel Visel
- Genomics Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Mailstop 84-171, Berkeley, CA 94720 USA ; DOE Joint Genome Institute, 2800 Mitchell Drive, Walnut Creek, CA 94598 USA
| | - Len A Pennacchio
- Genomics Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Mailstop 84-171, Berkeley, CA 94720 USA ; DOE Joint Genome Institute, 2800 Mitchell Drive, Walnut Creek, CA 94598 USA
| | - Mitchell J Weiss
- Department of Hematology, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105 USA
| | - Gerd A Blobel
- Division of Hematology, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104 USA ; Perelman School of Medicine at the University of Pennsylvania, 415 Curie Boulevard, Philadelphia, PA 19104 USA
| | - Ross C Hardison
- Center for Comparative Genomics and Bioinformatics, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, 304 Wartik Laboratory, University Park, PA 16802 USA
| |
Collapse
|
217
|
Singh K, Cassano M, Planet E, Sebastian S, Jang SM, Sohi G, Faralli H, Choi J, Youn HD, Dilworth FJ, Trono D. A KAP1 phosphorylation switch controls MyoD function during skeletal muscle differentiation. Genes Dev 2015; 29:513-25. [PMID: 25737281 PMCID: PMC4358404 DOI: 10.1101/gad.254532.114] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The transcriptional activator MyoD serves as a master controller of myogenesis. Singh et al. identify KAP1/TRIM28 as a key regulator of MyoD function. In myoblasts, KAP1 is present with MyoD and Mef2 at many muscle genes, where it acts as a scaffold to recruit not only coactivators such as p300 and LSD1 but also corepressors such as G9a and HDAC1, with promoter silencing as the net outcome. Upon differentiation, MSK1-mediated phosphorylation of KAP1 releases the corepressors from the scaffold, unleashing transcriptional activation by MyoD/Mef2 and their positive cofactors. The transcriptional activator MyoD serves as a master controller of myogenesis. Often in partnership with Mef2 (myocyte enhancer factor 2), MyoD binds to the promoters of hundreds of muscle genes in proliferating myoblasts yet activates these targets only upon receiving cues that launch differentiation. What regulates this off/on switch of MyoD function has been incompletely understood, although it is known to reflect the action of chromatin modifiers. Here, we identify KAP1 (KRAB [Krüppel-like associated box]-associated protein 1)/TRIM28 (tripartite motif protein 28) as a key regulator of MyoD function. In myoblasts, KAP1 is present with MyoD and Mef2 at many muscle genes, where it acts as a scaffold to recruit not only coactivators such as p300 and LSD1 but also corepressors such as G9a and HDAC1 (histone deacetylase 1), with promoter silencing as the net outcome. Upon differentiation, MSK1-mediated phosphorylation of KAP1 releases the corepressors from the scaffold, unleashing transcriptional activation by MyoD/Mef2 and their positive cofactors. Thus, our results reveal KAP1 as a previously unappreciated interpreter of cell signaling, which modulates the ability of MyoD to drive myogenesis.
Collapse
Affiliation(s)
- Kulwant Singh
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
| | - Marco Cassano
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Evarist Planet
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Soji Sebastian
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
| | - Suk Min Jang
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Gurjeev Sohi
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
| | - Hervé Faralli
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
| | - Jinmi Choi
- Department of Biomedical Sciences and Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Hong-Duk Youn
- Department of Biomedical Sciences and Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - F Jeffrey Dilworth
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ontario K1H 8L6, Canada
| | - Didier Trono
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland;
| |
Collapse
|
218
|
Brancaccio A, Palacios D. Chromatin signaling in muscle stem cells: interpreting the regenerative microenvironment. Front Aging Neurosci 2015; 7:36. [PMID: 25904863 PMCID: PMC4387924 DOI: 10.3389/fnagi.2015.00036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/04/2015] [Indexed: 12/12/2022] Open
Abstract
Muscle regeneration in the adult occurs in response to damage at expenses of a population of adult stem cells, the satellite cells. Upon injury, either physical or genetic, signals released within the satellite cell niche lead to the commitment, expansion and differentiation of the pool of muscle progenitors to repair damaged muscle. To achieve this goal satellite cells undergo a dramatic transcriptional reprogramming to coordinately activate and repress specific subset of genes. Although the epigenetics of muscle regeneration has been extensively discussed, less emphasis has been put on how extra-cellular cues are translated into the specific chromatin reorganization necessary for progression through the myogenic program. In this review we will focus on how satellite cells sense the regenerative microenvironment in physiological and pathological circumstances, paying particular attention to the mechanism through which the external stimuli are transduced to the nucleus to modulate chromatin structure and gene expression. We will discuss the pathways involved and how alterations in this chromatin signaling may contribute to satellite cells dysfunction during aging and disease.
Collapse
Affiliation(s)
- Arianna Brancaccio
- Laboratory of Epigenetics and Signaling, IRCCS Fondazione Santa Lucia Rome, Italy
| | - Daniela Palacios
- Laboratory of Epigenetics and Signaling, IRCCS Fondazione Santa Lucia Rome, Italy
| |
Collapse
|
219
|
Becker K, Beer C, Freitag M, Kück U. Genome-wide identification of target genes of a mating-type α-domain transcription factor reveals functions beyond sexual development. Mol Microbiol 2015; 96:1002-22. [DOI: 10.1111/mmi.12987] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Kordula Becker
- Christian Doppler Laboratory for Fungal Biotechnology; Lehrstuhl für Allgemeine und Molekulare Botanik; Ruhr-Universität Bochum; Universitätsstr. 150 D-44780 Bochum Germany
| | - Christina Beer
- Christian Doppler Laboratory for Fungal Biotechnology; Lehrstuhl für Allgemeine und Molekulare Botanik; Ruhr-Universität Bochum; Universitätsstr. 150 D-44780 Bochum Germany
| | - Michael Freitag
- Department of Biochemistry and Biophysics; Oregon State University; Corvallis Oregon 97331-7305 USA
| | - Ulrich Kück
- Christian Doppler Laboratory for Fungal Biotechnology; Lehrstuhl für Allgemeine und Molekulare Botanik; Ruhr-Universität Bochum; Universitätsstr. 150 D-44780 Bochum Germany
| |
Collapse
|
220
|
Fong AP, Yao Z, Zhong JW, Johnson NM, Farr GH, Maves L, Tapscott SJ. Conversion of MyoD to a neurogenic factor: binding site specificity determines lineage. Cell Rep 2015; 10:1937-46. [PMID: 25801030 DOI: 10.1016/j.celrep.2015.02.055] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 01/16/2015] [Accepted: 02/23/2015] [Indexed: 10/23/2022] Open
Abstract
MyoD and NeuroD2, master regulators of myogenesis and neurogenesis, bind to a "shared" E-box sequence (CAGCTG) and a "private" sequence (CAGGTG or CAGATG, respectively). To determine whether private-site recognition is sufficient to confer lineage specification, we generated a MyoD mutant with the DNA-binding specificity of NeuroD2. This chimeric mutant gained binding to NeuroD2 private sites but maintained binding to a subset of MyoD-specific sites, activating part of both the muscle and neuronal programs. Sequence analysis revealed an enrichment for PBX/MEIS motifs at the subset of MyoD-specific sites bound by the chimera, and point mutations that prevent MyoD interaction with PBX/MEIS converted the chimera to a pure neurogenic factor. Therefore, redirecting MyoD binding from MyoD private sites to NeuroD2 private sites, despite preserved binding to the MyoD/NeuroD2 shared sites, is sufficient to change MyoD from a master regulator of myogenesis to a master regulator of neurogenesis.
Collapse
Affiliation(s)
- Abraham P Fong
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Pediatrics, Division of Hematology-Oncology, University of Washington School of Medicine, Seattle, WA 98105, USA.
| | - Zizhen Yao
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jun Wen Zhong
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Nathan M Johnson
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Gist H Farr
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Lisa Maves
- Department of Pediatrics, Division of Cardiology, University of Washington, Seattle, WA 98105, USA; Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Stephen J Tapscott
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Neurology, University of Washington School of Medicine, Seattle, WA 98105, USA.
| |
Collapse
|
221
|
Wang L, Zhao Y, Bao X, Zhu X, Kwok YKY, Sun K, Chen X, Huang Y, Jauch R, Esteban MA, Sun H, Wang H. LncRNA Dum interacts with Dnmts to regulate Dppa2 expression during myogenic differentiation and muscle regeneration. Cell Res 2015; 25:335-50. [PMID: 25686699 PMCID: PMC4349245 DOI: 10.1038/cr.2015.21] [Citation(s) in RCA: 210] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 11/01/2014] [Accepted: 11/18/2014] [Indexed: 12/17/2022] Open
Abstract
Emerging studies document the roles of long non-coding RNAs (LncRNAs) in regulating gene expression at chromatin level but relatively less is known how they regulate DNA methylation. Here we identify an lncRNA, Dum (developmental pluripotency-associated 2 (Dppa2) Upstream binding Muscle lncRNA) in skeletal myoblast cells. The expression of Dum is dynamically regulated during myogenesis in vitro and in vivo. It is also transcriptionally induced by MyoD binding upon myoblast differentiation. Functional analyses show that it promotes myoblast differentiation and damage-induced muscle regeneration. Mechanistically, Dum was found to silence its neighboring gene, Dppa2, in cis through recruiting Dnmt1, Dnmt3a and Dnmt3b. Furthermore, intrachromosomal looping between Dum locus and Dppa2 promoter is necessary for Dum/Dppa2 interaction. Collectively, we have identified a novel lncRNA that interacts with Dnmts to regulate myogenesis.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Zhao
- Department of Obstetrics and Gynaecology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xichen Bao
- Laboratory of Chromatin and Human Disease, Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xihua Zhu
- Laboratory of Chromatin and Human Disease, Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yvonne Ka-yin Kwok
- Department of Obstetrics and Gynaecology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Kun Sun
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaona Chen
- Department of Obstetrics and Gynaecology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yongheng Huang
- Genome Regulation Laboratory, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ralf Jauch
- Genome Regulation Laboratory, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Miguel A Esteban
- Laboratory of Chromatin and Human Disease, Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Hao Sun
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Huating Wang
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
222
|
Suryamohan K, Halfon MS. Identifying transcriptional cis-regulatory modules in animal genomes. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2015; 4:59-84. [PMID: 25704908 PMCID: PMC4339228 DOI: 10.1002/wdev.168] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 11/04/2014] [Accepted: 11/16/2014] [Indexed: 11/08/2022]
Abstract
UNLABELLED Gene expression is regulated through the activity of transcription factors (TFs) and chromatin-modifying proteins acting on specific DNA sequences, referred to as cis-regulatory elements. These include promoters, located at the transcription initiation sites of genes, and a variety of distal cis-regulatory modules (CRMs), the most common of which are transcriptional enhancers. Because regulated gene expression is fundamental to cell differentiation and acquisition of new cell fates, identifying, characterizing, and understanding the mechanisms of action of CRMs is critical for understanding development. CRM discovery has historically been challenging, as CRMs can be located far from the genes they regulate, have few readily identifiable sequence characteristics, and for many years were not amenable to high-throughput discovery methods. However, the recent availability of complete genome sequences and the development of next-generation sequencing methods have led to an explosion of both computational and empirical methods for CRM discovery in model and nonmodel organisms alike. Experimentally, CRMs can be identified through chromatin immunoprecipitation directed against TFs or histone post-translational modifications, identification of nucleosome-depleted 'open' chromatin regions, or sequencing-based high-throughput functional screening. Computational methods include comparative genomics, clustering of known or predicted TF-binding sites, and supervised machine-learning approaches trained on known CRMs. All of these methods have proven effective for CRM discovery, but each has its own considerations and limitations, and each is subject to a greater or lesser number of false-positive identifications. Experimental confirmation of predictions is essential, although shortcomings in current methods suggest that additional means of validation need to be developed. For further resources related to this article, please visit the WIREs website. CONFLICT OF INTEREST The authors have declared no conflicts of interest for this article.
Collapse
Affiliation(s)
- Kushal Suryamohan
- Department of Biochemistry, University at Buffalo-State University of New York, Buffalo, NY 14203, USA
- NY State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA
| | - Marc S. Halfon
- Department of Biochemistry, University at Buffalo-State University of New York, Buffalo, NY 14203, USA
- Department of Biological Sciences, University at Buffalo-State University of New York, Buffalo, NY 14203, USA
- Department of Biomedical Informatics, University at Buffalo-State University of New York, Buffalo, NY 14203, USA
- NY State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA
- Molecular and Cellular Biology Department and Program in Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
223
|
Harada A, Mallappa C, Okada S, Butler JT, Baker SP, Lawrence JB, Ohkawa Y, Imbalzano AN. Spatial re-organization of myogenic regulatory sequences temporally controls gene expression. Nucleic Acids Res 2015; 43:2008-21. [PMID: 25653159 PMCID: PMC4344497 DOI: 10.1093/nar/gkv046] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 11/05/2014] [Accepted: 01/13/2015] [Indexed: 12/14/2022] Open
Abstract
During skeletal muscle differentiation, the activation of some tissue-specific genes occurs immediately while others are delayed. The molecular basis controlling temporal gene regulation is poorly understood. We show that the regulatory sequences, but not other regions of genes expressed at late times of myogenesis, are in close physical proximity in differentiating embryonic tissue and in differentiating culture cells, despite these genes being located on different chromosomes. Formation of these inter-chromosomal interactions requires the lineage-determinant MyoD and functional Brg1, the ATPase subunit of SWI/SNF chromatin remodeling enzymes. Ectopic expression of myogenin and a specific Mef2 isoform induced myogenic differentiation without activating endogenous MyoD expression. Under these conditions, the regulatory sequences of late gene loci were not in close proximity, and these genes were prematurely activated. The data indicate that the spatial organization of late genes contributes to temporal regulation of myogenic transcription by restricting late gene expression during the early stages of myogenesis.
Collapse
Affiliation(s)
- Akihito Harada
- Department of Advanced Medical Initiatives, JST-CREST, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan
| | - Chandrashekara Mallappa
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Seiji Okada
- Department of Advanced Medical Initiatives, JST-CREST, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan
| | - John T Butler
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Stephen P Baker
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jeanne B Lawrence
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Yasuyuki Ohkawa
- Department of Advanced Medical Initiatives, JST-CREST, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Anthony N Imbalzano
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
224
|
Weber D, Heisig J, Kneitz S, Wolf E, Eilers M, Gessler M. Mechanisms of epigenetic and cell-type specific regulation of Hey target genes in ES cells and cardiomyocytes. J Mol Cell Cardiol 2015; 79:79-88. [DOI: 10.1016/j.yjmcc.2014.11.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/07/2014] [Accepted: 11/06/2014] [Indexed: 01/20/2023]
|
225
|
Sohi G, Dilworth FJ. Noncoding RNAs as epigenetic mediators of skeletal muscle regeneration. FEBS J 2015; 282:1630-46. [PMID: 25483175 DOI: 10.1111/febs.13170] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/01/2014] [Accepted: 12/02/2014] [Indexed: 12/16/2022]
Abstract
Skeletal muscle regeneration is a well-characterized biological process in which resident adult stem cells must undertake a series of cell-fate decisions to ensure efficient repair of the damaged muscle fibers while also maintaining the stem cell niche. Satellite cells, the main stem cell contributing to the repaired muscle fiber, are maintained in a quiescent state in healthy muscle. Upon injury, the satellite cells become activated, and proliferate to expand the muscle progenitor cell population before returning to the quiescent state or differentiating to become myofibers. Importantly, the determination of cell fate is controlled at the epigenetic level in response to environmental cues. In this review, we discuss our current understanding of the role played by noncoding RNAs (both miRNAs and long-noncoding RNAs) in the epigenetic control of muscle regeneration.
Collapse
Affiliation(s)
- Gurjeev Sohi
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Canada
| | | |
Collapse
|
226
|
Puri D, Gala H, Mishra R, Dhawan J. High-wire act: the poised genome and cellular memory. FEBS J 2014; 282:1675-91. [PMID: 25440020 DOI: 10.1111/febs.13165] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 11/22/2014] [Accepted: 11/27/2014] [Indexed: 12/12/2022]
Abstract
Emerging evidence aided by genome-wide analysis of chromatin and transcriptional states has shed light on the mechanisms by which stem cells achieve cellular memory. The epigenetic and transcriptional plasticity governing stem cell behavior is highlighted by the identification of 'poised' genes, which permit cells to maintain readiness to undertake alternate developmental fates. This review focuses on two crucial mechanisms of gene poising: bivalent chromatin marks and RNA polymerase II stalling. We provide the context for these mechanisms by exploring the current consensus on the regulation of chromatin states, especially in quiescent adult stem cells, where poised genes are critical for recapitulating developmental choices, leading to regenerative function.
Collapse
Affiliation(s)
- Deepika Puri
- Council of Scientific and Industrial Research-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | | | | |
Collapse
|
227
|
Cattenoz PB, Giangrande A. New insights in the clockwork mechanism regulating lineage specification: Lessons from the Drosophila nervous system. Dev Dyn 2014; 244:332-41. [PMID: 25399853 DOI: 10.1002/dvdy.24228] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 11/06/2014] [Accepted: 11/07/2014] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Powerful transcription factors called fate determinants induce robust differentiation programs in multipotent cells and trigger lineage specification. These factors guarantee the differentiation of specific tissues/organs/cells at the right place and the right moment to form a fully functional organism. Fate determinants are activated by temporal, positional, epigenetic, and post-transcriptional cues, hence integrating complex and dynamic developmental networks. In turn, they activate specific transcriptional/epigenetic programs that secure novel molecular landscapes. RESULTS In this review, we use the Drosophila Gcm glial determinant as a model to discuss the mechanisms that allow lineage specification in the nervous system. The dynamic regulation of Gcm via interlocked loops has recently emerged as a key event in the establishment of stable identity. Gcm induces gliogenesis while triggering its own extinction, thus preventing the appearance of metastable states and neoplastic processes. CONCLUSIONS Using simple animal models that allow in vivo manipulations provides a key tool to disentangle the complex regulation of cell fate determinants.
Collapse
Affiliation(s)
- Pierre B Cattenoz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, Illkirch, France; Université de Strasbourg, Illkirch, France
| | | |
Collapse
|
228
|
Estrella NL, Desjardins CA, Nocco SE, Clark AL, Maksimenko Y, Naya FJ. MEF2 transcription factors regulate distinct gene programs in mammalian skeletal muscle differentiation. J Biol Chem 2014; 290:1256-68. [PMID: 25416778 DOI: 10.1074/jbc.m114.589838] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Skeletal muscle differentiation requires precisely coordinated transcriptional regulation of diverse gene programs that ultimately give rise to the specialized properties of this cell type. In Drosophila, this process is controlled, in part, by MEF2, the sole member of an evolutionarily conserved transcription factor family. By contrast, vertebrate MEF2 is encoded by four distinct genes, Mef2a, -b, -c, and -d, making it far more challenging to link this transcription factor to the regulation of specific muscle gene programs. Here, we have taken the first step in molecularly dissecting vertebrate MEF2 transcriptional function in skeletal muscle differentiation by depleting individual MEF2 proteins in myoblasts. Whereas MEF2A is absolutely required for proper myoblast differentiation, MEF2B, -C, and -D were found to be dispensable for this process. Furthermore, despite the extensive redundancy, we show that mammalian MEF2 proteins regulate a significant subset of nonoverlapping gene programs. These results suggest that individual MEF2 family members are able to recognize specific targets among the entire cohort of MEF2-regulated genes in the muscle genome. These findings provide opportunities to modulate the activity of MEF2 isoforms and their respective gene programs in skeletal muscle homeostasis and disease.
Collapse
Affiliation(s)
- Nelsa L Estrella
- From the Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215
| | - Cody A Desjardins
- From the Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215
| | - Sarah E Nocco
- From the Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215
| | - Amanda L Clark
- From the Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215
| | - Yevgeniy Maksimenko
- From the Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215
| | - Francisco J Naya
- From the Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215
| |
Collapse
|
229
|
Beck S, Lee BK, Rhee C, Song J, Woo AJ, Kim J. CpG island-mediated global gene regulatory modes in mouse embryonic stem cells. Nat Commun 2014; 5:5490. [PMID: 25405324 PMCID: PMC4236720 DOI: 10.1038/ncomms6490] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 10/03/2014] [Indexed: 12/13/2022] Open
Abstract
Both transcriptional and epigenetic regulations are fundamental for the control of eukaryotic gene expression. Here we perform a compendium analysis of >200 large sequencing data sets to elucidate the regulatory logic of global gene expression programs in mouse embryonic stem (ES) cells. We define four major classes of DNA-binding proteins (Core, PRC, MYC and CTCF) based on their target co-occupancy, and discover reciprocal regulation between the MYC and PRC classes for the activity of nearly all genes under the control of the CpG island (CGI)-containing promoters. This CGI-dependent regulatory mode explains the functional segregation between CGI-containing and CGI-less genes during early development. By defining active enhancers based on the co-occupancy of the Core class, we further demonstrate their additive roles in CGI-containing gene expression and cell type-specific roles in CGI-less gene expression. Altogether, our analyses provide novel insights into previously unknown CGI-dependent global gene regulatory modes.
Collapse
Affiliation(s)
- Samuel Beck
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, USA
| | - Bum-Kyu Lee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, USA
| | - Catherine Rhee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, USA
| | - Jawon Song
- Texas Advanced Computing Center, The University of Texas at Austin, Austin, Texas 78758, USA
| | - Andrew J Woo
- School of Medicine and Pharmacology, Royal Perth Hospital Unit, The University of Western Australia, Perth, WA 6000, Australia
| | - Jonghwan Kim
- 1] Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, USA [2] Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, USA [3] Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, Texas 78712, USA
| |
Collapse
|
230
|
Cho OH, Mallappa C, Hernández-Hernández JM, Rivera-Pérez JA, Imbalzano AN. Contrasting roles for MyoD in organizing myogenic promoter structures during embryonic skeletal muscle development. Dev Dyn 2014; 244:43-55. [PMID: 25329411 DOI: 10.1002/dvdy.24217] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/09/2014] [Accepted: 09/23/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Among the complexities of skeletal muscle differentiation is a temporal distinction in the onset of expression of different lineage-specific genes. The lineage-determining factor MyoD is bound to myogenic genes at the onset of differentiation whether gene activation is immediate or delayed. How temporal regulation of differentiation-specific genes is established remains unclear. RESULTS Using embryonic tissue, we addressed the molecular differences in the organization of the myogenin and muscle creatine kinase (MCK) gene promoters by examining regulatory factor binding as a function of both time and spatial organization during somitogenesis. At the myogenin promoter, binding of the homeodomain factor Pbx1 coincided with H3 hyperacetylation and was followed by binding of co-activators that modulate chromatin structure. MyoD and myogenin binding occurred subsequently, demonstrating that Pbx1 facilitates chromatin remodeling and modification before myogenic regulatory factor binding. At the same time, the MCK promoter was bound by HDAC2 and MyoD, and activating histone marks were largely absent. The association of HDAC2 and MyoD was confirmed by co-immunoprecipitation, proximity ligation assay (PLA), and sequential ChIP. CONCLUSIONS MyoD differentially promotes activated and repressed chromatin structures at myogenic genes early after the onset of skeletal muscle differentiation in the developing mouse embryo.
Collapse
Affiliation(s)
- Ok Hyun Cho
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | | | | | | | | |
Collapse
|
231
|
Segalés J, Perdiguero E, Muñoz-Cánoves P. Epigenetic control of adult skeletal muscle stem cell functions. FEBS J 2014; 282:1571-88. [PMID: 25251895 DOI: 10.1111/febs.13065] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/18/2014] [Accepted: 09/19/2014] [Indexed: 12/12/2022]
Abstract
Skeletal muscle regeneration in the adult (de novo myogenesis) depends on a resident population of muscle stem cells (satellite cells) that are normally quiescent. In response to injury or stress, satellite cells are activated and expand as myoblast cells that differentiate and fuse to form new muscle fibers or return to quiescence to maintain the stem cell pool (self-renewal). Satellite cell-dependent myogenesis is a well-characterized multi-step process orchestrated by muscle-specific transcription factors, such as Pax3/Pax7 and members of the MyoD family of muscle regulatory factors, and epigenetically controlled by mechanisms such as DNA methylation, covalent modification of histones and non-coding RNAs. Recent results from next-generation genome-wide sequencing have increased our understanding about the highly intricate layers of epigenetic regulation involved in satellite cell maintenance, activation, differentiation and self-renewal, and their cross-talk with the muscle-specific transcriptional machinery.
Collapse
Affiliation(s)
- Jessica Segalés
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University, Center for Networked Biomedical Research on Neurodegenerative Diseases, Barcelona, Spain
| | | | | |
Collapse
|
232
|
Forcales SV. The BAF60c-MyoD complex poises chromatin for rapid transcription. BIOARCHITECTURE 2014; 2:104-109. [PMID: 22880151 PMCID: PMC3414383 DOI: 10.4161/bioa.20970] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Chromatin remodeling by the SWI/SNF complex is required to activate the transcription of myogenic-specific genes. Our work addressed the details of how SWI/SNF is recruited to myogenic regulatory regions in response to differentiation signals. Surprisingly, the muscle determination factor MyoD and the SWI/SNF subunit BAF60c form a complex on the regulatory elements of MyoD-targeted genes in myogenic precursor cells. This Brg1-devoid MyoD-BAF60c complex flags the chromatin of myogenic-differentiation genes before transcription is activated. On differentiation, BAF60c phosphorylation on a conserved threonine by p38 α kinase promotes the incorporation of MyoD-BAF60c into a Brg1-based SWI/SNF complex, which remodels the chromatin and activates transcription of MyoD-target genes. Downregulation of BAF60c expression prevents MyoD access to the chromatin and the proper loading of an active myogenic transcriptosome preventing the expression of hundreds of myogenic genes. Our data support an unprecedented two-step model by which (1) pre-assembled BAF60c-MyoD complex poises the chromatin of myogenic genes for rapid transcription; (2) chromatin-bound BAF60c "senses" the myogenic differentiation cues and recruits an active SWI/SNF complex to remodel the chromatin allowing transcriptional activation.
Collapse
Affiliation(s)
- Sonia-Vanina Forcales
- Institute of Predictive and Personalized Medicine of Cancer; Badalona, Barcelona, Spain
| |
Collapse
|
233
|
Wales S, Hashemi S, Blais A, McDermott JC. Global MEF2 target gene analysis in cardiac and skeletal muscle reveals novel regulation of DUSP6 by p38MAPK-MEF2 signaling. Nucleic Acids Res 2014; 42:11349-62. [PMID: 25217591 PMCID: PMC4191398 DOI: 10.1093/nar/gku813] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
MEF2 plays a profound role in the regulation of transcription in cardiac and skeletal muscle lineages. To define the overlapping and unique MEF2A genomic targets, we utilized ChIP-exo analysis of cardiomyocytes and skeletal myoblasts. Of the 2783 and 1648 MEF2A binding peaks in skeletal myoblasts and cardiomyocytes, respectively, 294 common binding sites were identified. Genomic targets were compared to differentially expressed genes in RNA-seq analysis of MEF2A depleted myogenic cells, revealing two prominent genetic networks. Genes largely associated with muscle development were down-regulated by loss of MEF2A while up-regulated genes reveal a previously unrecognized function of MEF2A in suppressing growth/proliferative genes. Several up-regulated (Tprg, Mctp2, Kitl, Prrx1, Dusp6) and down-regulated (Atp1a2, Hspb7, Tmem182, Sorbs2, Lmod3) MEF2A target genes were chosen for further investigation. Interestingly, siRNA targeting of the MEF2A/D heterodimer revealed a somewhat divergent role in the regulation of Dusp6, a MAPK phosphatase, in cardiac and skeletal myogenic lineages. Furthermore, MEF2D functions as a p38MAPK-dependent repressor of Dusp6 in myoblasts. These data illustrate that MEF2 orchestrates both common and non-overlapping programs of signal-dependent gene expression in skeletal and cardiac muscle lineages.
Collapse
Affiliation(s)
- Stephanie Wales
- Department of Biology, York University, 4700 Keele Street Toronto, Ontario, M3J 1P3 Canada Muscle Health Research Centre (MHRC), York University, 4700 Keele Street, Toronto, Ontario, M3J 1P3 Canada Centre for Research on Biomolecular Interactions (CRBI), 4700 Keele Street, Toronto, Ontario, M3J 1P3 Canada
| | - Sara Hashemi
- Department of Biology, York University, 4700 Keele Street Toronto, Ontario, M3J 1P3 Canada Muscle Health Research Centre (MHRC), York University, 4700 Keele Street, Toronto, Ontario, M3J 1P3 Canada Centre for Research on Biomolecular Interactions (CRBI), 4700 Keele Street, Toronto, Ontario, M3J 1P3 Canada
| | - Alexandre Blais
- Ottawa Institute of Systems Biology, University of Ottawa, Health Sciences Campus, 451 Smyth Road, Ottawa, Ontario, K1H 8M5 Canada
| | - John C McDermott
- Department of Biology, York University, 4700 Keele Street Toronto, Ontario, M3J 1P3 Canada Muscle Health Research Centre (MHRC), York University, 4700 Keele Street, Toronto, Ontario, M3J 1P3 Canada Centre for Research on Biomolecular Interactions (CRBI), 4700 Keele Street, Toronto, Ontario, M3J 1P3 Canada Centre for Research in Mass Spectrometry (CRMS), York University, 4700 Keele Street, Toronto, Ontario, M3J 1P3 Canada
| |
Collapse
|
234
|
Zalc A, Hayashi S, Auradé F, Bröhl D, Chang T, Mademtzoglou D, Mourikis P, Yao Z, Cao Y, Birchmeier C, Relaix F. Antagonistic regulation of p57kip2 by Hes/Hey downstream of Notch signaling and muscle regulatory factors regulates skeletal muscle growth arrest. Development 2014; 141:2780-90. [PMID: 25005473 DOI: 10.1242/dev.110155] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A central question in development is to define how the equilibrium between cell proliferation and differentiation is temporally and spatially regulated during tissue formation. Here, we address how interactions between cyclin-dependent kinase inhibitors essential for myogenic growth arrest (p21(cip1) and p57(kip2)), the Notch pathway and myogenic regulatory factors (MRFs) orchestrate the proliferation, specification and differentiation of muscle progenitor cells. We first show that cell cycle exit and myogenic differentiation can be uncoupled. In addition, we establish that skeletal muscle progenitor cells require Notch signaling to maintain their cycling status. Using several mouse models combined with ex vivo studies, we demonstrate that Notch signaling is required to repress p21(cip1) and p57(kip2) expression in muscle progenitor cells. Finally, we identify a muscle-specific regulatory element of p57(kip2) directly activated by MRFs in myoblasts but repressed by the Notch targets Hes1/Hey1 in progenitor cells. We propose a molecular mechanism whereby information provided by Hes/Hey downstream of Notch as well as MRF activities are integrated at the level of the p57(kip2) enhancer to regulate the decision between progenitor cell maintenance and muscle differentiation.
Collapse
Affiliation(s)
- Antoine Zalc
- UPMC Paris 06, U 974, Paris, F-75013, France INSERM, Avenir Team, Pitié-Salpétrière, Paris, F-75013, France Institut de Myologie, Paris, F-75013, France
| | - Shinichiro Hayashi
- UPMC Paris 06, U 974, Paris, F-75013, France INSERM, Avenir Team, Pitié-Salpétrière, Paris, F-75013, France Institut de Myologie, Paris, F-75013, France
| | - Frédéric Auradé
- UPMC Paris 06, U 974, Paris, F-75013, France INSERM, Avenir Team, Pitié-Salpétrière, Paris, F-75013, France Institut de Myologie, Paris, F-75013, France
| | - Dominique Bröhl
- Max-Delbrück-Center for Molecular Medicine, Berlin 13125, Germany
| | - Ted Chang
- UPMC Paris 06, U 974, Paris, F-75013, France INSERM, Avenir Team, Pitié-Salpétrière, Paris, F-75013, France Institut de Myologie, Paris, F-75013, France
| | - Despoina Mademtzoglou
- UPMC Paris 06, U 974, Paris, F-75013, France INSERM, Avenir Team, Pitié-Salpétrière, Paris, F-75013, France Institut de Myologie, Paris, F-75013, France
| | - Philippos Mourikis
- UPMC Paris 06, U 974, Paris, F-75013, France INSERM, Avenir Team, Pitié-Salpétrière, Paris, F-75013, France Institut de Myologie, Paris, F-75013, France
| | - Zizhen Yao
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Yi Cao
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | - Frédéric Relaix
- UPMC Paris 06, U 974, Paris, F-75013, France INSERM, Avenir Team, Pitié-Salpétrière, Paris, F-75013, France Institut de Myologie, Paris, F-75013, France
| |
Collapse
|
235
|
Lau OS, Davies KA, Chang J, Adrian J, Rowe MH, Ballenger CE, Bergmann DC. Direct roles of SPEECHLESS in the specification of stomatal self-renewing cells. Science 2014; 345:1605-9. [PMID: 25190717 DOI: 10.1126/science.1256888] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Lineage-specific stem cells are critical for the production and maintenance of specific cell types and tissues in multicellular organisms. In Arabidopsis, the initiation and proliferation of stomatal lineage cells is controlled by the basic helix-loop-helix transcription factor SPEECHLESS (SPCH). SPCH-driven asymmetric and self-renewing divisions allow flexibility in stomatal production and overall organ growth. How SPCH directs stomatal lineage cell behaviors, however, is unclear. Here, we improved the chromatin immunoprecipitation (ChIP) assay and profiled the genome-wide targets of Arabidopsis SPCH in vivo. We found that SPCH controls key regulators of cell fate and asymmetric cell divisions and modulates responsiveness to peptide and phytohormone-mediated intercellular communication. Our results delineate the molecular pathways that regulate an essential adult stem cell lineage in plants.
Collapse
Affiliation(s)
- On Sun Lau
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Kelli A Davies
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Jessica Chang
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Jessika Adrian
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Matthew H Rowe
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | | | - Dominique C Bergmann
- Department of Biology, Stanford University, Stanford, CA 94305, USA. Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA. Carnegie Institution for Science, Stanford, CA 94305, USA.
| |
Collapse
|
236
|
Wackerhage H, Del Re DP, Judson RN, Sudol M, Sadoshima J. The Hippo signal transduction network in skeletal and cardiac muscle. Sci Signal 2014; 7:re4. [PMID: 25097035 DOI: 10.1126/scisignal.2005096] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The discovery of the Hippo pathway can be traced back to two areas of research. Genetic screens in fruit flies led to the identification of the Hippo pathway kinases and scaffolding proteins that function together to suppress cell proliferation and tumor growth. Independent research, often in the context of muscle biology, described Tead (TEA domain) transcription factors, which bind CATTCC DNA motifs to regulate gene expression. These two research areas were joined by the finding that the Hippo pathway regulates the activity of Tead transcription factors mainly through phosphorylation of the transcriptional coactivators Yap and Taz, which bind to and activate Teads. Additionally, many other signal transduction proteins crosstalk to members of the Hippo pathway forming a Hippo signal transduction network. We discuss evidence that the Hippo signal transduction network plays important roles in myogenesis, regeneration, muscular dystrophy, and rhabdomyosarcoma in skeletal muscle, as well as in myogenesis, organ size control, and regeneration of the heart. Understanding the role of Hippo kinases in skeletal and heart muscle physiology could have important implications for translational research.
Collapse
Affiliation(s)
- Henning Wackerhage
- School of Medical Sciences, University of Aberdeen, Health Sciences Building, Foresterhill, AB25 2ZD Aberdeen, Scotland, UK.
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Robert N Judson
- School of Medical Sciences, University of Aberdeen, Health Sciences Building, Foresterhill, AB25 2ZD Aberdeen, Scotland, UK. Biomedical Research Centre, University of British Columbia, 317-2194 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Marius Sudol
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Republic of Singapore. Department of Medicine, Mount Sinai School of Medicine, One Gustave Levy Place, New York, NY 10029, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 07103, USA
| |
Collapse
|
237
|
Battistelli C, Busanello A, Maione R. Functional interplay between MyoD and CTCF in regulating long-range chromatin interactions during differentiation. J Cell Sci 2014; 127:3757-67. [PMID: 25002401 DOI: 10.1242/jcs.149427] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Higher-order chromatin structures appear to be dynamically arranged during development and differentiation. However, the molecular mechanism underlying their maintenance or disruption and their functional relevance to gene regulation are poorly understood. We recently described a dynamic long-range chromatin interaction between the gene promoter of the cdk inhibitor p57(kip2) (also known as Cdkn1c) and the imprinting control region KvDMR1 in muscle cells. Here, we show that CTCF, the best characterized organizer of long-range chromatin interactions, binds to both the p57(kip2) promoter and KvDMR1 and is necessary for the maintenance of their physical contact. Moreover, we show that CTCF-mediated looping is required to prevent p57(kip2) expression before differentiation. Finally, we provide evidence that the induction of p57(kip2) during myogenesis involves the physical interaction of the muscle-regulatory factor MyoD with CTCF at KvDMR1, the displacement of the cohesin complex subunit Rad21 and the destabilization of the chromatin loop. The finding that MyoD affects chromatin looping at CTCF-binding sites represents the first evidence that a differentiation factor regulates chromatin-loop dynamics and provides a useful paradigm for gaining insights into the developmental regulation of long-range chromatin contacts.
Collapse
Affiliation(s)
- Cecilia Battistelli
- Pasteur Institute-Fondazione Cenci Bolognetti, Department of Cellular Biotechnologies and Haematology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Anna Busanello
- Pasteur Institute-Fondazione Cenci Bolognetti, Department of Cellular Biotechnologies and Haematology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Rossella Maione
- Pasteur Institute-Fondazione Cenci Bolognetti, Department of Cellular Biotechnologies and Haematology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| |
Collapse
|
238
|
Chandra S, Baribault C, Lacey M, Ehrlich M. Myogenic differential methylation: diverse associations with chromatin structure. BIOLOGY 2014; 3:426-51. [PMID: 24949935 PMCID: PMC4085616 DOI: 10.3390/biology3020426] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 05/21/2014] [Accepted: 05/21/2014] [Indexed: 11/16/2022]
Abstract
Employing a new algorithm for identifying differentially methylated regions (DMRs) from reduced representation bisulfite sequencing profiles, we identified 1972 hypermethylated and 3250 hypomethylated myogenic DMRs in a comparison of myoblasts (Mb) and myotubes (Mt) with 16 types of nonmuscle cell cultures. DMRs co-localized with a variety of chromatin structures, as deduced from ENCODE whole-genome profiles. Myogenic hypomethylation was highly associated with both weak and strong enhancer-type chromatin, while hypermethylation was infrequently associated with enhancer-type chromatin. Both myogenic hypermethylation and hypomethylation often overlapped weak transcription-type chromatin and Polycomb-repressed-type chromatin. For representative genes, we illustrate relationships between DNA methylation, the local chromatin state, DNaseI hypersensitivity, and gene expression. For example, MARVELD2 exhibited myogenic hypermethylation in transcription-type chromatin that overlapped a silenced promoter in Mb and Mt while TEAD4 had myogenic hypomethylation in intronic subregions displaying enhancer-type or transcription-type chromatin in these cells. For LSP1, alternative promoter usage and active promoter-type chromatin were linked to highly specific myogenic or lymphogenic hypomethylated DMRs. Lastly, despite its myogenesis-associated expression, TBX15 had multiple hypermethylated myogenic DMRs framing its promoter region. This could help explain why TBX15 was previously reported to be underexpressed and, unexpectedly, its promoter undermethylated in placentas exhibiting vascular intrauterine growth restriction.
Collapse
Affiliation(s)
- Sruti Chandra
- Center for Bioinformatics and Genomics, New Orleans, LA 70112, USA.
| | - Carl Baribault
- Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA 70112, USA.
| | - Michelle Lacey
- Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA 70112, USA.
| | - Melanie Ehrlich
- Center for Bioinformatics and Genomics, New Orleans, LA 70112, USA.
| |
Collapse
|
239
|
Santolini M, Mora T, Hakim V. A general pairwise interaction model provides an accurate description of in vivo transcription factor binding sites. PLoS One 2014; 9:e99015. [PMID: 24926895 PMCID: PMC4057186 DOI: 10.1371/journal.pone.0099015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 05/09/2014] [Indexed: 11/19/2022] Open
Abstract
The identification of transcription factor binding sites (TFBSs) on genomic DNA is of crucial importance for understanding and predicting regulatory elements in gene networks. TFBS motifs are commonly described by Position Weight Matrices (PWMs), in which each DNA base pair contributes independently to the transcription factor (TF) binding. However, this description ignores correlations between nucleotides at different positions, and is generally inaccurate: analysing fly and mouse in vivo ChIPseq data, we show that in most cases the PWM model fails to reproduce the observed statistics of TFBSs. To overcome this issue, we introduce the pairwise interaction model (PIM), a generalization of the PWM model. The model is based on the principle of maximum entropy and explicitly describes pairwise correlations between nucleotides at different positions, while being otherwise as unconstrained as possible. It is mathematically equivalent to considering a TF-DNA binding energy that depends additively on each nucleotide identity at all positions in the TFBS, like the PWM model, but also additively on pairs of nucleotides. We find that the PIM significantly improves over the PWM model, and even provides an optimal description of TFBS statistics within statistical noise. The PIM generalizes previous approaches to interdependent positions: it accounts for co-variation of two or more base pairs, and predicts secondary motifs, while outperforming multiple-motif models consisting of mixtures of PWMs. We analyse the structure of pairwise interactions between nucleotides, and find that they are sparse and dominantly located between consecutive base pairs in the flanking region of TFBS. Nonetheless, interactions between pairs of non-consecutive nucleotides are found to play a significant role in the obtained accurate description of TFBS statistics. The PIM is computationally tractable, and provides a general framework that should be useful for describing and predicting TFBSs beyond PWMs.
Collapse
Affiliation(s)
- Marc Santolini
- Laboratoire de Physique Statistique, CNRS, Université P. et M. Curie, Université D. Diderot, École Normale Supérieure, Paris, France
| | - Thierry Mora
- Laboratoire de Physique Statistique, CNRS, Université P. et M. Curie, Université D. Diderot, École Normale Supérieure, Paris, France
| | - Vincent Hakim
- Laboratoire de Physique Statistique, CNRS, Université P. et M. Curie, Université D. Diderot, École Normale Supérieure, Paris, France
| |
Collapse
|
240
|
Borromeo MD, Meredith DM, Castro DS, Chang JC, Tung KC, Guillemot F, Johnson JE. A transcription factor network specifying inhibitory versus excitatory neurons in the dorsal spinal cord. Development 2014; 141:2803-12. [PMID: 24924197 DOI: 10.1242/dev.105866] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The proper balance of excitatory and inhibitory neurons is crucial for normal processing of somatosensory information in the dorsal spinal cord. Two neural basic helix-loop-helix transcription factors (TFs), Ascl1 and Ptf1a, have contrasting functions in specifying these neurons. To understand how Ascl1 and Ptf1a function in this process, we identified their direct transcriptional targets genome-wide in the embryonic mouse neural tube using ChIP-Seq and RNA-Seq. We show that Ascl1 and Ptf1a directly regulate distinct homeodomain TFs that specify excitatory or inhibitory neuronal fates. In addition, Ascl1 directly regulates genes with roles in several steps of the neurogenic program, including Notch signaling, neuronal differentiation, axon guidance and synapse formation. By contrast, Ptf1a directly regulates genes encoding components of the neurotransmitter machinery in inhibitory neurons, and other later aspects of neural development distinct from those regulated by Ascl1. Moreover, Ptf1a represses the excitatory neuronal fate by directly repressing several targets of Ascl1. Ascl1 and Ptf1a bind sequences primarily enriched for a specific E-Box motif (CAGCTG) and for secondary motifs used by Sox, Rfx, Pou and homeodomain factors. Ptf1a also binds sequences uniquely enriched in the CAGATG E-box and in the binding motif for its co-factor Rbpj, providing two factors that influence the specificity of Ptf1a binding. The direct transcriptional targets identified for Ascl1 and Ptf1a provide a molecular understanding of how these DNA-binding proteins function in neuronal development, particularly as key regulators of homeodomain TFs required for neuronal subtype specification.
Collapse
Affiliation(s)
- Mark D Borromeo
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - David M Meredith
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Diogo S Castro
- Instituto Gulbenkian de Ciência, Molecular Neurobiology Laboratory, Oeiras, Portugal
| | - Joshua C Chang
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kuang-Chi Tung
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Francois Guillemot
- Division of Molecular Neurobiology, National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Jane E Johnson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
241
|
Sharov AA, Nishiyama A, Qian Y, Dudekula DB, Longo DL, Schlessinger D, Ko MSH. Chromatin properties of regulatory DNA probed by manipulation of transcription factors. J Comput Biol 2014; 21:569-77. [PMID: 24918633 DOI: 10.1089/cmb.2013.0126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Transcription factors (TFs) bind to DNA and regulate the transcription of nearby genes. However, only a small fraction of TF binding sites have such regulatory effects. Here we search for the predictors of functional binding sites by carrying out a systematic computational screening of a variety of contextual factors (histone modifications, nuclear lamin-bindings, and cofactor bindings). We used regression analysis to test if contextual factors are associated with upregulation or downregulation of neighboring genes following the induction or knockdown of the 9 TFs in mouse embryonic stem (ES) cells. Functional TF binding sites appeared to be either active (i.e., bound by P300, CHD7, mediator, cohesin, and SWI/SNF) or repressed (i.e., with H3K27me3 histone marks and bound by Polycomb factors). Active binding sites mediated the downregulation of nearby genes upon knocking down the activating TFs or inducing repressors. Repressed TF binding sites mediated the upregulation of nearby genes (e.g., poised developmental regulators) upon inducing TFs. In addition, repressed binding sites mediated repressive effects of TFs, identified by the downregulation of target genes after the induction of TFs or by the upregulation of target genes after the knockdown of TFs. The contextual factors associated with functions of DNA-bound TFs were used to improve the identification of candidate target genes regulated by TFs.
Collapse
Affiliation(s)
- Alexei A Sharov
- 1 National Institute on Aging, National Institutes of Health , Baltimore, Maryland
| | | | | | | | | | | | | |
Collapse
|
242
|
Terragni J, Zhang G, Sun Z, Pradhan S, Song L, Crawford GE, Lacey M, Ehrlich M. Notch signaling genes: myogenic DNA hypomethylation and 5-hydroxymethylcytosine. Epigenetics 2014; 9:842-50. [PMID: 24670287 PMCID: PMC4065182 DOI: 10.4161/epi.28597] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 03/11/2014] [Accepted: 03/19/2014] [Indexed: 12/31/2022] Open
Abstract
Notch intercellular signaling is critical for diverse developmental pathways and for homeostasis in various types of stem cells and progenitor cells. Because Notch gene products need to be precisely regulated spatially and temporally, epigenetics is likely to help control expression of Notch signaling genes. Reduced representation bisulfite sequencing (RRBS) indicated significant hypomethylation in myoblasts, myotubes, and skeletal muscle vs. many nonmuscle samples at intragenic or intergenic regions of the following Notch receptor or ligand genes: NOTCH1, NOTCH2, JAG2, and DLL1. An enzymatic assay of sites in or near these genes revealed unusually high enrichment of 5-hydroxymethylcytosine (up to 81%) in skeletal muscle, heart, and cerebellum. Epigenetics studies and gene expression profiles suggest that hypomethylation and/or hydroxymethylation help control expression of these genes in heart, brain, myoblasts, myotubes, and within skeletal muscle myofibers. Such regulation could promote cell renewal, cell maintenance, homeostasis, and a poised state for repair of tissue damage.
Collapse
Affiliation(s)
| | | | - Zhiyi Sun
- New England Biolabs; Ipswich, MA USA
| | | | - Lingyun Song
- Institute for Genome Sciences & Policy; Duke University; Durham, NC USA
| | | | - Michelle Lacey
- Tulane Cancer Center and Department of Mathematics; Tulane Health Sciences Center and Tulane University; New Orleans, LA USA
| | - Melanie Ehrlich
- Program in Human Genetics; Tulane Cancer Center; Center for Bioinformatics and Genomics; Tulane Health Sciences Center; New Orleans, LA USA
| |
Collapse
|
243
|
Cheng J, Blum R, Bowman C, Hu D, Shilatifard A, Shen S, Dynlacht BD. A role for H3K4 monomethylation in gene repression and partitioning of chromatin readers. Mol Cell 2014; 53:979-92. [PMID: 24656132 DOI: 10.1016/j.molcel.2014.02.032] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 12/23/2013] [Accepted: 02/03/2014] [Indexed: 01/28/2023]
Abstract
Monomethylation of lysine 4 on histone H3 (H3K4me1) is a well-established feature of enhancers and promoters, although its function is unknown. Here, we uncover roles for H3K4me1 in diverse cell types. Remarkably, we find that MLL3/4 provokes monomethylation of promoter regions and the conditional repression of muscle and inflammatory response genes in myoblasts. During myogenesis, muscle genes are activated, lose MLL3 occupancy, and become H3K4-trimethylated through an alternative COMPASS complex. Monomethylation-mediated repression was not restricted to skeletal muscle. Together with H3K27me3 and H4K20me1, H3K4me1 was associated with transcriptional silencing in embryonic fibroblasts, macrophages, and human embryonic stem cells (ESCs). On promoters of active genes, we find that H3K4me1 spatially demarcates the recruitment of factors that interact with H3K4me3, including ING1, which, in turn, recruits Sin3A. Our findings point to a unique role for H3K4 monomethylation in establishing boundaries that restrict the recruitment of chromatin-modifying enzymes to defined regions within promoters.
Collapse
Affiliation(s)
- Jemmie Cheng
- Department of Pathology and Cancer Institute, Smilow Research Center, New York University School of Medicine, New York, NY 10016, USA
| | - Roy Blum
- Department of Pathology and Cancer Institute, Smilow Research Center, New York University School of Medicine, New York, NY 10016, USA
| | - Christopher Bowman
- Department of Pathology and Cancer Institute, Smilow Research Center, New York University School of Medicine, New York, NY 10016, USA
| | - Deqing Hu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Ali Shilatifard
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Steven Shen
- GTC, Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY 10016, USA
| | - Brian D Dynlacht
- Department of Pathology and Cancer Institute, Smilow Research Center, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
244
|
Jin S, Kim J, Willert T, Klein-Rodewald T, Garcia-Dominguez M, Mosqueira M, Fink R, Esposito I, Hofbauer LC, Charnay P, Kieslinger M. Ebf factors and MyoD cooperate to regulate muscle relaxation via Atp2a1. Nat Commun 2014; 5:3793. [PMID: 24786561 DOI: 10.1038/ncomms4793] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/02/2014] [Indexed: 01/08/2023] Open
Abstract
Myogenic regulatory factors such as MyoD and Myf5 lie at the core of vertebrate muscle differentiation. However, E-boxes, the cognate binding sites for these transcription factors, are not restricted to the promoters/enhancers of muscle cell-specific genes. Thus, the specificity in myogenic transcription is poorly defined. Here we describe the transcription factor Ebf3 as a new determinant of muscle cell-specific transcription. In the absence of Ebf3 the lung does not unfold at birth, resulting in respiratory failure and perinatal death. This is due to a hypercontractile diaphragm with impaired Ca(2+) efflux-related muscle functions. Expression of the Ca(2+) pump Serca1 (Atp2a1) is downregulated in the absence of Ebf3, and its transgenic expression rescues this phenotype. Ebf3 binds directly to the promoter of Atp2a1 and synergises with MyoD in the induction of Atp2a1. In skeletal muscle, the homologous family member Ebf1 is strongly expressed and together with MyoD induces Atp2a1. Thus, Ebf3 is a new regulator of terminal muscle differentiation in the diaphragm, and Ebf factors cooperate with MyoD in the induction of muscle-specific genes.
Collapse
Affiliation(s)
- Saihong Jin
- Institute of Clinical Molecular Biology and Tumor Genetics, Helmholtz Zentrum München, National Research Center for Environmental Health, Marchioninistrasse 25, 81377 Munich, Germany
| | - Jeehee Kim
- Institute of Clinical Molecular Biology and Tumor Genetics, Helmholtz Zentrum München, National Research Center for Environmental Health, Marchioninistrasse 25, 81377 Munich, Germany
| | - Torsten Willert
- Institute of Clinical Molecular Biology and Tumor Genetics, Helmholtz Zentrum München, National Research Center for Environmental Health, Marchioninistrasse 25, 81377 Munich, Germany
| | - Tanja Klein-Rodewald
- Institute of Pathology, Helmholtz Zentrum München, National Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764 Neuherberg, 81377 Munich, Germany
| | - Mario Garcia-Dominguez
- 1] Developmental Biology Section, Ecole Normale Supérieure, Rue d'Ulm 46, 75230 Paris, France [2] Stem Cells Department, CABIMER (CISC), Av Américo Vespucio, 41092 Sevilla, Spain
| | - Matias Mosqueira
- Medical Biophysics Unit, Institute of Physiology and Pathophysiology, University of Heidelberg, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Rainer Fink
- Medical Biophysics Unit, Institute of Physiology and Pathophysiology, University of Heidelberg, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Irene Esposito
- 1] Institute of Pathology, Helmholtz Zentrum München, National Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764 Neuherberg, 81377 Munich, Germany [2] Institute of Pathology, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany
| | - Lorenz C Hofbauer
- Division of Endocrinology, Diabetes and Metabolic Bone Diseases, Department of Medicine III, TU Dresden Medical Center, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Patrick Charnay
- Developmental Biology Section, Ecole Normale Supérieure, Rue d'Ulm 46, 75230 Paris, France
| | - Matthias Kieslinger
- Institute of Clinical Molecular Biology and Tumor Genetics, Helmholtz Zentrum München, National Research Center for Environmental Health, Marchioninistrasse 25, 81377 Munich, Germany
| |
Collapse
|
245
|
Gene regulatory networks and transcriptional mechanisms that control myogenesis. Dev Cell 2014; 28:225-38. [PMID: 24525185 DOI: 10.1016/j.devcel.2013.12.020] [Citation(s) in RCA: 439] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 12/24/2013] [Accepted: 12/27/2013] [Indexed: 12/11/2022]
Abstract
We discuss the upstream regulators of myogenesis that lead to the activation of myogenic determination genes and subsequent differentiation, focusing on the mouse model. Key upstream genes, such as Pax3 and Pax7, Six1 and Six4, or Pitx2, participate in gene regulatory networks at different sites of skeletal muscle formation. MicroRNAs also intervene, with emerging evidence for the role of other noncoding RNAs. Myogenic determination and subsequent differentiation depend on members of the MyoD family. We discuss new insights into mechanisms underlying the transcriptional activity of these factors.
Collapse
|
246
|
Villar D, Flicek P, Odom DT. Evolution of transcription factor binding in metazoans - mechanisms and functional implications. Nat Rev Genet 2014; 15:221-33. [PMID: 24590227 PMCID: PMC4175440 DOI: 10.1038/nrg3481] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Differences in transcription factor binding can contribute to organismal evolution by altering downstream gene expression programmes. Genome-wide studies in Drosophila melanogaster and mammals have revealed common quantitative and combinatorial properties of in vivo DNA binding, as well as marked differences in the rate and mechanisms of evolution of transcription factor binding in metazoans. Here, we review the recently discovered rapid 're-wiring' of in vivo transcription factor binding between related metazoan species and summarize general principles underlying the observed patterns of evolution. We then consider what might explain the differences in genome evolution between metazoan phyla and outline the conceptual and technological challenges facing this research field.
Collapse
Affiliation(s)
- Diego Villar
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB1 01SD, UK
| | - Duncan T Odom
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|
247
|
Saccone V, Consalvi S, Giordani L, Mozzetta C, Barozzi I, Sandoná M, Ryan T, Rojas-Muñoz A, Madaro L, Fasanaro P, Borsellino G, De Bardi M, Frigè G, Termanini A, Sun X, Rossant J, Bruneau BG, Mercola M, Minucci S, Puri PL. HDAC-regulated myomiRs control BAF60 variant exchange and direct the functional phenotype of fibro-adipogenic progenitors in dystrophic muscles. Genes Dev 2014; 28:841-57. [PMID: 24682306 PMCID: PMC4003277 DOI: 10.1101/gad.234468.113] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Fibro-adipogenic progenitors (FAPs) are important components of the skeletal muscle regenerative environment. Whether FAPs support muscle regeneration or promote fibro-adipogenic degeneration is emerging as a key determinant in the pathogenesis of muscular diseases, including Duchenne muscular dystrophy (DMD). However, the molecular mechanism that controls FAP lineage commitment and activity is currently unknown. We show here that an HDAC-myomiR-BAF60 variant network regulates the fate of FAPs in dystrophic muscles of mdx mice. Combinatorial analysis of gene expression microarray, genome-wide chromatin remodeling by nuclease accessibility (NA) combined with next-generation sequencing (NA-seq), small RNA sequencing (RNA-seq), and microRNA (miR) high-throughput screening (HTS) against SWI/SNF BAF60 variants revealed that HDAC inhibitors (HDACis) derepress a "latent" myogenic program in FAPs from dystrophic muscles at early stages of disease. Specifically, HDAC inhibition induces two core components of the myogenic transcriptional machinery, MYOD and BAF60C, and up-regulates the myogenic miRs (myomiRs) (miR-1.2, miR-133, and miR-206), which target the alternative BAF60 variants BAF60A and BAF60B, ultimately directing promyogenic differentiation while suppressing the fibro-adipogenic phenotype. In contrast, FAPs from late stage dystrophic muscles are resistant to HDACi-induced chromatin remodeling at myogenic loci and fail to activate the promyogenic phenotype. These results reveal a previously unappreciated disease stage-specific bipotency of mesenchimal cells within the regenerative environment of dystrophic muscles. Resolution of such bipotency by epigenetic intervention with HDACis provides a molecular rationale for the in situ reprogramming of target cells to promote therapeutic regeneration of dystrophic muscles.
Collapse
Affiliation(s)
- Valentina Saccone
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, 00143 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Wu H, Whitfield TW, Gordon JAR, Dobson JR, Tai PWL, van Wijnen AJ, Stein JL, Stein GS, Lian JB. Genomic occupancy of Runx2 with global expression profiling identifies a novel dimension to control of osteoblastogenesis. Genome Biol 2014; 15:R52. [PMID: 24655370 PMCID: PMC4056528 DOI: 10.1186/gb-2014-15-3-r52] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 03/21/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteogenesis is a highly regulated developmental process and continues during the turnover and repair of mature bone. Runx2, the master regulator of osteoblastogenesis, directs a transcriptional program essential for bone formation through genetic and epigenetic mechanisms. While individual Runx2 gene targets have been identified, further insights into the broad spectrum of Runx2 functions required for osteogenesis are needed. RESULTS By performing genome-wide characterization of Runx2 binding at the three major stages of osteoblast differentiation--proliferation, matrix deposition and mineralization--we identify Runx2-dependent regulatory networks driving bone formation. Using chromatin immunoprecipitation followed by high-throughput sequencing over the course of these stages, we identify approximately 80,000 significantly enriched regions of Runx2 binding throughout the mouse genome. These binding events exhibit distinct patterns during osteogenesis, and are associated with proximal promoters and also non-promoter regions: upstream, introns, exons, transcription termination site regions, and intergenic regions. These peaks were partitioned into clusters that are associated with genes in complex biological processes that support bone formation. Using Affymetrix expression profiling of differentiating osteoblasts depleted of Runx2, we identify novel Runx2 targets including Ezh2, a critical epigenetic regulator; Crabp2, a retinoic acid signaling component; Adamts4 and Tnfrsf19, two remodelers of the extracellular matrix. We demonstrate by luciferase assays that these novel biological targets are regulated by Runx2 occupancy at non-promoter regions. CONCLUSIONS Our data establish that Runx2 interactions with chromatin across the genome reveal novel genes, pathways and transcriptional mechanisms that contribute to the regulation of osteoblastogenesis.
Collapse
|
249
|
A comparison of midline and tracheal gene regulation during Drosophila development. PLoS One 2014; 9:e85518. [PMID: 24465586 PMCID: PMC3896416 DOI: 10.1371/journal.pone.0085518] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 11/28/2013] [Indexed: 11/19/2022] Open
Abstract
Within the Drosophila embryo, two related bHLH-PAS proteins, Single-minded and Trachealess, control development of the central nervous system midline and the trachea, respectively. These two proteins are bHLH-PAS transcription factors and independently form heterodimers with another bHLH-PAS protein, Tango. During early embryogenesis, expression of Single-minded is restricted to the midline and Trachealess to the trachea and salivary glands, whereas Tango is ubiquitously expressed. Both Single-minded/Tango and Trachealess/Tango heterodimers bind to the same DNA sequence, called the CNS midline element (CME) within cis-regulatory sequences of downstream target genes. While Single-minded/Tango and Trachealess/Tango activate some of the same genes in their respective tissues during embryogenesis, they also activate a number of different genes restricted to only certain tissues. The goal of this research is to understand how these two related heterodimers bind different enhancers to activate different genes, thereby regulating the development of functionally diverse tissues. Existing data indicates that Single-minded and Trachealess may bind to different co-factors restricted to various tissues, causing them to interact with the CME only within certain sequence contexts. This would lead to the activation of different target genes in different cell types. To understand how the context surrounding the CME is recognized by different bHLH-PAS heterodimers and their co-factors, we identified and analyzed novel enhancers that drive midline and/or tracheal expression and compared them to previously characterized enhancers. In addition, we tested expression of synthetic reporter genes containing the CME flanked by different sequences. Taken together, these experiments identify elements overrepresented within midline and tracheal enhancers and suggest that sequences immediately surrounding a CME help dictate whether a gene is expressed in the midline or trachea.
Collapse
|
250
|
Zhu B, Zhang M, Byrum SD, Tackett AJ, Davie JK. TBX2 blocks myogenesis and promotes proliferation in rhabdomyosarcoma cells. Int J Cancer 2014; 135:785-97. [PMID: 24470334 DOI: 10.1002/ijc.28721] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 12/23/2013] [Accepted: 01/02/2014] [Indexed: 11/11/2022]
Abstract
Rhabdomyosarcomas (RMSs) are the most frequent soft tissue sarcomas in children that share many features of developing skeletal muscle. We have discovered that a T-box family member, TBX2, is highly upregulated in tumor cells of both major RMS subtypes. TBX2 is a repressor that is often overexpressed in cancer cells and is thought to function in bypassing cell growth control, including repression of p14 and p21. The cell cycle regulator p21 is required for the terminal differentiation of skeletal muscle cells and is silenced in RMS cells. We have found that TBX2 interacts with the myogenic regulatory factors MyoD and myogenin and inhibits the activity of these factors. TBX2 is expressed in primary myoblasts and C2C12 cells, but is strongly downregulated upon differentiation. TBX2 recruits the histone deacetylase HDAC1 and is a potent inhibitor of the expression of muscle-specific genes and the cell cycle regulators, p21 and p14. TBX2 promotes the proliferation of RMS cells and either depletions of TBX2 or dominant negative TBX2 upregulate p21- and muscle-specific genes. Significantly, depletion or interference with TBX2 completely inhibits tumor growth in a xenograft assay, highlighting the oncogenic role of TBX2 in RMS cells. Thus, the data demonstrate that elevated expression of TBX2 contributes to the pathology of RMS cells by promoting proliferation and repressing differentiation-specific gene expression. These results show that deregulated TBX2 serves as an oncogene in RMS, suggesting that TBX2 may serve as a new diagnostic marker or therapeutic target for RMS tumors.
Collapse
Affiliation(s)
- Bo Zhu
- Department of Biochemistry and Molecular Biology and Simmons Cancer Institute, Southern Illinois University School of Medicine, Carbondale, IL
| | | | | | | | | |
Collapse
|