1
|
Liu J, Liu S, Gao H, Han L, Chu X, Sheng Y, Shou W, Wang Y, Liu Y, Wan J, Yang L. Genome-wide studies reveal the essential and opposite roles of ARID1A in controlling human cardiogenesis and neurogenesis from pluripotent stem cells. Genome Biol 2020; 21:169. [PMID: 32646524 PMCID: PMC7350744 DOI: 10.1186/s13059-020-02082-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Early human heart and brain development simultaneously occur during embryogenesis. Notably, in human newborns, congenital heart defects strongly associate with neurodevelopmental abnormalities, suggesting a common gene or complex underlying both cardiogenesis and neurogenesis. However, due to lack of in vivo studies, the molecular mechanisms that govern both early human heart and brain development remain elusive. RESULTS Here, we report ARID1A, a DNA-binding subunit of the SWI/SNF epigenetic complex, controls both neurogenesis and cardiogenesis from human embryonic stem cells (hESCs) through distinct mechanisms. Knockout-of-ARID1A (ARID1A-/-) leads to spontaneous differentiation of neural cells together with globally enhanced expression of neurogenic genes in undifferentiated hESCs. Additionally, when compared with WT hESCs, cardiac differentiation from ARID1A -/- hESCs is prominently suppressed, whereas neural differentiation is significantly promoted. Whole genome-wide scRNA-seq, ATAC-seq, and ChIP-seq analyses reveal that ARID1A is required to open chromatin accessibility on promoters of essential cardiogenic genes, and temporally associated with key cardiogenic transcriptional factors T and MEF2C during early cardiac development. However, during early neural development, transcription of most essential neurogenic genes is dependent on ARID1A, which can interact with a known neural restrictive silencer factor REST/NRSF. CONCLUSIONS We uncover the opposite roles by ARID1A to govern both early cardiac and neural development from pluripotent stem cells. Global chromatin accessibility on cardiogenic genes is dependent on ARID1A, whereas transcriptional activity of neurogenic genes is under control by ARID1A, possibly through ARID1A-REST/NRSF interaction.
Collapse
Affiliation(s)
- Juli Liu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut Street, R4 272, Indianapolis, IN, 46202, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Lei Han
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut Street, R4 272, Indianapolis, IN, 46202, USA
| | - Xiaona Chu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yi Sheng
- Department of Obstetrics, Gynecology & Reproductive Sciences, Magee-Women's Research Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Weinian Shou
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut Street, R4 272, Indianapolis, IN, 46202, USA
| | - Yue Wang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of BioHealth Informatics, Indiana University School of Informatics and Computing, Indiana University - Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of BioHealth Informatics, Indiana University School of Informatics and Computing, Indiana University - Purdue University Indianapolis, Indianapolis, IN, 46202, USA.
| | - Lei Yang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut Street, R4 272, Indianapolis, IN, 46202, USA.
| |
Collapse
|
2
|
Bultman SJ. SETD7 interacts with other chromatin-modifying factors to regulate cardiac development. Stem Cell Investig 2019; 6:14. [PMID: 31304180 DOI: 10.21037/sci.2019.05.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 05/20/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Scott J Bultman
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
3
|
Jia G, Preussner J, Chen X, Guenther S, Yuan X, Yekelchyk M, Kuenne C, Looso M, Zhou Y, Teichmann S, Braun T. Single cell RNA-seq and ATAC-seq analysis of cardiac progenitor cell transition states and lineage settlement. Nat Commun 2018; 9:4877. [PMID: 30451828 PMCID: PMC6242939 DOI: 10.1038/s41467-018-07307-6] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 10/27/2018] [Indexed: 01/01/2023] Open
Abstract
Formation and segregation of cell lineages forming the heart have been studied extensively but the underlying gene regulatory networks and epigenetic changes driving cell fate transitions during early cardiogenesis are still only partially understood. Here, we comprehensively characterize mouse cardiac progenitor cells (CPCs) marked by Nkx2-5 and Isl1 expression from E7.5 to E9.5 using single-cell RNA sequencing and transposase-accessible chromatin profiling (ATAC-seq). By leveraging on cell-to-cell transcriptome and chromatin accessibility heterogeneity, we identify different previously unknown cardiac subpopulations. Reconstruction of developmental trajectories reveal that multipotent Isl1+ CPC pass through an attractor state before separating into different developmental branches, whereas extended expression of Nkx2-5 commits CPC to an unidirectional cardiomyocyte fate. Furthermore, we show that CPC fate transitions are associated with distinct open chromatin states critically depending on Isl1 and Nkx2-5. Our data provide a model of transcriptional and epigenetic regulations during cardiac progenitor cell fate decisions at single-cell resolution.
Collapse
Affiliation(s)
- Guangshuai Jia
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Jens Preussner
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, 60596, Germany
| | - Xi Chen
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Stefan Guenther
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, 60596, Germany
| | - Xuejun Yuan
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, 60596, Germany
| | - Michail Yekelchyk
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, 60596, Germany
| | - Carsten Kuenne
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, 60596, Germany
| | - Mario Looso
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, 60596, Germany
| | - Yonggang Zhou
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Sarah Teichmann
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
- Theory of Condensed Matter, Cavendish Laboratory, 19 JJ Thomson Ave, Cambridge, CB3 0HE, UK
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, 60596, Germany.
| |
Collapse
|
4
|
A Loss of Function Screen of Epigenetic Modifiers and Splicing Factors during Early Stage of Cardiac Reprogramming. Stem Cells Int 2018; 2018:3814747. [PMID: 29743891 PMCID: PMC5878887 DOI: 10.1155/2018/3814747] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 12/04/2017] [Indexed: 02/07/2023] Open
Abstract
Direct reprogramming of cardiac fibroblasts (CFs) to induced cardiomyocytes (iCMs) is a newly emerged promising approach for cardiac regeneration, disease modeling, and drug discovery. However, its potential has been drastically limited due to the low reprogramming efficiency and largely unknown underlying molecular mechanisms. We have previously screened and identified epigenetic factors related to histone modification during iCM reprogramming. Here, we used shRNAs targeting an additional battery of epigenetic factors involved in chromatin remodeling and RNA splicing factors to further identify inhibitors and facilitators of direct cardiac reprogramming. Knockdown of RNA splicing factors Sf3a1 or Sf3b1 significantly reduced the percentage and total number of cardiac marker positive iCMs accompanied with generally repressed gene expression. Removal of another RNA splicing factor Zrsr2 promoted the acquisition of CM molecular features in CFs and mouse embryonic fibroblasts (MEFs) at both protein and mRNA levels. Moreover, a consistent increase of reprogramming efficiency was observed in CFs and MEFs treated with shRNAs targeting Bcor (component of BCOR complex superfamily) or Stag2 (component of cohesin complex). Our work thus reveals several additional epigenetic and splicing factors that are either inhibitory to or required for iCM reprogramming and highlights the importance of epigenetic regulation and RNA splicing process during cell fate conversion.
Collapse
|
5
|
Willis MS, Holley DW, Wang Z, Chen X, Quintana M, Jensen BC, Tannu M, Parker J, Jeyaraj D, Jain MK, Wolfram JA, Lee HG, Bultman SJ. BRG1 and BRM function antagonistically with c-MYC in adult cardiomyocytes to regulate conduction and contractility. J Mol Cell Cardiol 2017; 105:99-109. [PMID: 28232072 DOI: 10.1016/j.yjmcc.2017.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/27/2017] [Accepted: 02/15/2017] [Indexed: 12/15/2022]
Abstract
RATIONALE The contractile dysfunction that underlies heart failure involves perturbations in multiple biological processes ranging from metabolism to electrophysiology. Yet the epigenetic mechanisms that are altered in this disease state have not been elucidated. SWI/SNF chromatin-remodeling complexes are plausible candidates based on mouse knockout studies demonstrating a combined requirement for the BRG1 and BRM catalytic subunits in adult cardiomyocytes. Brg1/Brm double mutants exhibit metabolic and mitochondrial defects and are not viable although their cause of death has not been ascertained. OBJECTIVE To determine the cause of death of Brg1/Brm double-mutant mice, to test the hypothesis that BRG1 and BRM are required for cardiac contractility, and to identify relevant downstream target genes. METHODS AND RESULTS A tamoxifen-inducible gene-targeting strategy utilizing αMHC-Cre-ERT was implemented to delete both SWI/SNF catalytic subunits in adult cardiomyocytes. Brg1/Brm double-mutant mice were monitored by echocardiography and electrocardiography, and they underwent rapidly progressive ventricular dysfunction including conduction defects and arrhythmias that culminated in heart failure and death within 3weeks. Mechanistically, BRG1/BRM repressed c-Myc expression, and enforced expression of a DOX-inducible c-MYC trangene in mouse cardiomyocytes phenocopied the ventricular conduction defects observed in Brg1/Brm double mutants. BRG1/BRM and c-MYC had opposite effects on the expression of cardiac conduction genes, and the directionality was consistent with their respective loss- and gain-of-function phenotypes. To support the clinical relevance of this mechanism, BRG1/BRM occupancy was diminished at the same target genes in human heart failure cases compared to controls, and this correlated with increased c-MYC expression and decreased CX43 and SCN5A expression. CONCLUSION BRG1/BRM and c-MYC have an antagonistic relationship regulating the expression of cardiac conduction genes that maintain contractility, which is reminiscent of their antagonistic roles as a tumor suppressor and oncogene in cancer.
Collapse
Affiliation(s)
- Monte S Willis
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA; Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Darcy Wood Holley
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Zhongjing Wang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Xin Chen
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, 250021 Jinan, PR China
| | - Megan Quintana
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian C Jensen
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Manasi Tannu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, 250021 Jinan, PR China
| | - Joel Parker
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Darwin Jeyaraj
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; Harrington Discovery Institute, University Hospitals Harrington Heart & Vascular Institute, Cleveland, OH 44106, USA
| | - Mukesh K Jain
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; Harrington Discovery Institute, University Hospitals Harrington Heart & Vascular Institute, Cleveland, OH 44106, USA
| | - Julie A Wolfram
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Hyoung-Gon Lee
- Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA.
| | - Scott J Bultman
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
6
|
Vecoli C, Pulignani S, Andreassi MG. Genetic and Epigenetic Mechanisms Linking Air Pollution and Congenital Heart Disease. J Cardiovasc Dev Dis 2016; 3:jcdd3040032. [PMID: 29367575 PMCID: PMC5715723 DOI: 10.3390/jcdd3040032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/08/2016] [Accepted: 11/26/2016] [Indexed: 12/11/2022] Open
Abstract
Epidemiological studies strongly suggest that parental air pollutants exposure during the periconceptional period may play a major role in causing fetal/newborn malformations, including a frequent heterogeneity in the methods applied and a difficulty in estimating the clear effect of environmental toxicants. Moreover, only some couples exposed to toxicants during the pre-conception period give birth to a child with congenital anomalies. The reasons for such phenomena remain elusive but they can be explained by the individual, innate ability to metabolize these contaminants that eventually defines the ultimate dose of a biological active toxicant. In this paper, we reviewed the major evidence regarding the role of parental air pollutant exposure on congenital heart disease (CHD) risk as well as the modulating effect on detoxification systems. Finally, major epigenetic alterations induced by adverse environment contaminants have been revised as possible mechanisms altering a correct heart morphogenesis.
Collapse
Affiliation(s)
- Cecilia Vecoli
- Institute of Clinical Physiology-National Research Council (CNR), Via Moruzzi, 1 56124 Pisa, Italy.
| | - Silvia Pulignani
- Institute of Clinical Physiology-National Research Council (CNR), Via Moruzzi, 1 56124 Pisa, Italy.
| | - Maria Grazia Andreassi
- Institute of Clinical Physiology-National Research Council (CNR), Via Moruzzi, 1 56124 Pisa, Italy.
| |
Collapse
|
7
|
Nimura K, Yamamoto M, Takeichi M, Saga K, Takaoka K, Kawamura N, Nitta H, Nagano H, Ishino S, Tanaka T, Schwartz RJ, Aburatani H, Kaneda Y. Regulation of alternative polyadenylation by Nkx2-5 and Xrn2 during mouse heart development. eLife 2016; 5. [PMID: 27331609 PMCID: PMC4982761 DOI: 10.7554/elife.16030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/21/2016] [Indexed: 11/21/2022] Open
Abstract
Transcription factors organize gene expression profiles by regulating promoter activity. However, the role of transcription factors after transcription initiation is poorly understood. Here, we show that the homeoprotein Nkx2-5 and the 5’-3’ exonuclease Xrn2 are involved in the regulation of alternative polyadenylation (APA) during mouse heart development. Nkx2-5 occupied not only the transcription start sites (TSSs) but also the downstream regions of genes, serving to connect these regions in primary embryonic cardiomyocytes (eCMs). Nkx2-5 deficiency affected Xrn2 binding to target loci and resulted in increases in RNA polymerase II (RNAPII) occupancy and in the expression of mRNAs with long 3’untranslated regions (3’ UTRs) from genes related to heart development. siRNA-mediated suppression of Nkx2-5 and Xrn2 led to heart looping anomaly. Moreover, Nkx2-5 genetically interacts with Xrn2 because Nkx2-5+/-Xrn2+/-, but neither Nkx2-5+/-nor Xrn2+/-, newborns exhibited a defect in ventricular septum formation, suggesting that the association between Nkx2-5 and Xrn2 is essential for heart development. Our results indicate that Nkx2-5 regulates not only the initiation but also the usage of poly(A) sites during heart development. Our findings suggest that tissue-specific transcription factors is involved in the regulation of APA. DOI:http://dx.doi.org/10.7554/eLife.16030.001 About one in every hundred babies is born with problems that either affect the structure of the heart or how it works. These problems are known as congenital heart disease, and result when the development of the heart is disrupted. How the heart develops is determined by thousands of genes whose activity or “expression” must be precisely regulated. Proteins called transcription factors can control gene expression; therefore, researchers may discover new ways of treating congenital heart disease if they can understand how transcription factors work during normal heart development. To produce a protein, the information in a gene must first be “transcribed” to form a molecule of messenger RNA (mRNA). Not all of the mRNA sequence is subsequently “translated” to form the protein; this includes a stretch at the end of the mRNA called the 3’ untranslated region. The length of the 3’ untranslated region for a particular mRNA may vary depending on the type of cell it has been produced in, and this length can influence how efficiently the mRNA is translated to form a protein. However, it was not clear what changes the length of the 3’ untranslated region. Nimura et al. have now studied mice to investigate the role of a transcription factor called Nkx2-5, which was known to be important for heart development. This revealed that in addition to its expected role in starting the transcription of genes that are important for heart development, Nkx2-5 also controls the length of 3’ untranslated regions of certain mRNAs. To do so, Nkx2-5 binds to a protein called Xrn2 that stops transcription when the end of the gene is reached. Mouse embryos that lacked Nkx2-5 produced mRNAs containing long 3’ untranslated regions from genes related to the development of the heart. Furthermore, suppressing the activity of both Nkx2-5 and Xrn2 resulted in the embryos developing heart defects. The findings of Nimura et al. suggest that transcription factors found in specific tissues are responsible for the different lengths of 3’ untranslated regions in mRNAs in different tissues. Furthermore, incorrectly regulating the length of these regions appears to be linked to the development of congenital heart disease. The next step is to understand exactly how the failure to correctly regulate the length of 3’ untranslated regions contributes to congenital heart disease. DOI:http://dx.doi.org/10.7554/eLife.16030.002
Collapse
Affiliation(s)
- Keisuke Nimura
- Division of Gene Therapy Science, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masamichi Yamamoto
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Makiko Takeichi
- Division of Gene Therapy Science, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kotaro Saga
- Division of Gene Therapy Science, Osaka University Graduate School of Medicine, Suita, Japan
| | - Katsuyoshi Takaoka
- Developmental Genetics Group, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Norihiko Kawamura
- Division of Gene Therapy Science, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hirohisa Nitta
- Division of Gene Therapy Science, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiromichi Nagano
- Division of Gene Therapy Science, Osaka University Graduate School of Medicine, Suita, Japan
| | - Saki Ishino
- Center for Medical Research and Education, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tatsuya Tanaka
- Center for Medical Research and Education, Osaka University Graduate School of Medicine, Suita, Japan
| | - Robert J Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, Unites States
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| | - Yasufumi Kaneda
- Division of Gene Therapy Science, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
8
|
van Weerd JH, Christoffels VM. The formation and function of the cardiac conduction system. Development 2016; 143:197-210. [PMID: 26786210 DOI: 10.1242/dev.124883] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The cardiac conduction system (CCS) consists of distinctive components that initiate and conduct the electrical impulse required for the coordinated contraction of the cardiac chambers. CCS development involves complex regulatory networks that act in stage-, tissue- and dose-dependent manners, and recent findings indicate that the activity of these networks is sensitive to common genetic variants associated with cardiac arrhythmias. Here, we review how these findings have provided novel insights into the regulatory mechanisms and transcriptional networks underlying CCS formation and function.
Collapse
Affiliation(s)
- Jan Hendrik van Weerd
- Department of Anatomy, Embryology & Physiology, Academic Medical Center, Amsterdam 1105 AZ, The Netherlands
| | - Vincent M Christoffels
- Department of Anatomy, Embryology & Physiology, Academic Medical Center, Amsterdam 1105 AZ, The Netherlands
| |
Collapse
|
9
|
The Chromatin Remodeling Complex Chd4/NuRD Controls Striated Muscle Identity and Metabolic Homeostasis. Cell Metab 2016; 23:881-92. [PMID: 27166947 DOI: 10.1016/j.cmet.2016.04.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 02/19/2016] [Accepted: 04/13/2016] [Indexed: 01/01/2023]
Abstract
Heart muscle maintains blood circulation, while skeletal muscle powers skeletal movement. Despite having similar myofibrilar sarcomeric structures, these striated muscles differentially express specific sarcomere components to meet their distinct contractile requirements. The mechanism responsible is still unclear. We show here that preservation of the identity of the two striated muscle types depends on epigenetic repression of the alternate lineage gene program by the chromatin remodeling complex Chd4/NuRD. Loss of Chd4 in the heart triggers aberrant expression of the skeletal muscle program, causing severe cardiomyopathy and sudden death. Conversely, genetic depletion of Chd4 in skeletal muscle causes inappropriate expression of cardiac genes and myopathy. In both striated tissues, mitochondrial function was also dependent on the Chd4/NuRD complex. We conclude that an epigenetic mechanism controls cardiac and skeletal muscle structural and metabolic identities and that loss of this regulation leads to hybrid striated muscle tissues incompatible with life.
Collapse
|
10
|
Abstract
With the impressive advancement in high-throughput 'omics' technologies over the past two decades, epigenetic mechanisms have emerged as the regulatory interface between the genome and environmental factors. These mechanisms include DNA methylation, histone modifications, ATP-dependent chromatin remodeling and RNA-based mechanisms. Their highly interdependent and coordinated action modulates the chromatin structure controlling access of the transcription machinery and thereby regulating expression of target genes. Given the rather limited proliferative capability of human cardiomyocytes, epigenetic regulation appears to play a particularly important role in the myocardium. The highly dynamic nature of the epigenome allows the heart to adapt to environmental challenges and to respond quickly and properly to cardiac stress. It is now becoming evident that histone-modifying and chromatin-remodeling enzymes as well as numerous non-coding RNAs play critical roles in cardiac development and function, while their dysregulation contributes to the onset and development of pathological cardiac remodeling culminating in HF. This review focuses on up-to-date knowledge about the epigenetic mechanisms and highlights their emerging role in the healthy and failing heart. Uncovering the determinants of epigenetic regulation holds great promise to accelerate the development of successful new diagnostic and therapeutic strategies in human cardiac disease.
Collapse
Affiliation(s)
- José Marín-García
- The Molecular Cardiology and Neuromuscular Institute, 75 Raritan Ave., Highland Park, NJ, 08904, USA,
| | | |
Collapse
|
11
|
Cui H, Schlesinger J, Schoenhals S, Tönjes M, Dunkel I, Meierhofer D, Cano E, Schulz K, Berger MF, Haack T, Abdelilah-Seyfried S, Bulyk ML, Sauer S, Sperling SR. Phosphorylation of the chromatin remodeling factor DPF3a induces cardiac hypertrophy through releasing HEY repressors from DNA. Nucleic Acids Res 2015; 44:2538-53. [PMID: 26582913 PMCID: PMC4824069 DOI: 10.1093/nar/gkv1244] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 11/01/2015] [Indexed: 01/09/2023] Open
Abstract
DPF3 (BAF45c) is a member of the BAF chromatin remodeling complex. Two isoforms have been described, namely DPF3a and DPF3b. The latter binds to acetylated and methylated lysine residues of histones. Here, we elaborate on the role of DPF3a and describe a novel pathway of cardiac gene transcription leading to pathological cardiac hypertrophy. Upon hypertrophic stimuli, casein kinase 2 phosphorylates DPF3a at serine 348. This initiates the interaction of DPF3a with the transcriptional repressors HEY, followed by the release of HEY from the DNA. Moreover, BRG1 is bound by DPF3a, and is thus recruited to HEY genomic targets upon interaction of the two components. Consequently, the transcription of downstream targets such as NPPA and GATA4 is initiated and pathological cardiac hypertrophy is established. In human, DPF3a is significantly up-regulated in hypertrophic hearts of patients with hypertrophic cardiomyopathy or aortic stenosis. Taken together, we show that activation of DPF3a upon hypertrophic stimuli switches cardiac fetal gene expression from being silenced by HEY to being activated by BRG1. Thus, we present a novel pathway for pathological cardiac hypertrophy, whose inhibition is a long-term therapeutic goal for the treatment of the course of heart failure.
Collapse
Affiliation(s)
- Huanhuan Cui
- Department of Cardiovascular Genetics, Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany Group of Cardiovascular Genetics, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Jenny Schlesinger
- Department of Cardiovascular Genetics, Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany Group of Cardiovascular Genetics, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Sophia Schoenhals
- Department of Cardiovascular Genetics, Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Martje Tönjes
- Group of Cardiovascular Genetics, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Ilona Dunkel
- Group of Cardiovascular Genetics, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - David Meierhofer
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Elena Cano
- Department of Cardiovascular Genetics, Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Kerstin Schulz
- Department of Cardiovascular Genetics, Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Michael F Berger
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA Committee on Higher Degrees in Biophysics, Harvard University, Cambridge, MA 02138, USA
| | - Timm Haack
- Hannover Medical School, Institute of Molecular Biology, Carl-Neuberg Str. 1, D-30625 Hannover, Germany
| | - Salim Abdelilah-Seyfried
- Hannover Medical School, Institute of Molecular Biology, Carl-Neuberg Str. 1, D-30625 Hannover, Germany Potsdam University, Institute of Biochemistry and Biology, Department of Animal Physiology, Karl-Liebknecht Str. 24-25, 14476 Potsdam-Golm, Germany
| | - Martha L Bulyk
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA Committee on Higher Degrees in Biophysics, Harvard University, Cambridge, MA 02138, USA Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Sascha Sauer
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany CU Systems Medicine, University of Würzburg, 97080 Würzburg, Germany
| | - Silke R Sperling
- Department of Cardiovascular Genetics, Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany Group of Cardiovascular Genetics, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| |
Collapse
|
12
|
Elucidating the mechanisms of transcription regulation during heart development by next-generation sequencing. J Hum Genet 2015. [PMID: 26202577 DOI: 10.1038/jhg.2015.84] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Dysregulation of transcription is associated with the pathogenesis of cardiovascular diseases, including congenital heart diseases and heart failure. However, it remains unclear how transcription factors regulate transcription in the heart and which genes are associated with cardiovascular diseases in humans. Development of genome-wide analyses using next-generation sequencers provides powerful methods to determine how these transcription factors and chromatin regulators control gene expressions and to identify causative genes in cardiovascular diseases. These technologies have revealed that transcription during heart development is elaborately regulated by multiple cardiac transcription factors. In this review, we discuss the recent progress toward understanding the molecular mechanisms of how transcriptional dysregulation leads to cardiovascular diseases.
Collapse
|
13
|
Vecoli C, Pulignani S, Foffa I, Andreassi MG. Congenital heart disease: the crossroads of genetics, epigenetics and environment. Curr Genomics 2014; 15:390-9. [PMID: 25435801 PMCID: PMC4245698 DOI: 10.2174/1389202915666140716175634] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 06/24/2014] [Accepted: 07/07/2014] [Indexed: 02/07/2023] Open
Abstract
Congenital heart diseases (CHDs) are recognized as the most common type of birth malformations. Although recent advances in pre- and neonatal diagnosis as well as in surgical procedures have reduced the morbidity and mortality for many CHD, the etiology for CHD remains undefined. In non-syndromic and isolated (without a familial history or a Mendelian inheritance) forms of CHDs, a multifactorial pathogenesis with interplay between inherited and non-inherited causes is recognized. In this paper, we discuss the current knowledge of the potential molecular mechanisms, mediating abnormal cardiac development in non-syndromic and isolated CHD, including mutations in cardiac transcription factors, the role of somatic mutations and epigenetic alterations as well as the influence of gene-environment interactions. In the near future, the advent of high-throughput genomic technologies with the integration of system biology will expand our understanding of isolated, non-syndromic CHDs for their prevention, early diagnosis and therapy.
Collapse
Affiliation(s)
| | | | - Ilenia Foffa
- CNR, Institute of Clinical Physiology, Massa, Italy
| | - Maria Grazia Andreassi
- CNR, Institute of Clinical Physiology, Pisa, Italy ; Fondazione Toscana G.Monasterio, Massa, Italy
| |
Collapse
|
14
|
Garnatz AS, Gao Z, Broman M, Martens S, Earley JU, Svensson EC. FOG-2 mediated recruitment of the NuRD complex regulates cardiomyocyte proliferation during heart development. Dev Biol 2014; 395:50-61. [PMID: 25196150 DOI: 10.1016/j.ydbio.2014.08.030] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 08/05/2014] [Accepted: 08/26/2014] [Indexed: 12/21/2022]
Abstract
FOG-2 is a multi-zinc finger protein that binds the transcriptional activator GATA4 and modulates GATA4-mediated regulation of target genes during heart development. Our previous work has demonstrated that the Nucleosome Remodeling and Deacetylase (NuRD) complex physically interacts with FOG-2 and is necessary for FOG-2 mediated repression of GATA4 activity in vitro. However, the relevance of this interaction for FOG-2 function in vivo has remained unclear. In this report, we demonstrate the importance of FOG-2/NuRD interaction through the generation and characterization of mice homozygous for a mutation in FOG-2 that disrupts NuRD binding (FOG-2(R3K5A)). These mice exhibit a perinatal lethality and have multiple cardiac malformations, including ventricular and atrial septal defects and a thin ventricular myocardium. To investigate the etiology of the thin myocardium, we measured the rate of cardiomyocyte proliferation in wild-type and FOG-2(R3K5A) developing hearts. We found cardiomyocyte proliferation was reduced by 31±8% in FOG-2(R3K5A) mice. Gene expression analysis indicated that the cell cycle inhibitor Cdkn1a (p21(cip1)) is up-regulated 2.0±0.2-fold in FOG-2(R3K5A) hearts. In addition, we demonstrate that FOG-2 can directly repress the activity of the Cdkn1a gene promoter, suggesting a model by which FOG-2/NuRD promotes ventricular wall thickening by repression of this cell cycle inhibitor. Consistent with this notion, the genetic ablation of Cdkn1a in FOG-2(R3K5A) mice leads to an improvement in left ventricular function and a partial rescue of left ventricular wall thickness. Taken together, our results define a novel mechanism in which FOG-2/NuRD interaction is required for cardiomyocyte proliferation by directly down-regulating the cell cycle inhibitor Cdkn1a during heart development.
Collapse
Affiliation(s)
- Audrey S Garnatz
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Zhiguang Gao
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael Broman
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Spencer Martens
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Judy U Earley
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Eric C Svensson
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
15
|
Baf250a orchestrates an epigenetic pathway to repress the Nkx2.5-directed contractile cardiomyocyte program in the sinoatrial node. Cell Res 2014; 24:1201-13. [PMID: 25145359 PMCID: PMC4185344 DOI: 10.1038/cr.2014.113] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 06/30/2014] [Accepted: 07/15/2014] [Indexed: 02/06/2023] Open
Abstract
The sinoatrial node (SAN) is essential for rhythmic beating of the heart; however, our understanding of what controls proper functioning of the SAN remains primitive. To explore molecular control of SAN function, we specifically deleted Baf250a, a key regulatory component of the ATP-dependent chromatin remodeling complex SWI/SNF, in the SAN. Deletion of Baf250a in the SAN led to sinus bradycardia. Time series analysis of dysregulated genes after deletion of Baf250a reveals a transcriptional hierarchy maintaining pacemaker cell identity, i.e., Baf250a activates the expression of Tbx3, and Baf250a, Tbx3 and histone deacetylase 3 coordinately repress the expression of Nkx2.5. Disruption of this repressive pathway switches on expression of Nkx2.5, which stimulates expression of Gata4 and Tbx5. These three cardiac transcription factors further turn on a contractile cardiomyocyte program in the SAN, which eventually leads to sick sinus disease (SSD). Our study suggests that disruption of key genetic pathways regulating cardiac lineage segregation may cause SSD and cardiac arrhythmias in general.
Collapse
|
16
|
Abstract
The development of specialized helper T cells has garnered much attention because of their critical role in coordinating the immune response to invading pathogens. Recent research emphasizing novel functions for specialized helper T cells in a variety of infectious disease settings, as well as autoimmune states, has reshaped our view on the capabilities of helper T cells. Notably, one previously underappreciated aspect of the lifespan of helper T cells is that they often retain the capacity to respond to changes in the environment by altering the composition of helper T cell lineage-specifying transcription factors they express, which, in turn, changes their phenotype. This emerging realization is changing our views on the stability versus flexibility of specialized helper T cell subtypes. Now, there is a new concerted effort to define the mechanistic events that contribute to the potential for flexibility in specialized helper T cell gene expression programs in the different environmental circumstances that allow for the re-expression of helper T cell lineage-specifying transcription factors. In addition, we are also now beginning to appreciate that "helper T cell" lineage-specifying transcription factors are expressed in diverse types of innate and adaptive immune cells and this may allow them to play roles in coordinating aspects of the immune response. Our current challenges include defining the conserved mechanisms that are utilized by these lineage-specifying transcription factors to coordinate gene expression programs in different settings as well as the mechanistic events that contribute to the differential downstream consequences that these factors mediate in unique cellular environments. In this review, we will explore our evolving views on these topics, often times using the Th1-lineage-specifying transcription factor T-bet as an example.
Collapse
|
17
|
Singh AP, Archer TK. Analysis of the SWI/SNF chromatin-remodeling complex during early heart development and BAF250a repression cardiac gene transcription during P19 cell differentiation. Nucleic Acids Res 2013; 42:2958-75. [PMID: 24335282 PMCID: PMC3950667 DOI: 10.1093/nar/gkt1232] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The regulatory networks of differentiation programs and the molecular mechanisms of lineage-specific gene regulation in mammalian embryos remain only partially defined. We document differential expression and temporal switching of BRG1-associated factor (BAF) subunits, core pluripotency factors and cardiac-specific genes during post-implantation development and subsequent early organogenesis. Using affinity purification of BRG1 ATPase coupled to mass spectrometry, we characterized the cardiac-enriched remodeling complexes present in E8.5 mouse embryos. The relative abundance and combinatorial assembly of the BAF subunits provides functional specificity to Switch/Sucrose NonFermentable (SWI/SNF) complexes resulting in a unique gene expression profile in the developing heart. Remarkably, the specific depletion of the BAF250a subunit demonstrated differential effects on cardiac-specific gene expression and resulted in arrhythmic contracting cardiomyocytes in vitro. Indeed, the BAF250a physically interacts and functionally cooperates with Nucleosome Remodeling and Histone Deacetylase (NURD) complex subunits to repressively regulate chromatin structure of the cardiac genes by switching open and poised chromatin marks associated with active and repressed gene expression. Finally, BAF250a expression modulates BRG1 occupancy at the loci of cardiac genes regulatory regions in P19 cell differentiation. These findings reveal specialized and novel cardiac-enriched SWI/SNF chromatin-remodeling complexes, which are required for heart formation and critical for cardiac gene expression regulation at the early stages of heart development.
Collapse
Affiliation(s)
- Ajeet Pratap Singh
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | |
Collapse
|
18
|
Monte E, Mouillesseaux K, Chen H, Kimball T, Ren S, Wang Y, Chen JN, Vondriska TM, Franklin S. Systems proteomics of cardiac chromatin identifies nucleolin as a regulator of growth and cellular plasticity in cardiomyocytes. Am J Physiol Heart Circ Physiol 2013; 305:H1624-38. [PMID: 24077883 PMCID: PMC3882469 DOI: 10.1152/ajpheart.00529.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 09/10/2013] [Indexed: 11/22/2022]
Abstract
Myocyte hypertrophy antecedent to heart failure involves changes in global gene expression, although the preceding mechanisms to coordinate DNA accessibility on a genomic scale are unknown. Chromatin-associated proteins alter chromatin structure by changing their association with DNA, thereby altering the gene expression profile. Little is known about the global changes in chromatin subproteomes that accompany heart failure, and the mechanisms by which these proteins alter chromatin structure. The present study tests the fundamental hypothesis that cardiac growth and plasticity in the setting of disease recapitulates conserved developmental chromatin remodeling events. We used quantitative proteomics to identify chromatin-associated proteins extracted via detergent and to quantify changes in their abundance during disease. Our study identified 321 proteins in this subproteome, demonstrating it to have modest conservation (37%) with that revealed using strong acid. Of these proteins, 176 exhibited altered expression during cardiac hypertrophy and failure; we conducted extensive functional characterization of one of these proteins, Nucleolin. Morpholino-based knockdown of nucleolin nearly abolished protein expression but surprisingly had little impact on gross morphological development. However, hearts of fish lacking Nucleolin displayed severe developmental impairment, abnormal chamber patterning and functional deficits, ostensibly due to defects in cardiac looping and myocyte differentiation. The mechanisms underlying these defects involve perturbed bone morphogenetic protein 4 expression, decreased rRNA transcription, and a shift to more heterochromatic chromatin. This study reports the quantitative analysis of a new chromatin subproteome in the normal and diseased mouse heart. Validation studies in the complementary model system of zebrafish examine the role of Nucleolin to orchestrate genomic reprogramming events shared between development and disease.
Collapse
Affiliation(s)
- Emma Monte
- Department of Anesthesiology, University of California, Los Angeles, Los Angeles, California
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Liu Y, Harmelink C, Peng Y, Chen Y, Wang Q, Jiao K. CHD7 interacts with BMP R-SMADs to epigenetically regulate cardiogenesis in mice. Hum Mol Genet 2013; 23:2145-56. [PMID: 24293546 DOI: 10.1093/hmg/ddt610] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Haploinsufficiency for CHD7, an ATP-dependent nucleosome remodeling factor, is the leading cause of CHARGE syndrome. While congenital heart defects (CHDs) are major clinical features of CHARGE syndrome, affecting >75% of patients, it remains unclear whether CHD7 can directly regulate cardiogenic genes in embryos. Our complementary yeast two-hybrid and biochemical assays reveal that CHD7 is a novel interaction partner of canonical BMP signaling pathway nuclear mediators, SMAD1/5/8, in the embryonic heart. Moreover, CHD7 associates in a BMP-dependent manner with the enhancers of a critical cardiac transcription factor, Nkx2.5, that contain functional SMAD1-binding elements. Both the active epigenetic signature of Nkx2.5 regulatory elements and its proper expression in cardiomyocytes require CHD7. Finally, inactivation of Chd7 in mice impairs multiple BMP signaling-regulated cardiogenic processes. Our results thus support the model that CHD7 is recruited by SMAD1/5/8 to the enhancers of BMP-targeted cardiogenic genes to epigenetically regulate their expression. Impaired BMP activities in embryonic hearts may thus have a major contribution to CHDs in CHARGE syndrome.
Collapse
Affiliation(s)
- Yuelong Liu
- Department of Genetics and Department of Cell, Developmental and Integrative Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
20
|
Martinez-Fernandez A, Li X, Hartjes KA, Terzic A, Nelson TJ. Natural cardiogenesis-based template predicts cardiogenic potential of induced pluripotent stem cell lines. ACTA ACUST UNITED AC 2013; 6:462-71. [PMID: 24036272 DOI: 10.1161/circgenetics.113.000045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cardiac development is a complex process resulting in an integrated, multilineage tissue with developmental corruption in early embryogenesis leading to congenital heart disease. Interrogation of individual genes has provided the backbone for cardiac developmental biology, yet a comprehensive transcriptome derived from natural cardiogenesis is required to gauge innate developmental milestones. METHODS AND RESULTS Stage-specific cardiac structures were dissected from 8 distinctive mouse embryonic time points to produce genome-wide expressome analysis across cardiogenesis. With reference to this native cardiogenic expression roadmap, divergent induced pluripotent stem cell-derived cardiac expression profiles were mapped from procardiogenic 3-factor (SOX2, OCT4, KLF4) and less-cardiogenic 4-factor (plus c-MYC) reprogrammed cells. Expression of cardiac-related genes from 3-factor-induced pluripotent stem cell differentiated in vitro at days 5 and 11 and recapitulated expression profiles of natural embryos at days E7.5-E8.5 and E14.5-E18.5, respectively. By contrast, 4-factor-induced pluripotent stem cells demonstrated incomplete cardiogenic gene expression profiles beginning at day 5 of differentiation. Differential gene expression within the pluripotent state revealed 23 distinguishing candidate genes among pluripotent cell lines with divergent cardiogenic potentials. A confirmed panel of 12 genes, differentially expressed between high and low cardiogenic lines, was transformed into a predictive score sufficient to discriminate individual induced pluripotent stem cell lines according to relative cardiogenic potential. CONCLUSIONS Transcriptome analysis attuned to natural embryonic cardiogenesis provides a robust platform to probe coordinated cardiac specification and maturation from bioengineered stem cell-based model systems. A panel of developmental-related genes allowed differential prognosis of cardiogenic competency, thus prioritizing cell lines according to natural blueprint to streamline functional applications.
Collapse
Affiliation(s)
- Almudena Martinez-Fernandez
- Division of Cardiovascular Diseases, Department of Medicine, Department of Molecular Pharmacology and Experimental Therapeutics, Division of General Internal Medicine Transplant Center, Division of Biomedical Statistics and Informatics, and Center for Regenerative Medicine, Mayo Clinic, Rochester, MN
| | | | | | | | | |
Collapse
|
21
|
The G-protein-coupled receptor APJ is expressed in the second heart field and regulates Cerberus–Baf60c axis in embryonic stem cell cardiomyogenesis. Cardiovasc Res 2013; 100:95-104. [DOI: 10.1093/cvr/cvt166] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
22
|
Corsten-Janssen N, Kerstjens-Frederikse WS, du Marchie Sarvaas GJ, Baardman ME, Bakker MK, Bergman JE, Hove HD, Heimdal KR, Rustad CF, Hennekam RC, Hofstra RM, Hoefsloot LH, Van Ravenswaaij-Arts CM, Kapusta L. The Cardiac Phenotype in Patients With a
CHD7
Mutation. ACTA ACUST UNITED AC 2013; 6:248-54. [DOI: 10.1161/circgenetics.113.000054] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Loss-of-function mutations in
CHD7
cause Coloboma, Heart Disease, Atresia of Choanae, Retardation of Growth and/or Development, Genital Hypoplasia, and Ear Abnormalities With or Without Deafness (CHARGE) syndrome, a variable combination of multiple congenital malformations including heart defects. Heart defects are reported in 70% to 92% of patients with a
CHD7
mutation, but most studies are small and do not provide a detailed classification of the defects. We present the first, detailed, descriptive study on the cardiac phenotype of 299 patients with a
CHD7
mutation and discuss the role of CHD7 in cardiac development.
Methods and Results—
We collected information on congenital heart defects in 299 patients with a pathogenic
CHD7
mutation, of whom 220 (74%) had a congenital heart defect. Detailed information on the heart defects was available for 202 of these patients. We classified the heart defects based on embryonic cardiac development and compared the distribution to 1007 equally classified nonsyndromic heart defects of patients registered by EUROCAT, a European Registry of Congenital Anomalies. Heart defects are highly variable in patients with
CHD7
mutations, but atrioventricular septal defects and conotruncal heart defects are over-represented. Sex did not have an effect on the presence of heart defects, but truncating
CHD7
mutations resulted in a heart defect significantly more often than missense or splice-site mutations (χ
2
,
P
<0.001).
Conclusions—
CHD7 plays an important role in cardiac development, given that we found a wide range of heart defects in 74% of a large cohort of patients with a CHD7 mutation. Conotruncal defects and atrioventricular septal defects are over-represented in patients with
CHD7
mutations compared with patients with nonsyndromic heart defects.
Collapse
Affiliation(s)
- Nicole Corsten-Janssen
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| | - Wilhelmina S. Kerstjens-Frederikse
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| | - Gideon J. du Marchie Sarvaas
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| | - Maria E. Baardman
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| | - Marian K. Bakker
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| | - Jorieke E.H. Bergman
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| | - Hanne D. Hove
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| | - Ketil R. Heimdal
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| | - Cecilie F. Rustad
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| | - Raoul C.M. Hennekam
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| | - Robert M.W. Hofstra
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| | - Lies H. Hoefsloot
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| | - Conny M.A. Van Ravenswaaij-Arts
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| | - Livia Kapusta
- From the Department of Genetics (N.C.-J., W.S.K.-F., M.E.B., M.K.B., J.E.H.B., C.M.A.V.R.-A.) and Center for Congenital Heart Diseases (G.J.d.M.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark (H.D.H.); Department of Medical Genetics, Oslo University Hospital, Oslo, Norway (K.R.H., C.F.R.); Department of Pediatrics and Genetics, Academic Medical Center, University of Amsterdam,
| |
Collapse
|
23
|
Abstract
Congenital heart disease (CHD) is the most common congenital anomaly in newborn babies. Cardiac malformations have been produced in multiple experimental animal models, by perturbing selected molecules that function in the developmental pathways involved in myocyte specification, differentiation, or cardiac morphogenesis. In contrast, the precise genetic, epigenetic, or environmental basis for these perturbations in humans remains poorly understood. Over the past few decades, researchers have tried to bridge this knowledge gap through conventional genome-wide analyses of rare Mendelian CHD families, and by sequencing candidate genes in CHD cohorts. Although yielding few, usually highly penetrant, disease gene mutations, these discoveries provided 3 notable insights. First, human CHD mutations impact a heterogeneous set of molecules that orchestrate cardiac development. Second, CHD mutations often alter gene/protein dosage. Third, identical pathogenic CHD mutations cause a variety of distinct malformations, implying that higher order interactions account for particular CHD phenotypes. The advent of contemporary genomic technologies including single nucleotide polymorphism arrays, next-generation sequencing, and copy number variant platforms are accelerating the discovery of genetic causes of CHD. Importantly, these approaches enable study of sporadic cases, the most common presentation of CHD. Emerging results from ongoing genomic efforts have validated earlier observations learned from the monogenic CHD families. In this review, we explore how continued use of these technologies and integration of systems biology is expected to expand our understanding of the genetic architecture of CHD.
Collapse
Affiliation(s)
- Akl C Fahed
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
24
|
Bruneau BG. Signaling and transcriptional networks in heart development and regeneration. Cold Spring Harb Perspect Biol 2013; 5:a008292. [PMID: 23457256 DOI: 10.1101/cshperspect.a008292] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The mammalian heart is the first functional organ, the first indicator of life. Its normal formation and function are essential for fetal life. Defects in heart formation lead to congenital heart defects, underscoring the finesse with which the heart is assembled. Understanding the regulatory networks controlling heart development have led to significant insights into its lineage origins and morphogenesis and illuminated important aspects of mammalian embryology, while providing insights into human congenital heart disease. The mammalian heart has very little regenerative potential, and thus, any damage to the heart is life threatening and permanent. Knowledge of the developing heart is important for effective strategies of cardiac regeneration, providing new hope for future treatments for heart disease. Although we still have an incomplete picture of the mechanisms controlling development of the mammalian heart, our current knowledge has important implications for embryology and better understanding of human heart disease.
Collapse
Affiliation(s)
- Benoit G Bruneau
- Gladstone Institute of Cardiovascular Disease, San Francisco, California 94158, and Department of Pediatrics and Cardiovascular Research Institute, University of California, San Francisco, California 94158, USA.
| |
Collapse
|
25
|
Gregoire S, Karra R, Passer D, Deutsch MA, Krane M, Feistritzer R, Sturzu A, Domian I, Saga Y, Wu SM. Essential and unexpected role of Yin Yang 1 to promote mesodermal cardiac differentiation. Circ Res 2013; 112:900-10. [PMID: 23307821 DOI: 10.1161/circresaha.113.259259] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
RATIONALE Cardiogenesis is regulated by a complex interplay between transcription factors. However, little is known about how these interactions regulate the transition from mesodermal precursors to cardiac progenitor cells (CPCs). OBJECTIVE To identify novel regulators of mesodermal cardiac lineage commitment. METHODS AND RESULTS We performed a bioinformatic-based transcription factor binding site analysis on upstream promoter regions of genes that are enriched in embryonic stem cell-derived CPCs. From 32 candidate transcription factors screened, we found that Yin Yang 1 (YY1), a repressor of sarcomeric gene expression, is present in CPCs in vivo. Interestingly, we uncovered the ability of YY1 to transcriptionally activate Nkx2.5, a key marker of early cardiogenic commitment. YY1 regulates Nkx2.5 expression via a 2.1-kb cardiac-specific enhancer as demonstrated by in vitro luciferase-based assays, in vivo chromatin immunoprecipitation, and genome-wide sequencing analysis. Furthermore, the ability of YY1 to activate Nkx2.5 expression depends on its cooperative interaction with Gata4 at a nearby chromatin. Cardiac mesoderm-specific loss-of-function of YY1 resulted in early embryonic lethality. This was corroborated in vitro by embryonic stem cell-based assays in which we showed that the overexpression of YY1 enhanced the cardiogenic differentiation of embryonic stem cells into CPCs. CONCLUSIONS These results demonstrate an essential and unexpected role for YY1 to promote cardiogenesis as a transcriptional activator of Nkx2.5 and other CPC-enriched genes.
Collapse
Affiliation(s)
- Serge Gregoire
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA (
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Smith-Roe SL, Bultman SJ. Combined gene dosage requirement for SWI/SNF catalytic subunits during early mammalian development. Mamm Genome 2012; 24:21-9. [PMID: 23076393 DOI: 10.1007/s00335-012-9433-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 09/10/2012] [Indexed: 01/17/2023]
Abstract
Mammalian SWI/SNF complexes utilize either BRG1 or BRM as alternative catalytic subunits with DNA-dependent ATPase activity to remodel chromatin. Although the two proteins are 75 % identical, broadly expressed, and have similar biochemical activities in vitro, BRG1 is essential for mouse embryonic development, while BRM is dispensable. To investigate whether BRG1 and BRM have overlapping functions during mouse embryogenesis, we performed double-heterozygous intercrosses using constitutive null mutations previously created by gene targeting. The progeny of these crosses had a distribution of genotypes that was significantly skewed relative to their combined gene dosage. This was most pronounced at the top and bottom of the gene dosage hierarchy, with a 1.5-fold overrepresentation of Brg1 (+/+) ;Brm (+/+) mice and a corresponding 1.6-fold underrepresentation of Brg1 (+/-) ;Brm (-/-) mice. To account for the underrepresentation of Brg1 (+/-) ;Brm (-/-) mice, timed matings and blastocyst outgrowth assays demonstrated that ~50 % of these embryos failed to develop beyond the peri-implantation stage. These results challenge the idea that BRG1 is the exclusive catalytic subunit of SWI/SNF complexes in ES cells and suggest that BRM also interacts with the pluripotency transcription factors to facilitate self-renewal of the inner cell mass. In contrast to implantation, the Brm genotype did not influence an exencephaly phenotype that arises because of Brg1 haploinsufficiency during neural tube closure and that results in peri-natal lethality. Taken together, these results support the idea that BRG1 and BRM have overlapping functions for certain developmental processes but not others during embryogenesis.
Collapse
Affiliation(s)
- Stephanie L Smith-Roe
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599-7264, USA
| | | |
Collapse
|
27
|
van den Boogaard M, Wong LE, Tessadori F, Bakker ML, Dreizehnter LK, Wakker V, Bezzina CR, ‘t Hoen PA, Bakkers J, Barnett P, Christoffels VM. Genetic variation in T-box binding element functionally affects SCN5A/SCN10A enhancer. J Clin Invest 2012; 122:2519-30. [PMID: 22706305 PMCID: PMC3386824 DOI: 10.1172/jci62613] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 05/10/2012] [Indexed: 12/26/2022] Open
Abstract
The contraction pattern of the heart relies on the activation and conduction of the electrical impulse. Perturbations of cardiac conduction have been associated with congenital and acquired arrhythmias as well as cardiac arrest. The pattern of conduction depends on the regulation of heterogeneous gene expression by key transcription factors and transcriptional enhancers. Here, we assessed the genome-wide occupation of conduction system-regulating transcription factors TBX3, NKX2-5, and GATA4 and of enhancer-associated coactivator p300 in the mouse heart, uncovering cardiac enhancers throughout the genome. Many of the enhancers colocalized with ion channel genes repressed by TBX3, including the clustered sodium channel genes Scn5a, essential for cardiac function, and Scn10a. We identified 2 enhancers in the Scn5a/Scn10a locus, which were regulated by TBX3 and its family member and activator, TBX5, and are functionally conserved in humans. We also provided evidence that a SNP in the SCN10A enhancer associated with alterations in cardiac conduction patterns in humans disrupts TBX3/TBX5 binding and reduces the cardiac activity of the enhancer in vivo. Thus, the identification of key regulatory elements for cardiac conduction helps to explain how genetic variants in noncoding regulatory DNA sequences influence the regulation of cardiac conduction and the predisposition for cardiac arrhythmias.
Collapse
Affiliation(s)
- Malou van den Boogaard
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - L.Y. Elaine Wong
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Federico Tessadori
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Martijn L. Bakker
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisa K. Dreizehnter
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Vincent Wakker
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Connie R. Bezzina
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A.C. ‘t Hoen
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen Bakkers
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Phil Barnett
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Vincent M. Christoffels
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
28
|
Willis MS, Homeister JW, Rosson GB, Annayev Y, Holley D, Holly SP, Madden VJ, Godfrey V, Parise LV, Bultman SJ. Functional redundancy of SWI/SNF catalytic subunits in maintaining vascular endothelial cells in the adult heart. Circ Res 2012; 111:e111-22. [PMID: 22740088 DOI: 10.1161/circresaha.112.265587] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
RATIONALE Mating type switching/sucrose non-fermenting (SWI/SNF) chromatin-remodeling complexes utilize either BRG1 or BRM as a catalytic subunit to alter nucleosome position and regulate gene expression. BRG1 is required for vascular endothelial cell (VEC) development and embryonic survival, whereas BRM is dispensable. OBJECTIVE To circumvent embryonic lethality and study Brg1 function in adult tissues, we used conditional gene targeting. To evaluate possible Brg1-Brm redundancy, we analyzed Brg1 mutant mice on wild-type and Brm-deficient backgrounds. METHODS AND RESULTS The inducible Mx1-Cre driver was used to mutate Brg1 in adult mice. These conditional-null mutants exhibited a tissue-specific phenotype and unanticipated functional compensation between Brg1 and Brm. Brg1 single mutants were healthy and had a normal lifespan, whereas Brg1/Brm double mutants exhibited cardiovascular defects and died within 1 month. BRG1 and BRM were required for the viability of VECs but not other cell types where both genes were also knocked out. The VEC phenotype was most evident in the heart, particularly in the microvasculature of the outer myocardium, and was recapitulated in primary cells ex vivo. VEC death resulted in vascular leakage, cardiac hemorrhage, secondary death of cardiomyocytes due to ischemia, and ventricular dissections. CONCLUSIONS BRG1-catalyzed SWI/SNF complexes are particularly important in cardiovascular tissues. However, in contrast to embryonic development, in which Brm does not compensate, Brg1 is required in adult VECs only when Brm is also mutated. These results demonstrate for the first time that Brm functionally compensates for Brg1 in vivo and that there are significant changes in the relative importance of BRG1- and BRM-catalyzed SWI/SNF complexes during the development of an essential cell lineage.
Collapse
Affiliation(s)
- Monte S Willis
- 120 Mason Farm Rd, Genetic Medicine Bldg, Room 5060, Chapel Hill, NC 27516-7264, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Franklin S, Chen H, Mitchell-Jordan S, Ren S, Wang Y, Vondriska TM. Quantitative analysis of the chromatin proteome in disease reveals remodeling principles and identifies high mobility group protein B2 as a regulator of hypertrophic growth. Mol Cell Proteomics 2012; 11:M111.014258. [PMID: 22270000 DOI: 10.1074/mcp.m111.014258] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
A fundamental question in biology is how genome-wide changes in gene expression are enacted in response to a finite stimulus. Recent studies have mapped changes in nucleosome localization, determined the binding preferences for individual transcription factors, and shown that the genome adopts a nonrandom structure in vivo. What remains unclear is how global changes in the proteins bound to DNA alter chromatin structure and gene expression. We have addressed this question in the mouse heart, a system in which global gene expression and massive phenotypic changes occur without cardiac cell division, making the mechanisms of chromatin remodeling centrally important. To determine factors controlling genomic plasticity, we used mass spectrometry to measure chromatin-associated proteins. We have characterized the abundance of 305 chromatin-associated proteins in normal cells and measured changes in 108 proteins that accompany the progression of heart disease. These studies were conducted on a high mass accuracy instrument and confirmed in multiple biological replicates, facilitating statistical analysis and allowing us to interrogate the data bioinformatically for modules of proteins involved in similar processes. Our studies reveal general principles for global shifts in chromatin accessibility: altered linker to core histone ratio; differing abundance of chromatin structural proteins; and reprogrammed histone post-translational modifications. Using small interfering RNA-mediated loss-of-function in isolated cells, we demonstrate that the non-histone chromatin structural protein HMGB2 (but not HMGB1) suppresses pathologic cell growth in vivo and controls a gene expression program responsible for hypertrophic cell growth. Our findings reveal the basis for alterations in chromatin structure necessary for genome-wide changes in gene expression. These studies have fundamental implications for understanding how global chromatin remodeling occurs with specificity and accuracy, demonstrating that isoform-specific alterations in chromatin structural proteins can impart these features.
Collapse
Affiliation(s)
- Sarah Franklin
- Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Barnett P, van den Boogaard M, Christoffels V. Localized and temporal gene regulation in heart development. Curr Top Dev Biol 2012; 100:171-201. [PMID: 22449844 DOI: 10.1016/b978-0-12-387786-4.00004-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The heart is a structurally complex and functionally heterogeneous organ. The repertoire of genes active in a given cardiac cell defines its shapes and function. This process of localized or heterogeneous gene expression is regulated to a large extent at the level of transcription, dictating the degree particular genes in a cell are active. Therefore, errors in the regulation of localized gene expression are at the basis of misregulation of the delicate process of heart development and function. In this review, we provide an overview of the origin of the different components of the vertebrate heart, and discuss our current understanding of the regulation of localized gene expression in the developing heart. We will also discuss where future research may lead to gain more insight into this process, which should provide much needed insight into the dysregulation of heart development and function, and the etiology of congenital defects.
Collapse
Affiliation(s)
- Phil Barnett
- Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
31
|
van Weerd JH, Koshiba-Takeuchi K, Kwon C, Takeuchi JK. Epigenetic factors and cardiac development. Cardiovasc Res 2011; 91:203-11. [PMID: 21606181 DOI: 10.1093/cvr/cvr138] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Congenital heart malformations remain the leading cause of death related to birth defects. Recent advances in developmental and regenerative cardiology have shed light on a mechanistic understanding of heart development that is controlled by a transcriptional network of genetic and epigenetic factors. This article reviews the roles of chromatin remodelling factors important for cardiac development with the current knowledge of cardiac morphogenesis, regeneration, and direct cardiac differentiation. In the last 5 years, critical roles of epigenetic factors have been revealed in the cardiac research field.
Collapse
Affiliation(s)
- Jan Hendrick van Weerd
- Cardiovascular Regeneration, Institute of Molecular and Cellular Biosciences, University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
32
|
Chromatin remodelling complex dosage modulates transcription factor function in heart development. Nat Commun 2011; 2:187. [PMID: 21304516 PMCID: PMC3096875 DOI: 10.1038/ncomms1187] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 01/11/2011] [Indexed: 02/07/2023] Open
Abstract
Dominant mutations in cardiac transcription factor genes cause human inherited congenital heart defects (CHDs); however, their molecular basis is not understood. Interactions between transcription factors and the Brg1/Brm-associated factor (BAF) chromatin remodelling complex suggest potential mechanisms; however, the role of BAF complexes in cardiogenesis is not known. In this study, we show that dosage of Brg1 is critical for mouse and zebrafish cardiogenesis. Disrupting the balance between Brg1 and disease-causing cardiac transcription factors, including Tbx5, Tbx20 and Nkx2–5, causes severe cardiac anomalies, revealing an essential allelic balance between Brg1 and these cardiac transcription factor genes. This suggests that the relative levels of transcription factors and BAF complexes are important for heart development, which is supported by reduced occupancy of Brg1 at cardiac gene promoters in Tbx5 haploinsufficient hearts. Our results reveal complex dosage-sensitive interdependence between transcription factors and BAF complexes, providing a potential mechanism underlying transcription factor haploinsufficiency, with implications for multigenic inheritance of CHDs. Inherited congenital heart defects are prevalent in the human population, but the molecular mechanisms are poorly understood. In this article, deficiency in the chromatin remodelling factor, Brg1, is shown to alter cardiac development in both mouse and zebrafish laboratory models.
Collapse
|