1
|
Guijarro C, Song S, Aigouy B, Clément R, Villoutreix P, Kelly RG. Single-cell morphometrics reveals T-box gene-dependent patterns of epithelial tension in the Second Heart field. Nat Commun 2024; 15:9512. [PMID: 39496595 DOI: 10.1038/s41467-024-53612-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/17/2024] [Indexed: 11/06/2024] Open
Abstract
The vertebrate heart tube extends by progressive addition of epithelial second heart field (SHF) progenitor cells from the dorsal pericardial wall. The interplay between epithelial mechanics and genetic mechanisms during SHF deployment is unknown. Here, we present a quantitative single-cell morphometric analysis of SHF cells during heart tube extension, including force inference analysis of epithelial stress. Joint spatial Principal Component Analysis reveals that cell orientation and stress direction are the main parameters defining apical cell morphology and distinguishes cells adjacent to the arterial and venous poles. Cell shape and mechanical forces display a dynamic relationship during heart tube formation. Moreover, while the T-box transcription factor Tbx1 is necessary for cell orientation towards the arterial pole, activation of Tbx5 in the posterior SHF correlates with the establishment of epithelial stress and SHF deletion of Tbx5 relaxes the progenitor epithelium. Integrating findings from cell-scale feature patterning and mechanical stress provides new insights into cardiac morphogenesis.
Collapse
Affiliation(s)
- Clara Guijarro
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Turing Centre for Living Systems, Marseille, France
- Aix-Marseille Université, LIS, UMR 7020, Turing Centre for Living Systems, Marseille, France
- Aix-Marseille Université, MMG, Inserm U1251, Turing Centre for Living Systems, Marseille, France
| | - Solène Song
- Aix-Marseille Université, LIS, UMR 7020, Turing Centre for Living Systems, Marseille, France
- Aix-Marseille Université, MMG, Inserm U1251, Turing Centre for Living Systems, Marseille, France
| | - Benoit Aigouy
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Raphaël Clément
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Paul Villoutreix
- Aix-Marseille Université, LIS, UMR 7020, Turing Centre for Living Systems, Marseille, France.
- Aix-Marseille Université, MMG, Inserm U1251, Turing Centre for Living Systems, Marseille, France.
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Turing Centre for Living Systems, Marseille, France.
| |
Collapse
|
2
|
Kelly RG. Molecular Pathways and Animal Models of Tetralogy of Fallot and Double Outlet Right Ventricle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:645-659. [PMID: 38884739 DOI: 10.1007/978-3-031-44087-8_37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Tetralogy of Fallot and double-outlet right ventricle are outflow tract (OFT) alignment defects situated on a continuous disease spectrum. A myriad of upstream causes can impact on ventriculoarterial alignment that can be summarized as defects in either i) OFT elongation during looping morphogenesis or ii) OFT remodeling during cardiac septation. Embryological processes underlying these two developmental steps include deployment of second heart field cardiac progenitor cells, establishment and transmission of embryonic left/right information driving OFT rotation and OFT cushion and valve morphogenesis. The formation and remodeling of pulmonary trunk infundibular myocardium is a critical component of both steps. Defects in myocardial, endocardial, or neural crest cell lineages can result in alignment defects, reflecting the complex intercellular signaling events that coordinate arterial pole development. Importantly, however, OFT alignment is mechanistically distinct from neural crest-driven OFT septation, although neural crest cells impact indirectly on alignment through their role in modulating signaling during SHF development. As yet poorly understood nongenetic causes of alignment defects that impact the above processes include hemodynamic changes, maternal exposure to environmental teratogens, and stochastic events. The heterogeneity of causes converging on alignment defects characterizes the OFT as a hotspot of congenital heart defects.
Collapse
Affiliation(s)
- Robert G Kelly
- Aix Marseille Université, Institut de Biologie du Dévelopment de Marseille, Marseille, France.
| |
Collapse
|
3
|
Gill E, Bamforth SD. Molecular Pathways and Animal Models of Truncus Arteriosus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:853-865. [PMID: 38884754 DOI: 10.1007/978-3-031-44087-8_52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
In normal cardiovascular development in birds and mammals, the outflow tract of the heart is divided into two distinct channels to separate the oxygenated systemic blood flow from the deoxygenated pulmonary circulation. When the process of outflow tract septation fails, a single common outflow vessel persists resulting in a serious clinical condition known as persistent truncus arteriosus or common arterial trunk. In this chapter, we will review molecular pathways and the cells that are known to play a role in the formation and development of the outflow tract and how genetic manipulation of these pathways in animal models can result in common arterial trunk.
Collapse
Affiliation(s)
- Eleanor Gill
- Newcastle University Biosciences Institute, Newcastle, UK
| | | |
Collapse
|
4
|
Gill E, Bamforth SD. Molecular Pathways and Animal Models of Semilunar Valve and Aortic Arch Anomalies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:777-796. [PMID: 38884748 DOI: 10.1007/978-3-031-44087-8_46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The great arteries of the vertebrate carry blood from the heart to the systemic circulation and are derived from the pharyngeal arch arteries. In higher vertebrates, the pharyngeal arch arteries are a symmetrical series of blood vessels that rapidly remodel during development to become the asymmetric aortic arch arteries carrying oxygenated blood from the left ventricle via the outflow tract. At the base of the aorta, as well as the pulmonary trunk, are the semilunar valves. These valves each have three leaflets and prevent the backflow of blood into the heart. During development, the process of aortic arch and valve formation may go wrong, resulting in cardiovascular defects, and these may, at least in part, be caused by genetic mutations. In this chapter, we will review models harboring genetic mutations that result in cardiovascular defects affecting the great arteries and the semilunar valves.
Collapse
Affiliation(s)
- Eleanor Gill
- Newcastle University Biosciences Institute, Newcastle upon Tyne, UK
| | - Simon D Bamforth
- Newcastle University Biosciences Institute, Newcastle upon Tyne, UK.
| |
Collapse
|
5
|
Astrof S, Arriagada C, Saijoh Y, Francou A, Kelly RG, Moon A. Aberrant differentiation of second heart field mesoderm prefigures cellular defects in the outflow tract in response to loss of FGF8. Dev Biol 2023; 499:10-21. [PMID: 37060937 PMCID: PMC10686765 DOI: 10.1016/j.ydbio.2023.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/17/2023]
Abstract
Development of the outflow tract of the heart requires specification, proliferation and deployment of a progenitor cell population from the second heart field to generate the myocardium at the arterial pole of the heart. Disruption of these processes leads to lethal defects in rotation and septation of the outflow tract. We previously showed that Fibroblast Growth Factor 8 (FGF8) directs a signaling cascade in the second heart field that regulates critical aspects of OFT morphogenesis. Here we show that in addition to the survival and proliferation cues previously described, FGF8 provides instructive and patterning information to OFT myocardial cells and their progenitors that prevents their aberrant differentiation along a working myocardial program.
Collapse
Affiliation(s)
- Sophie Astrof
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Cecilia Arriagada
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Yukio Saijoh
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, USA
| | - Alexandre Francou
- Aix-Marseille Université, CNRS UMR 7288, Developmental Biology Institute of Marseille, Marseille, France
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, Developmental Biology Institute of Marseille, Marseille, France
| | - Anne Moon
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, PA, USA; Department of Human Genetics, University of Utah, Salt Lake City, UT, USA; The Mindich Child Health and Development Institute, Hess Center for Science and Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
6
|
De Bono C, Liu Y, Ferrena A, Valentine A, Zheng D, Morrow BE. Single-cell transcriptomics uncovers a non-autonomous Tbx1-dependent genetic program controlling cardiac neural crest cell development. Nat Commun 2023; 14:1551. [PMID: 36941249 PMCID: PMC10027855 DOI: 10.1038/s41467-023-37015-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 02/28/2023] [Indexed: 03/23/2023] Open
Abstract
Disruption of cardiac neural crest cells (CNCCs) results in congenital heart disease, yet we do not understand the cell fate dynamics as these cells differentiate to vascular smooth muscle cells. Here we performed single-cell RNA-sequencing of NCCs from the pharyngeal apparatus with the heart in control mouse embryos and when Tbx1, the gene for 22q11.2 deletion syndrome, is inactivated. We uncover three dynamic transitions of pharyngeal NCCs expressing Tbx2 and Tbx3 through differentiated CNCCs expressing cardiac transcription factors with smooth muscle genes. These transitions are altered non-autonomously by loss of Tbx1. Further, inactivation of Tbx2 and Tbx3 in early CNCCs results in aortic arch branching defects due to failed smooth muscle differentiation. Loss of Tbx1 interrupts mesoderm to CNCC cell-cell communication with upregulation and premature activation of BMP signaling and reduced MAPK signaling, as well as alteration of other signaling, and failed dynamic transitions of CNCCs leading to disruption of aortic arch artery formation and cardiac outflow tract septation.
Collapse
Affiliation(s)
- Christopher De Bono
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Yang Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alexander Ferrena
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Clinical and Translational Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aneesa Valentine
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
- Departments of Obstetrics and Gynecology; and Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
7
|
Zhao K, Yang Z. The second heart field: the first 20 years. Mamm Genome 2022:10.1007/s00335-022-09975-8. [PMID: 36550326 DOI: 10.1007/s00335-022-09975-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
In 2001, three independent groups reported the identification of a novel cluster of progenitor cells that contribute to heart development in mouse and chicken embryos. This population of progenitor cells was designated as the second heart field (SHF), and a new research direction in heart development was launched. Twenty years have since passed and a comprehensive understanding of the SHF has been achieved. This review provides retrospective insights in to the contribution, the signaling regulatory networks and the epithelial properties of the SHF. It also includes the spatiotemporal characteristics of SHF development and interactions between the SHF and other types of cells during heart development. Although considerable efforts will be required to investigate the cellular heterogeneity of the SHF, together with its intricate regulatory networks and undefined mechanisms, it is expected that the burgeoning new technology of single-cell sequencing and precise lineage tracing will advance the comprehension of SHF function and its molecular signals. The advances in SHF research will translate to clinical applications and to the treatment of congenital heart diseases, especially conotruncal defects, as well as to regenerative medicine.
Collapse
Affiliation(s)
- Ke Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China.
| |
Collapse
|
8
|
Lania G, Franzese M, Noritaka A, Bilio M, Flore G, Russo A, D'Agostino E, Angelini C, Kelly RG, Baldini A. A phenotypic rescue approach identifies lineage regionalization defects in a mouse model of DiGeorge syndrome. Dis Model Mech 2022; 15:276264. [PMID: 35946435 PMCID: PMC9555768 DOI: 10.1242/dmm.049415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 07/26/2022] [Indexed: 11/24/2022] Open
Abstract
TBX1 is a key regulator of pharyngeal apparatus (PhAp) development. Vitamin B12 (vB12) treatment partially rescues aortic arch patterning defects of Tbx1+/− embryos. Here, we show that it also improves cardiac outflow tract septation and branchiomeric muscle anomalies of Tbx1 hypomorphic mutants. At the molecular level, in vivo vB12 treatment enabled us to identify genes that were dysregulated by Tbx1 haploinsufficiency and rescued by treatment. We found that SNAI2, also known as SLUG, encoded by the rescued gene Snai2, identified a population of mesodermal cells that was partially overlapping with, but distinct from, ISL1+ and TBX1+ populations. In addition, SNAI2+ cells were mislocalized and had a greater tendency to aggregate in Tbx1+/− and Tbx1−/− embryos, and vB12 treatment restored cellular distribution. Adjacent neural crest-derived mesenchymal cells, which do not express TBX1, were also affected, showing enhanced segregation from cardiopharyngeal mesodermal cells. We propose that TBX1 regulates cell distribution in the core mesoderm and the arrangement of multiple lineages within the PhAp.
Collapse
Affiliation(s)
- Gabriella Lania
- Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy
| | - Monica Franzese
- Istituto per le Applicazione del Calcolo, National Research Council (CNR), Naples, Italy.,IRCCS SDN, Naples, Italy
| | - Adachi Noritaka
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Marchesa Bilio
- Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy
| | - Gemma Flore
- Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy
| | - Annalaura Russo
- Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy
| | - Erika D'Agostino
- Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy
| | - Claudia Angelini
- Istituto per le Applicazione del Calcolo, National Research Council (CNR), Naples, Italy
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Antonio Baldini
- Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| |
Collapse
|
9
|
Steele RE, Sanders R, Phillips HM, Bamforth SD. PAX Genes in Cardiovascular Development. Int J Mol Sci 2022; 23:7713. [PMID: 35887061 PMCID: PMC9324344 DOI: 10.3390/ijms23147713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 01/25/2023] Open
Abstract
The mammalian heart is a four-chambered organ with systemic and pulmonary circulations to deliver oxygenated blood to the body, and a tightly regulated genetic network exists to shape normal development of the heart and its associated major arteries. A key process during cardiovascular morphogenesis is the septation of the outflow tract which initially forms as a single vessel before separating into the aorta and pulmonary trunk. The outflow tract connects to the aortic arch arteries which are derived from the pharyngeal arch arteries. Congenital heart defects are a major cause of death and morbidity and are frequently associated with a failure to deliver oxygenated blood to the body. The Pax transcription factor family is characterised through their highly conserved paired box and DNA binding domains and are crucial in organogenesis, regulating the development of a wide range of cells, organs and tissues including the cardiovascular system. Studies altering the expression of these genes in murine models, notably Pax3 and Pax9, have found a range of cardiovascular patterning abnormalities such as interruption of the aortic arch and common arterial trunk. This suggests that these Pax genes play a crucial role in the regulatory networks governing cardiovascular development.
Collapse
Affiliation(s)
| | | | | | - Simon D. Bamforth
- Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Centre for Life, Newcastle NE1 3BZ, UK; (R.E.S.); (R.S.); (H.M.P.)
| |
Collapse
|
10
|
Single cell multi-omic analysis identifies a Tbx1-dependent multilineage primed population in murine cardiopharyngeal mesoderm. Nat Commun 2021; 12:6645. [PMID: 34789765 PMCID: PMC8599455 DOI: 10.1038/s41467-021-26966-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
The poles of the heart and branchiomeric muscles of the face and neck are formed from the cardiopharyngeal mesoderm within the pharyngeal apparatus. They are disrupted in patients with 22q11.2 deletion syndrome, due to haploinsufficiency of TBX1, encoding a T-box transcription factor. Here, using single cell RNA-sequencing, we now identify a multilineage primed population within the cardiopharyngeal mesoderm, marked by Tbx1, which has bipotent properties to form cardiac and branchiomeric muscle cells. The multilineage primed cells are localized within the nascent mesoderm of the caudal lateral pharyngeal apparatus and provide a continuous source of cardiopharyngeal mesoderm progenitors. Tbx1 regulates the maturation of multilineage primed progenitor cells to cardiopharyngeal mesoderm derivatives while restricting ectopic non-mesodermal gene expression. We further show that TBX1 confers this balance of gene expression by direct and indirect regulation of enriched genes in multilineage primed progenitors and downstream pathways, partly through altering chromatin accessibility, the perturbation of which can lead to congenital defects in individuals with 22q11.2 deletion syndrome.
Collapse
|
11
|
Jiang X, Li T, Liu S, Fu Q, Li F, Chen S, Sun K, Xu R, Xu Y. Variants in a cis-regulatory element of TBX1 in conotruncal heart defect patients impair GATA6-mediated transactivation. Orphanet J Rare Dis 2021; 16:334. [PMID: 34332615 PMCID: PMC8325851 DOI: 10.1186/s13023-021-01981-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 07/25/2021] [Indexed: 12/14/2022] Open
Abstract
Background TBX1 (T-box transcription factor 1) is a major candidate gene that likely contributes to the etiology of velo-cardio-facial syndrome/DiGeorge syndrome (VCFS/DGS). Although the haploinsufficiency of TBX1 in both mice and humans results in congenital cardiac malformations, little has been elucidated about its upstream regulation. We aimed to explore the transcriptional regulation and dysregulation of TBX1. Methods Different TBX1 promoter reporters were constructed. Luciferase assays and electrophoretic mobility shift assays (EMSAs) were used to identify a cis-regulatory element within the TBX1 promoter region and its trans-acting factor. The expression of proteins was identified by immunohistochemistry and immunofluorescence. Variants in the cis-regulatory element were screened in conotruncal defect (CTD) patients. In vitro functional assays were performed to show the effects of the variants found in CTD patients on the transactivation of TBX1. Results We identified a cis-regulatory element within intron 1 of TBX1 that was found to be responsive to GATA6 (GATA binding protein 6), a transcription factor crucial for cardiogenesis. The expression patterns of GATA6 and TBX1 overlapped in the pharyngeal arches of human embryos. Transfection experiments and EMSA indicated that GATA6 could activate the transcription of TBX1 by directly binding with its GATA cis-regulatory element in vitro. Furthermore, sequencing analyses of 195 sporadic CTD patients without the 22q11.2 deletion or duplication identified 3 variants (NC_000022.11:g.19756832C > G, NC_000022.11:g.19756845C > T, and NC_000022.11:g. 19756902G > T) in the non-coding cis-regulatory element of TBX1. Luciferase assays showed that all 3 variants led to reduced transcription of TBX1 when incubated with GATA6. Conclusions Our findings showed that TBX1 might be a direct transcriptional target of GATA6, and variants in the non-coding cis-regulatory element of TBX1 disrupted GATA6-mediated transactivation. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-01981-4.
Collapse
Affiliation(s)
- Xuechao Jiang
- Scientific Research Center, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Tingting Li
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Sijie Liu
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Qihua Fu
- Medical Laboratory, Shanghai Children's Medical Center, Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai, 200127, China
| | - Fen Li
- Department of Pediatric Cardiology, Shanghai Children's Medical Center, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Sun Chen
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Rang Xu
- Scientific Research Center, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Yuejuan Xu
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
12
|
A BMP4-p38 MAPK signaling axis controls ISL1 protein stability and activity during cardiogenesis. Stem Cell Reports 2021; 16:1894-1905. [PMID: 34329593 PMCID: PMC8365108 DOI: 10.1016/j.stemcr.2021.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 12/03/2022] Open
Abstract
During development, cells respond rapidly to intra- and intercellular signals, which induce signaling cascades regulating the activity of transcription factors at the transcriptional and/or post-translational level. The transcription factor ISL1 plays a key role in second heart field development and cardiac differentiation, and its mRNA levels are tightly regulated during cardiogenesis. Here, we show that a BMP-p38 MAPK signaling axis controls ISL1 protein function at the post-translational level. BMP-mediated activation of p38 MAPK leads to ISL1 phosphorylation at S269 by p38, which prevents ISL1 degradation and ensures its transcriptional activity during cardiogenesis. Interfering with p38 MAPK signaling leads to the degradation of ISL1 by the proteasome, resulting in defects in cardiomyocyte differentiation and impaired zebrafish and mouse heart morphogenesis and function. Given the critical role of the tight control of ISL1 activity during cardiac lineage diversification, modulation of BMP4-p38 MAPK signaling could direct differentiation into specific cardiac cell subpopulations. ISL1 is phosphorylated by p38 MAPK at serine 269 A BMP4-p38 MAPK signaling axis controls ISL1 protein stability Phosphorylation of ISL1 by p38 regulates its activity during cardiogenesis p38 Inhibition in vivo results in ISL1 degradation and second heart field defects
Collapse
|
13
|
Kalisch-Smith JI, Ved N, Szumska D, Munro J, Troup M, Harris SE, Rodriguez-Caro H, Jacquemot A, Miller JJ, Stuart EM, Wolna M, Hardman E, Prin F, Lana-Elola E, Aoidi R, Fisher EMC, Tybulewicz VLJ, Mohun TJ, Lakhal-Littleton S, De Val S, Giannoulatou E, Sparrow DB. Maternal iron deficiency perturbs embryonic cardiovascular development in mice. Nat Commun 2021; 12:3447. [PMID: 34103494 PMCID: PMC8187484 DOI: 10.1038/s41467-021-23660-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 05/07/2021] [Indexed: 02/05/2023] Open
Abstract
Congenital heart disease (CHD) is the most common class of human birth defects, with a prevalence of 0.9% of births. However, two-thirds of cases have an unknown cause, and many of these are thought to be caused by in utero exposure to environmental teratogens. Here we identify a potential teratogen causing CHD in mice: maternal iron deficiency (ID). We show that maternal ID in mice causes severe cardiovascular defects in the offspring. These defects likely arise from increased retinoic acid signalling in ID embryos. The defects can be prevented by iron administration in early pregnancy. It has also been proposed that teratogen exposure may potentiate the effects of genetic predisposition to CHD through gene-environment interaction. Here we show that maternal ID increases the severity of heart and craniofacial defects in a mouse model of Down syndrome. It will be important to understand if the effects of maternal ID seen here in mice may have clinical implications for women.
Collapse
Affiliation(s)
- Jacinta I Kalisch-Smith
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Nikita Ved
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Dorota Szumska
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Jacob Munro
- Victor Chang Cardiac Research Institute, Molecular, Structural and Computational Biology Division, Sydney, NSW, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Michael Troup
- Victor Chang Cardiac Research Institute, Molecular, Structural and Computational Biology Division, Sydney, NSW, Australia
| | - Shelley E Harris
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Helena Rodriguez-Caro
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Aimée Jacquemot
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
- Ealing Hospital, London, UK
| | - Jack J Miller
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
- Department of Physics, Clarendon Laboratory, University of Oxford, Oxford, UK
- Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Oxford, UK
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Eleanor M Stuart
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Magda Wolna
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Emily Hardman
- Heart Development Laboratory, The Francis Crick Institute, London, UK
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Fabrice Prin
- Heart Development Laboratory, The Francis Crick Institute, London, UK
- Advanced Light Microscopy Facility, The Francis Crick Institute, London, UK
| | - Eva Lana-Elola
- Immune Cell Biology and Down Syndrome Laboratory, The Francis Crick Institute, London, UK
| | - Rifdat Aoidi
- Immune Cell Biology and Down Syndrome Laboratory, The Francis Crick Institute, London, UK
| | | | - Victor L J Tybulewicz
- Immune Cell Biology and Down Syndrome Laboratory, The Francis Crick Institute, London, UK
- Imperial College London, London, UK
| | - Timothy J Mohun
- Heart Development Laboratory, The Francis Crick Institute, London, UK
| | - Samira Lakhal-Littleton
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Sarah De Val
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
- Ludwig Institute for Cancer Research Limited, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Eleni Giannoulatou
- Victor Chang Cardiac Research Institute, Molecular, Structural and Computational Biology Division, Sydney, NSW, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Duncan B Sparrow
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, UK.
| |
Collapse
|
14
|
Stefanovic S, Laforest B, Desvignes JP, Lescroart F, Argiro L, Maurel-Zaffran C, Salgado D, Plaindoux E, De Bono C, Pazur K, Théveniau-Ruissy M, Béroud C, Puceat M, Gavalas A, Kelly RG, Zaffran S. Hox-dependent coordination of mouse cardiac progenitor cell patterning and differentiation. eLife 2020; 9:55124. [PMID: 32804075 PMCID: PMC7462617 DOI: 10.7554/elife.55124] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 08/16/2020] [Indexed: 12/15/2022] Open
Abstract
Perturbation of addition of second heart field (SHF) cardiac progenitor cells to the poles of the heart tube results in congenital heart defects (CHD). The transcriptional programs and upstream regulatory events operating in different subpopulations of the SHF remain unclear. Here, we profile the transcriptome and chromatin accessibility of anterior and posterior SHF sub-populations at genome-wide levels and demonstrate that Hoxb1 negatively regulates differentiation in the posterior SHF. Spatial mis-expression of Hoxb1 in the anterior SHF results in hypoplastic right ventricle. Activation of Hoxb1 in embryonic stem cells arrests cardiac differentiation, whereas Hoxb1-deficient mouse embryos display premature cardiac differentiation. Moreover, ectopic differentiation in the posterior SHF of embryos lacking both Hoxb1 and its paralog Hoxa1 results in atrioventricular septal defects. Our results show that Hoxb1 plays a key role in patterning cardiac progenitor cells that contribute to both cardiac poles and provide new insights into the pathogenesis of CHD.
Collapse
Affiliation(s)
- Sonia Stefanovic
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Marseille, France
| | - Brigitte Laforest
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Marseille, France
| | | | - Fabienne Lescroart
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Marseille, France
| | - Laurent Argiro
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Marseille, France
| | | | - David Salgado
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Marseille, France
| | - Elise Plaindoux
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Marseille, France
| | | | - Kristijan Pazur
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustave Carus of TU Dresden, Helmoholtz Zentrum München, German Center for Diabetes Research (DZD), Dresden, Germany
| | - Magali Théveniau-Ruissy
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Marseille, France.,Aix Marseille Univ, CNRS UMR7288, IBDM, Marseille, France
| | - Christophe Béroud
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Marseille, France
| | - Michel Puceat
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Marseille, France
| | - Anthony Gavalas
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustave Carus of TU Dresden, Helmoholtz Zentrum München, German Center for Diabetes Research (DZD), Dresden, Germany
| | - Robert G Kelly
- Aix Marseille Univ, CNRS UMR7288, IBDM, Marseille, France
| | - Stephane Zaffran
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Marseille, France
| |
Collapse
|
15
|
Holowiecki A, Linstrum K, Ravisankar P, Chetal K, Salomonis N, Waxman JS. Pbx4 limits heart size and fosters arch artery formation by partitioning second heart field progenitors and restricting proliferation. Development 2020; 147:dev185652. [PMID: 32094112 PMCID: PMC7063670 DOI: 10.1242/dev.185652] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 02/06/2020] [Indexed: 12/11/2022]
Abstract
Vertebrate heart development requires the integration of temporally distinct differentiating progenitors. However, few signals are understood that restrict the size of the later-differentiating outflow tract (OFT). We show that improper specification and proliferation of second heart field (SHF) progenitors in zebrafish lazarus (lzr) mutants, which lack the transcription factor Pbx4, produces enlarged hearts owing to an increase in ventricular and smooth muscle cells. Specifically, Pbx4 initially promotes the partitioning of the SHF into anterior progenitors, which contribute to the OFT, and adjacent endothelial cell progenitors, which contribute to posterior pharyngeal arches. Subsequently, Pbx4 limits SHF progenitor (SHFP) proliferation. Single cell RNA sequencing of nkx2.5+ cells revealed previously unappreciated distinct differentiation states and progenitor subpopulations that normally reside within the SHF and arterial pole of the heart. Specifically, the transcriptional profiles of Pbx4-deficient nkx2.5+ SHFPs are less distinct and display characteristics of normally discrete proliferative progenitor and anterior, differentiated cardiomyocyte populations. Therefore, our data indicate that the generation of proper OFT size and arch arteries requires Pbx-dependent stratification of unique differentiation states to facilitate both homeotic-like transformations and limit progenitor production within the SHF.
Collapse
Affiliation(s)
- Andrew Holowiecki
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
| | - Kelsey Linstrum
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
- Molecular Genetics Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Padmapriyadarshini Ravisankar
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
| | - Kashish Chetal
- Bioinformatics Division, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
| | - Nathan Salomonis
- Bioinformatics Division, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Joshua S Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
16
|
Tseng YT, Ko CL, Chang CT, Lee YH, Huang Fu WC, Liu IH. Leucine-rich repeat containing 8A contributes to the expansion of brain ventricles in zebrafish embryos. Biol Open 2020; 9:bio048264. [PMID: 31941702 PMCID: PMC6994961 DOI: 10.1242/bio.048264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/23/2019] [Indexed: 12/16/2022] Open
Abstract
The sodium osmotic gradient is necessary for the initiation of brain ventricle inflation, but a previous study predicted that organic and inorganic osmolytes play equivalently important roles in osmotic homeostasis in astrocytes. To test whether organic osmoregulation also plays a role in brain ventricle inflation, the core component for volume-regulated anion and organic osmolyte channel, lrrc8a, was investigated in the zebrafish model. RT-PCR and whole-mount in situ hybridization indicated that both genes were ubiquitously expressed through to 12 hpf, and around the ventricular layer of neural tubes and the cardiogenic region at 24 hpf. Knocking down either one lrrc8a paralog with morpholino oligos resulted in abnormalities in circulation at 32 hpf. Morpholino oligos or CRISPR interference against either paralog led to smaller brain ventricles at 24 hpf. Either lrrc8aa or lrrc8ab mRNA rescued the phenotypic penetrance in both lrrc8aa and lrrc8ab morphants. Supplementation of taurine in the E3 medium and overexpression csad mRNA also rescued lrrc8aa and lrrc8ab morphants. Our results indicate that the two zebrafish lrrc8a paralogs are maternal message genes and are ubiquitously expressed in early embryos. The two genes play redundant roles in the expansion of brain ventricles and the circulatory system and taurine contributes to brain ventricle expansion via the volume-regulated anion and organic osmolyte channels.
Collapse
Affiliation(s)
- Yen-Tzu Tseng
- Department of Animal Science and Technology, National Taiwan University, Taipei, 106, Taiwan
| | - Chun-Lin Ko
- Department of Animal Science and Technology, National Taiwan University, Taipei, 106, Taiwan
| | - Chia-Teng Chang
- Department of Animal Science and Technology, National Taiwan University, Taipei, 106, Taiwan
| | - Yen-Hua Lee
- Department of Animal Science and Technology, National Taiwan University, Taipei, 106, Taiwan
| | - Wei-Chun Huang Fu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 110 Taiwan
| | - I-Hsuan Liu
- Department of Animal Science and Technology, National Taiwan University, Taipei, 106, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, 106, Taiwan
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan
| |
Collapse
|
17
|
Morrow BE, McDonald-McGinn DM, Emanuel BS, Vermeesch JR, Scambler PJ. Molecular genetics of 22q11.2 deletion syndrome. Am J Med Genet A 2019; 176:2070-2081. [PMID: 30380194 DOI: 10.1002/ajmg.a.40504] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 06/11/2018] [Accepted: 07/17/2018] [Indexed: 02/02/2023]
Abstract
The 22q11.2 deletion syndrome (22q11.2DS) is a congenital malformation and neuropsychiatric disorder caused by meiotic chromosome rearrangements. One of the goals of this review is to summarize the current state of basic research studies of 22q11.2DS. It highlights efforts to understand the mechanisms responsible for the 22q11.2 deletion that occurs in meiosis. This mechanism involves the four sets of low copy repeats (LCR22) that are dispersed in the 22q11.2 region and the deletion is mediated by nonallelic homologous recombination events. This review also highlights selected genes mapping to the 22q11.2 region that may contribute to the typical clinical findings associated with the disorder and explain that mutations in genes on the remaining allele can uncover rare recessive conditions. Another important aspect of 22q11.2DS is the existence of phenotypic heterogeneity. While some patients are mildly affected, others have severe medical, cognitive, and/or psychiatric challenges. Variability may be due in part to the presence of genetic modifiers. This review discusses current genome-wide efforts to identify such modifiers that could shed light on molecular pathways required for normal human development, cognition or behavior.
Collapse
Affiliation(s)
- Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Donna M McDonald-McGinn
- Division of Human Genetics, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Beverly S Emanuel
- Division of Human Genetics, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Joris R Vermeesch
- Center for Human Genetics, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Peter J Scambler
- Institute of Child Health, University College London, London, UK
| |
Collapse
|
18
|
Phillips HM, Stothard CA, Shaikh Qureshi WM, Kousa AI, Briones-Leon JA, Khasawneh RR, O'Loughlin C, Sanders R, Mazzotta S, Dodds R, Seidel K, Bates T, Nakatomi M, Cockell SJ, Schneider JE, Mohun TJ, Maehr R, Kist R, Peters H, Bamforth SD. Pax9 is required for cardiovascular development and interacts with Tbx1 in the pharyngeal endoderm to control 4th pharyngeal arch artery morphogenesis. Development 2019; 146:dev.177618. [PMID: 31444215 PMCID: PMC6765178 DOI: 10.1242/dev.177618] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 08/14/2019] [Indexed: 12/16/2022]
Abstract
Developmental defects affecting the heart and aortic arch arteries are a significant phenotype observed in individuals with 22q11 deletion syndrome and are caused by a microdeletion on chromosome 22q11. TBX1, one of the deleted genes, is expressed throughout the pharyngeal arches and is considered a key gene, when mutated, for the arch artery defects. Pax9 is expressed in the pharyngeal endoderm and is downregulated in Tbx1 mutant mice. We show here that Pax9-deficient mice are born with complex cardiovascular malformations that affect the outflow tract and aortic arch arteries with failure of the 3rd and 4th pharyngeal arch arteries to form correctly. Transcriptome analysis indicated that Pax9 and Tbx1 may function together, and mice double heterozygous for Tbx1/Pax9 presented with a significantly increased incidence of interrupted aortic arch when compared with Tbx1 heterozygous mice. Using a novel Pax9Cre allele, we demonstrated that the site of this Tbx1-Pax9 genetic interaction is the pharyngeal endoderm, therefore revealing that a Tbx1-Pax9-controlled signalling mechanism emanating from the pharyngeal endoderm is required for crucial tissue interactions during normal morphogenesis of the pharyngeal arch artery system. Summary: A strong genetic interaction between Tbx1 and Pax9 that leads to 4th PAA-derived defects in double heterozygous mice is cell-autonomous within the pharyngeal endoderm.
Collapse
Affiliation(s)
- Helen M Phillips
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Catherine A Stothard
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | | | | | | | - Ramada R Khasawneh
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Chloe O'Loughlin
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Rachel Sanders
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Silvia Mazzotta
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Rebecca Dodds
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Kerstin Seidel
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Timothy Bates
- School of Dental Sciences, Newcastle University, Newcastle-upon-Tyne NE2 4BW, UK
| | - Mitsushiro Nakatomi
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Simon J Cockell
- Bioinformatics Support Unit, Newcastle University, Newcastle-upon-Tyne NE2 4HH, UK
| | | | | | - René Maehr
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ralf Kist
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK.,School of Dental Sciences, Newcastle University, Newcastle-upon-Tyne NE2 4BW, UK
| | - Heiko Peters
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Simon D Bamforth
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| |
Collapse
|
19
|
Abstract
The vertebrate heart tube forms from epithelial progenitor cells in the early embryo and subsequently elongates by progressive addition of second heart field (SHF) progenitor cells from adjacent splanchnic mesoderm. Failure to maximally elongate the heart results in a spectrum of morphological defects affecting the cardiac poles, including outflow tract alignment and atrioventricular septal defects, among the most common congenital birth anomalies. SHF cells constitute an atypical apicobasally polarized epithelium with dynamic basal filopodia, located in the dorsal wall of the pericardial cavity. Recent studies have highlighted the importance of epithelial architecture and cell adhesion in the SHF, particularly for signaling events that control the progenitor cell niche during heart tube elongation. The 22q11.2 deletion syndrome gene Tbx1 regulates progenitor cell status through modulating cell shape and filopodial activity and is required for SHF contributions to both cardiac poles. Noncanonical Wnt signaling and planar cell polarity pathway genes control epithelial polarity in the dorsal pericardial wall, as progenitor cells differentiate in a transition zone at the arterial pole. Defects in these pathways lead to outflow tract shortening. Moreover, new biomechanical models of heart tube elongation have been proposed based on analysis of tissue-wide forces driving epithelial morphogenesis in the SHF, including regional cell intercalation, cell cohesion, and epithelial tension. Regulation of the epithelial properties of SHF cells is thus emerging as a key step during heart tube elongation, adding a new facet to our understanding of the mechanisms underlying both heart morphogenesis and congenital heart defects.
Collapse
Affiliation(s)
- Claudio Cortes
- From Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, France
| | - Alexandre Francou
- From Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, France
| | - Christopher De Bono
- From Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, France
| | - Robert G Kelly
- From Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, France.
| |
Collapse
|
20
|
A single-cell transcriptional roadmap for cardiopharyngeal fate diversification. Nat Cell Biol 2019; 21:674-686. [PMID: 31160712 PMCID: PMC7491489 DOI: 10.1038/s41556-019-0336-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/29/2019] [Indexed: 01/06/2023]
Abstract
In vertebrates, multipotent progenitors located in the pharyngeal mesoderm form cardiomyocytes and branchiomeric head muscles, but the dynamic gene expression programmes and mechanisms underlying cardiopharyngeal multipotency and heart versus head muscle fate choices remain elusive. Here, we used single-cell genomics in the simple chordate model Ciona to reconstruct developmental trajectories forming first and second heart lineages and pharyngeal muscle precursors and characterize the molecular underpinnings of cardiopharyngeal fate choices. We show that FGF-MAPK signalling maintains multipotency and promotes the pharyngeal muscle fate, whereas signal termination permits the deployment of a pan-cardiac programme, shared by the first and second heart lineages, to define heart identity. In the second heart lineage, a Tbx1/10-Dach pathway actively suppresses the first heart lineage programme, conditioning later cell diversity in the beating heart. Finally, cross-species comparisons between Ciona and the mouse evoke the deep evolutionary origins of cardiopharyngeal networks in chordates.
Collapse
|
21
|
De Bono C, Thellier C, Bertrand N, Sturny R, Jullian E, Cortes C, Stefanovic S, Zaffran S, Théveniau-Ruissy M, Kelly RG. T-box genes and retinoic acid signaling regulate the segregation of arterial and venous pole progenitor cells in the murine second heart field. Hum Mol Genet 2019; 27:3747-3760. [PMID: 30016433 DOI: 10.1093/hmg/ddy266] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/11/2018] [Indexed: 01/10/2023] Open
Abstract
The arterial and venous poles of the mammalian heart are hotspots of congenital heart defects (CHD) such as those observed in 22q11.2 deletion (or DiGeorge) and Holt-Oram syndromes. These regions of the heart are derived from late differentiating cardiac progenitor cells of the Second Heart Field (SHF) located in pharyngeal mesoderm contiguous with the elongating heart tube. The T-box transcription factor Tbx1, encoded by the major 22q11.2 deletion syndrome gene, regulates SHF addition to both cardiac poles from a common progenitor population. Despite the significance of this cellular addition the mechanisms regulating the deployment of common progenitor cells to alternate cardiac poles remain poorly understood. Here we demonstrate that Tbx5, mutated in Holt-Oram syndrome and essential for venous pole development, is activated in Tbx1 expressing cells in the posterior region of the SHF at early stages of heart tube elongation. A subset of the SHF transcriptional program, including Tbx1 expression, is subsequently downregulated in Tbx5 expressing cells, generating a transcriptional boundary between Tbx1-positive arterial pole and Tbx5-positive venous pole progenitor cell populations. We show that normal downregulation of the definitive arterial pole progenitor cell program in the posterior SHF is dependent on both Tbx1 and Tbx5. Furthermore, retinoic acid (RA) signaling is required for Tbx5 activation in Tbx1-positive cells and blocking RA signaling at the time of Tbx5 activation results in atrioventricular septal defects at fetal stages. Our results reveal sequential steps of cardiac progenitor cell patterning and provide mechanistic insights into the origin of common forms of CHD.
Collapse
Affiliation(s)
| | | | | | - Rachel Sturny
- Aix-Marseille Univ, CNRS UMR 7288, IBDM, Marseille, France
| | | | - Claudio Cortes
- Aix-Marseille Univ, CNRS UMR 7288, IBDM, Marseille, France
| | | | | | | | - Robert G Kelly
- Aix-Marseille Univ, CNRS UMR 7288, IBDM, Marseille, France
| |
Collapse
|
22
|
Kaplan NA, Wang W, Christiaen L. Initial characterization of Wnt-Tcf functions during Ciona heart development. Dev Biol 2019; 448:199-209. [PMID: 30635127 PMCID: PMC6487219 DOI: 10.1016/j.ydbio.2018.12.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 12/16/2022]
Abstract
In vertebrate embryos, the cardiopharyngeal mesoderm gives rise to both cardiac and branchiomeric head muscles. The canonical Wnt signaling pathway regulates many aspects of cardiomyocyte specification, and modulates a balance between skeletal and cardiac myogenesis during vertebrate head muscle development. However, the role of Wnt signaling during ascidian cardiopharyngeal development remains elusive. Here, we documented the expression of Wnt pathway components during cardiopharyngeal development in Ciona, and generated tools to investigate potential roles for Wnt signaling, and its transcriptional effector Tcf, on heart vs. pharyngeal muscle fate specification. Neither focused functional analyses nor lineage-specific transcriptome profiling uncovered a significant role for Tcf during early cardiac vs. pharyngeal muscle fate choice. By contrast, Wnt gene expression patterns of Frizzled4 and Lrp4/8 and CRISPR/Cas9-mediated Tcf knock-down suggested a later requirement for Wnt signaling during heart morphogenesis and/or cardiomyocyte differentiation. This study provides a provisional set of reagents to study Wnt signaling function in Ciona, and promising insights for future analyses of Wnt functions during heart organogenesis.
Collapse
Affiliation(s)
- Nicole A Kaplan
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, USA
| | - Wei Wang
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, USA
| | - Lionel Christiaen
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, USA.
| |
Collapse
|
23
|
Hasten E, McDonald-McGinn DM, Crowley TB, Zackai E, Emanuel BS, Morrow BE, Racedo SE. Dysregulation of TBX1 dosage in the anterior heart field results in congenital heart disease resembling the 22q11.2 duplication syndrome. Hum Mol Genet 2019; 27:1847-1857. [PMID: 29509905 DOI: 10.1093/hmg/ddy078] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/27/2018] [Indexed: 12/27/2022] Open
Abstract
Non-allelic homologous recombination events on chromosome 22q11.2 during meiosis can result in either the deletion (22q11.2DS) or duplication (22q11.2DupS) syndrome. Although the spectrum and frequency of congenital heart disease (CHD) are known for 22q11.2DS, there is less known for 22q11.2DupS. We now evaluated cardiac phenotypes in 235 subjects with 22q11.2DupS including 102 subjects we collected and 133 subjects that were previously reported as a confirmation and found 25% have CHD, mostly affecting the cardiac outflow tract (OFT). Previous studies have shown that global loss or gain of function (LOF; GOF) of mouse Tbx1, encoding a T-box transcription factor mapping to the region of synteny to 22q11.2, results in similar OFT defects. To further evaluate Tbx1 function in the progenitor cells forming the cardiac OFT, termed the anterior heart field, Tbx1 was overexpressed using the Mef2c-AHF-Cre driver (Tbx1 GOF). Here we found that all resulting conditional GOF embryos had a persistent truncus arteriosus (PTA), similar to what was previously reported for conditional Tbx1 LOF mutant embryos. To understand the basis for the PTA in the conditional GOF embryos, we found that proliferation in the Mef2c-AHF-Cre lineage cells before migrating to the heart, was reduced and critical genes were oppositely changed in this tissue in Tbx1 GOF embryos versus conditional LOF embryos. These results suggest that a major function of TBX1 in the AHF is to maintain the normal balance of expression of key cardiac developmental genes required to form the aorta and pulmonary trunk, which is disrupted in 22q11.2DS and 22q11.2DupS.
Collapse
Affiliation(s)
- Erica Hasten
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Donna M McDonald-McGinn
- Division of Human Genetics, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Terrence B Crowley
- Division of Human Genetics, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elaine Zackai
- Division of Human Genetics, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Beverly S Emanuel
- Division of Human Genetics, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Silvia E Racedo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
24
|
Hong N, Zhang E, Wang Q, Zhang X, Li F, Fu Q, Xu R, Yu Y, Chen S, Xu Y, Sun K. A loss-of-function mutation p.T52S in RIPPLY3 is a potential predisposing genetic risk factor for Chinese Han conotruncal heart defect patients without the 22q11.2 deletion/duplication. J Transl Med 2018; 16:260. [PMID: 30241482 PMCID: PMC6151064 DOI: 10.1186/s12967-018-1633-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 09/16/2018] [Indexed: 01/18/2023] Open
Abstract
Background Conotruncal heart defect (CTD) is a complex congenital heart disease with a complex and poorly understood etiology. The transcriptional corepressor RIPPLY3 plays a pivotal role in heart development as a negative regulator of the key cardiac transcription factor TBX1. A previous study showed that RIPPLY3 contribute to cardiac outflow tract development in mice, however, the relationship between RIPPLY3 and human cardiac malformation has not been reported. Methods 615 unrelated CTD Chinese Han patients were enrolled, we excluded the 22q11.2 deletion/duplication using a modified multiplex ligation-dependent probe amplification method—CNVplex®, and investigated the variants of RIPPLY3 in 577 patients without the 22q11.2 deletion/duplication by target sequencing. Functional assays were performed to testify the potential pathogenicity of nonsynonymous variants found in these CTD patients. Results Four rare heterozygous nonsynonymous variants (p.P30L, p.T52S, p.D113N and p.V179D) were identified in four CTD patients, the variant NM_018962.2:c.155C>G (p.T52S) is referred as rs745539198, and the variant NM_018962.2:c.337G>A (p.D113N) is referred as rs747419773. However, variants p.P30L and p.V179D were not found in multiple online human gene variation databases. Western blot analysis and immunofluorescence showed that there were no significant difference between wild type RIPPLY3 and these four variants. Luciferase assays revealed that the p.T52S variant altered the inhibition of TBX1 transcriptional activity in vitro, and co-immunoprecipitation assays showed that the p.T52S variant reduced the physical interaction of RIPPLY3 with TBX1. In addition to the results from pathogenicity prediction tools and evolutionary protein conservation, the p.T52S variant was thought to be a potentially deleterious variant. Conclusion Our results provide evidence that deleterious variants in RIPPLY3 are potential molecular mechanisms involved in the pathogenesis of human CTD. Electronic supplementary material The online version of this article (10.1186/s12967-018-1633-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nanchao Hong
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Room 505, Scientific Building, Shanghai, 200092, China
| | - Erge Zhang
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Room 505, Scientific Building, Shanghai, 200092, China
| | - Qingjie Wang
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Room 505, Scientific Building, Shanghai, 200092, China
| | - Xiaoqing Zhang
- Medical Laboratory, Shanghai Children's Medical Center, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Fen Li
- Department of Pediatric Cardiology, Shanghai Children's Medical Center, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Qihua Fu
- Medical Laboratory, Shanghai Children's Medical Center, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Rang Xu
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Room 505, Scientific Building, Shanghai, 200092, China.,Scientific Research Center, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yu Yu
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Room 505, Scientific Building, Shanghai, 200092, China
| | - Sun Chen
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Room 505, Scientific Building, Shanghai, 200092, China
| | - Yuejuan Xu
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Room 505, Scientific Building, Shanghai, 200092, China.
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Room 505, Scientific Building, Shanghai, 200092, China.
| |
Collapse
|
25
|
Pane LS, Fulcoli FG, Cirino A, Altomonte A, Ferrentino R, Bilio M, Baldini A. Tbx1 represses Mef2c gene expression and is correlated with histone 3 deacetylation of the anterior heart field enhancer. Dis Model Mech 2018; 11:11/9/dmm029967. [PMID: 30166330 PMCID: PMC6176997 DOI: 10.1242/dmm.029967] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/09/2018] [Indexed: 12/19/2022] Open
Abstract
The TBX1 gene is haploinsufficient in 22q11.2 deletion syndrome (22q11.2DS), and genetic evidence from human patients and mouse models points to a major role of this gene in the pathogenesis of this syndrome. Tbx1 can activate and repress transcription, and previous work has shown that one of its functions is to negatively modulate cardiomyocyte differentiation. Tbx1 occupies the anterior heart field (AHF) enhancer of the Mef2c gene, which encodes a key cardiac differentiation transcription factor. Here, we show that increased dosage of Tbx1 correlates with downregulation of Mef2c expression and reduced acetylation of its AHF enhancer in cultured mouse myoblasts. Consistently, 22q11.2DS-derived and in vitro-differentiated human induced pluripotent stem cells (hiPSCs) expressed higher levels of MEF2C and showed increased AHF acetylation, compared with hiPSCs from a healthy donor. Most importantly, we show that in mouse embryos, loss of Tbx1 enhances the expression of the Mef2c-AHF-Cre transgene in a specific region of the splanchnic mesoderm, and in a dosage-dependent manner, providing an in vivo correlate of our cell culture data. These results indicate that Tbx1 regulates the Mef2c AHF enhancer by inducing histone deacetylation.
Collapse
Affiliation(s)
- Luna Simona Pane
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Filomena Gabriella Fulcoli
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Andrea Cirino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Napoli, Italy
| | - Alessandra Altomonte
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Rosa Ferrentino
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Marchesa Bilio
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Antonio Baldini
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, 80131 Napoli, Italy .,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Napoli, Italy
| |
Collapse
|
26
|
Lin JR, Zhang Q, Cai Y, Morrow BE, Zhang ZD. Integrated rare variant-based risk gene prioritization in disease case-control sequencing studies. PLoS Genet 2017; 13:e1007142. [PMID: 29281626 PMCID: PMC5760082 DOI: 10.1371/journal.pgen.1007142] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 01/09/2018] [Accepted: 12/01/2017] [Indexed: 12/17/2022] Open
Abstract
Rare variants of major effect play an important role in human complex diseases and can be discovered by sequencing-based genome-wide association studies. Here, we introduce an integrated approach that combines the rare variant association test with gene network and phenotype information to identify risk genes implicated by rare variants for human complex diseases. Our data integration method follows a 'discovery-driven' strategy without relying on prior knowledge about the disease and thus maintains the unbiased character of genome-wide association studies. Simulations reveal that our method can outperform a widely-used rare variant association test method by 2 to 3 times. In a case study of a small disease cohort, we uncovered putative risk genes and the corresponding rare variants that may act as genetic modifiers of congenital heart disease in 22q11.2 deletion syndrome patients. These variants were missed by a conventional approach that relied on the rare variant association test alone. Case-control sequencing studies are a promising design to uncover risk genes of human complex diseases implicated by rare variants. The recent development of different types of rare variant association tests has improved the statistical power to identify disease genes that harbor risk rare variants. However, none of the recent sequencing-based genome-wide association studies identified robust disease association of rare variants or genes based on them. Due to limited sample sizes that can be feasibly achieved in real applications, current rare variant association tests can only generate marginal association signals for most risk genes. Here we proposed an integrated method that combined association signals with orthogonal biological evidence to uncover risk genes in sequencing studies. Designed to address the lack-of-power issue, our method was shown to effectively uncover risk genes with marginal association signals in data simulation. Indeed, in a real application demonstrated in our case study our method disclosed important risk genes of congenital heart disease in 22q11.2 deletion syndrome that were missed by the previous study.
Collapse
Affiliation(s)
- Jhih-Rong Lin
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Quanwei Zhang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Ying Cai
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Zhengdong D Zhang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
27
|
Shi H, O'Reilly VC, Moreau JLM, Bewes TR, Yam MX, Chapman BE, Grieve SM, Stocker R, Graham RM, Chapman G, Sparrow DB, Dunwoodie SL. Gestational stress induces the unfolded protein response, resulting in heart defects. Development 2017; 143:2561-72. [PMID: 27436040 DOI: 10.1242/dev.136820] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 05/20/2016] [Indexed: 12/20/2022]
Abstract
Congenital heart disease (CHD) is an enigma. It is the most common human birth defect and yet, even with the application of modern genetic and genomic technologies, only a minority of cases can be explained genetically. This is because environmental stressors also cause CHD. Here we propose a plausible non-genetic mechanism for induction of CHD by environmental stressors. We show that exposure of mouse embryos to short-term gestational hypoxia induces the most common types of heart defect. This is mediated by the rapid induction of the unfolded protein response (UPR), which profoundly reduces FGF signaling in cardiac progenitor cells of the second heart field. Thus, UPR activation during human pregnancy might be a common cause of CHD. Our findings have far-reaching consequences because the UPR is activated by a myriad of environmental or pathophysiological conditions. Ultimately, our discovery could lead to preventative strategies to reduce the incidence of human CHD.
Collapse
Affiliation(s)
- Hongjun Shi
- The Victor Chang Cardiac Research Institute, Sydney, New South Wales 2010, Australia St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Victoria C O'Reilly
- The Victor Chang Cardiac Research Institute, Sydney, New South Wales 2010, Australia
| | - Julie L M Moreau
- The Victor Chang Cardiac Research Institute, Sydney, New South Wales 2010, Australia
| | - Therese R Bewes
- The Victor Chang Cardiac Research Institute, Sydney, New South Wales 2010, Australia
| | - Michelle X Yam
- The Victor Chang Cardiac Research Institute, Sydney, New South Wales 2010, Australia
| | - Bogdan E Chapman
- School of Molecular Bioscience, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Stuart M Grieve
- School of Molecular Bioscience, University of Sydney, Sydney, New South Wales 2006, Australia Sydney Translational Imaging Laboratory, Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia Department of Radiology, Royal Prince Alfred Hospital, Sydney, New South Wales 2050, Australia
| | - Roland Stocker
- The Victor Chang Cardiac Research Institute, Sydney, New South Wales 2010, Australia School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Robert M Graham
- The Victor Chang Cardiac Research Institute, Sydney, New South Wales 2010, Australia St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Gavin Chapman
- The Victor Chang Cardiac Research Institute, Sydney, New South Wales 2010, Australia St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Duncan B Sparrow
- The Victor Chang Cardiac Research Institute, Sydney, New South Wales 2010, Australia St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Sally L Dunwoodie
- The Victor Chang Cardiac Research Institute, Sydney, New South Wales 2010, Australia St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
28
|
Francou A, De Bono C, Kelly RG. Epithelial tension in the second heart field promotes mouse heart tube elongation. Nat Commun 2017; 8:14770. [PMID: 28357999 PMCID: PMC5379109 DOI: 10.1038/ncomms14770] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 01/27/2017] [Indexed: 12/29/2022] Open
Abstract
Extension of the vertebrate heart tube is driven by progressive addition of second heart field (SHF) progenitor cells to the poles of the heart. Defects in this process cause a spectrum of congenital anomalies. SHF cells form an epithelial layer in splanchnic mesoderm in the dorsal wall of the pericardial cavity. Here we report oriented cell elongation, polarized actomyosin distribution and nuclear YAP/TAZ in a proliferative centre in the posterior dorsal pericardial wall during heart tube extension. These parameters are indicative of mechanical stress, further supported by analysis of cell shape changes in wound assays. Time course and mutant analysis identifies SHF deployment as a source of epithelial tension. Moreover, cell division and oriented growth in the dorsal pericardial wall align with the axis of cell elongation, suggesting that epithelial tension in turn contributes to heart tube extension. Our results implicate tissue-level forces in the regulation of heart tube extension. Epithelial progenitor cell growth in the second heart field contributes to heart morphogenesis but how this is regulated at the tissue level is unclear. Here, the authors show that cell elongation, polarized actomyosin and nuclear YAP/TAZ drive epithelial growth and correlate with mechanical tension.
Collapse
Affiliation(s)
- Alexandre Francou
- Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, Campus De Luminy Case 907, 13288 Marseille Cedex 9, France
| | - Christopher De Bono
- Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, Campus De Luminy Case 907, 13288 Marseille Cedex 9, France
| | - Robert G Kelly
- Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, Campus De Luminy Case 907, 13288 Marseille Cedex 9, France
| |
Collapse
|
29
|
Reduced dosage of β-catenin provides significant rescue of cardiac outflow tract anomalies in a Tbx1 conditional null mouse model of 22q11.2 deletion syndrome. PLoS Genet 2017; 13:e1006687. [PMID: 28346476 PMCID: PMC5386301 DOI: 10.1371/journal.pgen.1006687] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/10/2017] [Accepted: 03/13/2017] [Indexed: 11/19/2022] Open
Abstract
The 22q11.2 deletion syndrome (22q11.2DS; velo-cardio-facial syndrome; DiGeorge syndrome) is a congenital anomaly disorder in which haploinsufficiency of TBX1, encoding a T-box transcription factor, is the major candidate for cardiac outflow tract (OFT) malformations. Inactivation of Tbx1 in the anterior heart field (AHF) mesoderm in the mouse results in premature expression of pro-differentiation genes and a persistent truncus arteriosus (PTA) in which septation does not form between the aorta and pulmonary trunk. Canonical Wnt/β-catenin has major roles in cardiac OFT development that may act upstream of Tbx1. Consistent with an antagonistic relationship, we found the opposite gene expression changes occurred in the AHF in β-catenin loss of function embryos compared to Tbx1 loss of function embryos, providing an opportunity to test for genetic rescue. When both alleles of Tbx1 and one allele of β-catenin were inactivated in the Mef2c-AHF-Cre domain, 61% of them (n = 34) showed partial or complete rescue of the PTA defect. Upregulated genes that were oppositely changed in expression in individual mutant embryos were normalized in significantly rescued embryos. Further, β-catenin was increased in expression when Tbx1 was inactivated, suggesting that there may be a negative feedback loop between canonical Wnt and Tbx1 in the AHF to allow the formation of the OFT. We suggest that alteration of this balance may contribute to variable expressivity in 22q11.2DS.
Collapse
|
30
|
A 3 base pair deletion in TBX1 leads to reduced protein expression and transcriptional activity. Sci Rep 2017; 7:44165. [PMID: 28272434 PMCID: PMC5341160 DOI: 10.1038/srep44165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 01/23/2017] [Indexed: 02/06/2023] Open
Abstract
Transcription factor TBX1 plays a pivotal role in heart development and has been implicated in 22q11.2 deletion syndrome. The structure of this protein has been elucidated, and several mutations have been identified that disrupt TBX1 localization, DNA/protein-binding, or mRNA expression. This study reports a mutation in the TBX1 gene that leads to significantly reduced expression of the mutant protein. A total of 773 conotruncal heart defect patients and 516 unrelated healthy control individuals were enrolled, none of which harbored a 22q11.2 deletion or duplication. We identified a mutation, c.303-305delGAA, located in the third exon of TBX1 that does not disrupt TBX1 mRNA expression or DNA binding activity, but results in decreased TBX1 protein levels and transcriptional activity. Through protein degradation studies we demonstrated that TBX1 is degraded primarily in proteasomes. Although the c.303-305delGAA mutation leads to low levels of the mutant protein, we found that increased protein degradation was not the cause, and we hypothesize that an alternate mechanism, such as translational inhibition, may be the cause.
Collapse
|
31
|
Abstract
Recent data have paved the way to mechanistic studies into the role of Tbx1 during development. Tbx1 is haploinsufficient and is involved in an important genetic disorder. The gene encodes a T-box transcription factor that is expressed from approximately E7.5 in mouse embryos and continues to be expressed in a highly dynamic manner. It is neither a strong transcriptional activator nor a strong repressor, but it regulates a large number of genes through epigenetic modifications. Here, we review recent literature concerning mechanisms of gene regulation by Tbx1 and its role in mammalian development, with a special focus on the cardiac, vascular, and central nervous systems.
Collapse
|
32
|
Abstract
Cardiac cell specification and the genetic determinants that govern this process are highly conserved among Chordates. Recent studies have established the importance of evolutionarily-conserved mechanisms in the study of congenital heart defects and disease, as well as cardiac regeneration. As a basal Chordate, the Ciona model system presents a simple scaffold that recapitulates the basic blueprint of cardiac development in Chordates. Here we will focus on the development and cellular structure of the heart of the ascidian Ciona as compared to other Chordates, principally vertebrates. Comparison of the Ciona model system to heart development in other Chordates presents great potential for dissecting the genetic mechanisms that underlie congenital heart defects and disease at the cellular level and might provide additional insight into potential pathways for therapeutic cardiac regeneration.
Collapse
|
33
|
Baardman ME, Zwier MV, Wisse LJ, Gittenberger-de Groot AC, Kerstjens-Frederikse WS, Hofstra RMW, Jurdzinski A, Hierck BP, Jongbloed MRM, Berger RMF, Plösch T, DeRuiter MC. Common arterial trunk and ventricular non-compaction in Lrp2 knockout mice indicate a crucial role of LRP2 in cardiac development. Dis Model Mech 2016; 9:413-25. [PMID: 26822476 PMCID: PMC4852499 DOI: 10.1242/dmm.022053] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 01/20/2016] [Indexed: 01/22/2023] Open
Abstract
Lipoprotein-related receptor protein 2 (LRP2) is important for development of the embryonic neural crest and brain in both mice and humans. Although a role in cardiovascular development can be expected, the hearts of Lrp2 knockout (KO) mice have not yet been investigated. We studied the cardiovascular development of Lrp2 KO mice between embryonic day 10.5 (E10.5) and E15.5, applying morphometry and immunohistochemistry, using antibodies against Tfap2α (neural crest cells), Nkx2.5 (second heart field), WT1 (epicardium derived cells), tropomyosin (myocardium) and LRP2. The Lrp2 KO mice display a range of severe cardiovascular abnormalities, including aortic arch anomalies, common arterial trunk (persistent truncus arteriosus) with coronary artery anomalies, ventricular septal defects, overriding of the tricuspid valve and marked thinning of the ventricular myocardium. Both the neural crest cells and second heart field, which are essential for the lengthening and growth of the right ventricular outflow tract, are abnormally positioned in the Lrp2 KO. This explains the absence of the aorto-pulmonary septum, which leads to common arterial trunk and ventricular septal defects. Severe blebbing of the epicardial cells covering the ventricles is seen. Epithelial-mesenchymal transition does occur; however, there are fewer WT1-positive epicardium-derived cells in the ventricular wall as compared to normal, coinciding with the myocardial thinning and deep intertrabecular spaces. LRP2 plays a crucial role in cardiovascular development in mice. This corroborates findings of cardiac anomalies in humans with LRP2 mutations. Future studies should reveal the underlying signaling mechanisms in which LRP2 is involved during cardiogenesis. Summary: This paper sheds a new light on the role of the second heart field and neural crest cells in outflow tract formation in the mouse embryo. Depletion of the LPR2 results in a disturbed contribution pattern and subsequent common arterial trunk.
Collapse
Affiliation(s)
- Maria E Baardman
- Department of Genetics, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| | - Mathijs V Zwier
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| | - Lambertus J Wisse
- Department of Anatomy and Embryology, Leiden University Medical Center, PO-Box 9600, Leiden 2300 RC, The Netherlands
| | | | - Wilhelmina S Kerstjens-Frederikse
- Department of Genetics, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| | - Robert M W Hofstra
- Department of Clinical Genetics, Erasmus Medical Center Rotterdam, PO-Box 2040, Rotterdam 3000 CA, The Netherlands Neural Development and Gastroenterology Units, UCL Institute of Child Health, London WC1 NEH, UK
| | - Angelika Jurdzinski
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| | - Beerend P Hierck
- Department of Anatomy and Embryology, Leiden University Medical Center, PO-Box 9600, Leiden 2300 RC, The Netherlands
| | - Monique R M Jongbloed
- Department of Cardiology and Department of Anatomy and Embryology, Leiden University Medical Center, PO-Box 9600, Leiden 2300 RC, The Netherlands
| | - Rolf M F Berger
- Center for Congenital Heart Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| | - Torsten Plösch
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| | - Marco C DeRuiter
- Department of Anatomy and Embryology, Leiden University Medical Center, PO-Box 9600, Leiden 2300 RC, The Netherlands
| |
Collapse
|
34
|
Théveniau-Ruissy M, Pérez-Pomares JM, Parisot P, Baldini A, Miquerol L, Kelly RG. Coronary stem development in wild-type and Tbx1 null mouse hearts. Dev Dyn 2016; 245:445-59. [PMID: 26708418 DOI: 10.1002/dvdy.24380] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/17/2015] [Accepted: 12/18/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Coronary artery (CA) stems connect the ventricular coronary tree with the aorta. Defects in proximal CA patterning are a cause of sudden cardiac death. In mice lacking Tbx1, common arterial trunk is associated with an abnormal trajectory of the proximal left CA. Here we investigate CA stem development in wild-type and Tbx1 null embryos. RESULTS Genetic lineage tracing reveals that limited outgrowth of aortic endothelium contributes to proximal CA stems. Immunohistochemistry and fluorescent tracer injections identify a periarterial vascular plexus present at the onset of CA stem development. Transplantation experiments in avian embryos indicate that the periarterial plexus originates in mesenchyme distal to the outflow tract. Tbx1 is required for the patterning but not timing of CA stem development and a Tbx1 reporter allele is expressed in myocardium adjacent to the left but not right CA stem. This expression domain is maintained in Sema3c(-/-) hearts with a common arterial trunk and leftward positioned CA. Ectopic myocardial differentiation is observed on the left side of the Tbx1(-/-) common arterial trunk. CONCLUSIONS A periarterial plexus bridges limited outgrowth of the aortic endothelium with the ventricular plexus during CA stem development. Molecular differences associated with left and right CA stems provide new insights into the etiology of CA patterning defects.
Collapse
Affiliation(s)
| | - José-Maria Pérez-Pomares
- Department of Animal Biology, Instituto de Investigación Biomedica de Málaga (IBIMA), Faculty of Science, University of Málaga, Málaga, Spain.,Andalusian Centre for Nanomedicine and Biotechnology (BIONAND), Málaga, Spain
| | - Pauline Parisot
- Aix-Marseille University, CNRS, IBDM UMR 7288, Marseille, France
| | | | - Lucile Miquerol
- Aix-Marseille University, CNRS, IBDM UMR 7288, Marseille, France
| | - Robert G Kelly
- Aix-Marseille University, CNRS, IBDM UMR 7288, Marseille, France
| |
Collapse
|
35
|
Dorn T, Goedel A, Lam JT, Haas J, Tian Q, Herrmann F, Bundschu K, Dobreva G, Schiemann M, Dirschinger R, Guo Y, Kühl SJ, Sinnecker D, Lipp P, Laugwitz KL, Kühl M, Moretti A. Direct nkx2-5 transcriptional repression of isl1 controls cardiomyocyte subtype identity. Stem Cells 2016; 33:1113-29. [PMID: 25524439 PMCID: PMC6750130 DOI: 10.1002/stem.1923] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 10/29/2014] [Accepted: 11/08/2014] [Indexed: 12/31/2022]
Abstract
During cardiogenesis, most myocytes arise from cardiac progenitors expressing the transcription factors Isl1 and Nkx2-5. Here, we show that a direct repression of Isl1 by Nkx2-5 is necessary for proper development of the ventricular myocardial lineage. Overexpression of Nkx2-5 in mouse embryonic stem cells (ESCs) delayed specification of cardiac progenitors and inhibited expression of Isl1 and its downstream targets in Isl1(+) precursors. Embryos deficient for Nkx2-5 in the Isl1(+) lineage failed to downregulate Isl1 protein in cardiomyocytes of the heart tube. We demonstrated that Nkx2-5 directly binds to an Isl1 enhancer and represses Isl1 transcriptional activity. Furthermore, we showed that overexpression of Isl1 does not prevent cardiac differentiation of ESCs and in Xenopus laevis embryos. Instead, it leads to enhanced specification of cardiac progenitors, earlier cardiac differentiation, and increased cardiomyocyte number. Functional and molecular characterization of Isl1-overexpressing cardiomyocytes revealed higher beating frequencies in both ESC-derived contracting areas and Xenopus Isl1-gain-of-function hearts, which associated with upregulation of nodal-specific genes and downregulation of transcripts of working myocardium. Immunocytochemistry of cardiomyocyte lineage-specific markers demonstrated a reduction of ventricular cells and an increase of cells expressing the pacemaker channel Hcn4. Finally, optical action potential imaging of single cardiomyocytes combined with pharmacological approaches proved that Isl1 overexpression in ESCs resulted in normally electrophysiologically functional cells, highly enriched in the nodal subtype at the expense of the ventricular lineage. Our findings provide an Isl1/Nkx2-5-mediated mechanism that coordinately regulates the specification of cardiac progenitors toward the different myocardial lineages and ensures proper acquisition of myocyte subtype identity.
Collapse
Affiliation(s)
- Tatjana Dorn
- I. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
TBX1 Represses Vegfr2 Gene Expression and Enhances the Cardiac Fate of VEGFR2+ Cells. PLoS One 2015; 10:e0138525. [PMID: 26382615 PMCID: PMC4575176 DOI: 10.1371/journal.pone.0138525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/01/2015] [Indexed: 02/04/2023] Open
Abstract
The T-box transcription factor TBX1 has critical roles in maintaining proliferation and inhibiting differentiation of cardiac progenitor cells of the second heart field (SHF). Haploinsufficiency of the gene that encodes it is a cause of congenital heart disease. Here, we developed an embryonic stem (ES) cell-based model in which Tbx1 expression can be modulated by tetracycline. Using this model, we found that TBX1 down regulates the expression of VEGFR2, and we confirmed this finding in vivo during embryonic development. In addition, we found a Vegfr2 domain of expression, not previously described, in the posterior SHF and this expression is extended by loss of Tbx1. VEGFR2 has been previously described as a marker of a subpopulation of cardiac progenitors. Clonal analysis of ES-derived VEGFR2+ cells indicated that 12.5% of clones expressed three markers of cardiac lineage (cardiomyocyte, smooth muscle and endothelium). However, a pulse of Tbx1 expression was sufficient to increase the percentage to 20.8%. In addition, the percentage of clones expressing markers of multiple cardiac lineages increased from 41.6% to 79.1% after Tbx1 pulse. These results suggest that TBX1 plays a role in maintaining a progenitor state in VEGFR2+ cells.
Collapse
|
37
|
Yang S, Wang S, Sun F, Zhang M, Wu F, Xu F, Ding Z. Protective effects of puerarin against tetrabromobisphenol a-induced apoptosis and cardiac developmental toxicity in zebrafish embryo-larvae. ENVIRONMENTAL TOXICOLOGY 2015; 30:1014-1023. [PMID: 24596333 DOI: 10.1002/tox.21975] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 02/10/2014] [Accepted: 02/23/2014] [Indexed: 06/03/2023]
Abstract
Tetrabromobisphenol A (TBBPA), a brominated flame retardant, is detected commonly in aquatic environments, where it is thought to be highly toxic to the development of aquatic life. In this study, zebrafish embryos and larvae were used to investigate the protective effects of puerarin after exposure to TBBPA. Malformation, blood flow disorders, pericardial edema, and spawn coagulation rates increased, whereas survival decreased significantly after exposure to 0.5 and 1.0 mg L(-1) TBBPA. The measured indices of morphological toxicity improved after treatment with puerarin. TBBPA also induced reactive oxygen species (ROS) production in a dose-dependent manner. Acridine orange staining results revealed that TBBPA exposure caused cardiomyocyte apoptosis and induced the expression of three proapoptotic genes: P53, Bax, and Caspase9. In contrast, the expression of the antiapoptotic gene Bcl2 was down-regulated. When genes related to cardiac development were assessed, the expression of Tbx1, Raldh2, and Bmp2b changed after exposure to the combination of TBBPA and puerarin. These results suggest that TBBPA induces cardiomyocyte apoptosis and ROS production, resulting in cardiac developmental toxicity in zebrafish embryos or larvae. Therefore, puerarin regulates the expression of cardiac developmental genes, such as Tbx1, Bmp2b, and Raldh2 by inhibiting ROS production, and subsequently modulates cardiac development after the exposure of zebrafish larvae to TBBPA.
Collapse
Affiliation(s)
- Suwen Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Shengrui Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Fengchao Sun
- Department of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mengmeng Zhang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Fengchang Wu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Fanfan Xu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Zhishan Ding
- Department of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| |
Collapse
|
38
|
Zhang R, Cao P, Yang Z, Wang Z, Wu JL, Chen Y, Pan Y. Heparan Sulfate Biosynthesis Enzyme, Ext1, Contributes to Outflow Tract Development of Mouse Heart via Modulation of FGF Signaling. PLoS One 2015; 10:e0136518. [PMID: 26295701 PMCID: PMC4546591 DOI: 10.1371/journal.pone.0136518] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 08/05/2015] [Indexed: 11/19/2022] Open
Abstract
Glycosaminoglycans are important regulators of multiple signaling pathways. As a major constituent of the heart extracellular matrix, glycosaminoglycans are implicated in cardiac morphogenesis through interactions with different signaling morphogens. Ext1 is a glycosyltransferase responsible for heparan sulfate synthesis. Here, we evaluate the function of Ext1 in heart development by analyzing Ext1 hypomorphic mutant and conditional knockout mice. Outflow tract alignment is sensitive to the dosage of Ext1. Deletion of Ext1 in the mesoderm induces a cardiac phenotype similar to that of a mutant with conditional deletion of UDP-glucose dehydrogenase, a key enzyme responsible for synthesis of all glycosaminoglycans. The outflow tract defect in conditional Ext1 knockout(Ext1f/f:Mesp1Cre) mice is attributable to the reduced contribution of second heart field and neural crest cells. Ext1 deletion leads to downregulation of FGF signaling in the pharyngeal mesoderm. Exogenous FGF8 ameliorates the defects in the outflow tract and pharyngeal explants. In addition, Ext1 expression in second heart field and neural crest cells is required for outflow tract remodeling. Our results collectively indicate that Ext1 is crucial for outflow tract formation in distinct progenitor cells, and heparan sulfate modulates FGF signaling during early heart development.
Collapse
Affiliation(s)
- Rui Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Peijuan Cao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Zhongzhou Yang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Zhenzhen Wang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Jiu-Lin Wu
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou, China
| | - Yan Chen
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
- * E-mail: (YC); (YP)
| | - Yi Pan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
- * E-mail: (YC); (YP)
| |
Collapse
|
39
|
Chung JH, Cai J, Suskin BG, Zhang Z, Coleman K, Morrow BE. Whole-Genome Sequencing and Integrative Genomic Analysis Approach on Two 22q11.2 Deletion Syndrome Family Trios for Genotype to Phenotype Correlations. Hum Mutat 2015; 36:797-807. [PMID: 25981510 DOI: 10.1002/humu.22814] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 05/01/2015] [Indexed: 12/20/2022]
Abstract
The 22q11.2 deletion syndrome (22q11DS) affects 1:4,000 live births and presents with highly variable phenotype expressivity. In this study, we developed an analytical approach utilizing whole-genome sequencing (WGS) and integrative analysis to discover genetic modifiers. Our pipeline combined available tools in order to prioritize rare, predicted deleterious, coding and noncoding single-nucleotide variants (SNVs), and insertion/deletions from WGS. We sequenced two unrelated probands with 22q11DS, with contrasting clinical findings, and their unaffected parents. Proband P1 had cognitive impairment, psychotic episodes, anxiety, and tetralogy of Fallot (TOF), whereas proband P2 had juvenile rheumatoid arthritis but no other major clinical findings. In P1, we identified common variants in COMT and PRODH on 22q11.2 as well as rare potentially deleterious DNA variants in other behavioral/neurocognitive genes. We also identified a de novo SNV in ADNP2 (NM_014913.3:c.2243G>C), encoding a neuroprotective protein that may be involved in behavioral disorders. In P2, we identified a novel nonsynonymous SNV in ZFPM2 (NM_012082.3:c.1576C>T), a known causative gene for TOF, which may act as a protective variant downstream of TBX1, haploinsufficiency of which is responsible for congenital heart disease in individuals with 22q11DS.
Collapse
Affiliation(s)
- Jonathan H Chung
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Jinlu Cai
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Barrie G Suskin
- Department of Obstetrics & Gynecology and Women's Health, Montefiore Medical Center, Bronx, New York
| | - Zhengdong Zhang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Karlene Coleman
- Children's Healthcare of Atlanta at Egleston, Atlanta, Georgia
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
40
|
Vanyai HK, Thomas T, Voss AK. Mesodermal expression of Moz is necessary for cardiac septum development. Dev Biol 2015; 403:22-9. [DOI: 10.1016/j.ydbio.2015.04.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 04/09/2015] [Accepted: 04/10/2015] [Indexed: 10/23/2022]
|
41
|
Zhou L, Liu J, Olson P, Zhang K, Wynne J, Xie L. Tbx5 and Osr1 interact to regulate posterior second heart field cell cycle progression for cardiac septation. J Mol Cell Cardiol 2015; 85:1-12. [PMID: 25986147 DOI: 10.1016/j.yjmcc.2015.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 05/05/2015] [Accepted: 05/07/2015] [Indexed: 11/30/2022]
Abstract
RATIONALE Mutations of TBX5 cause Holt-Oram syndrome (HOS) in humans, a disease characterized by atrial or occasionally ventricular septal defects in the heart and skeletal abnormalities of the upper extremity. Previous studies have demonstrated that Tbx5 regulates Osr1 expression in the second heart field (SHF) of E9.5 mouse embryos. However, it is unknown whether and how Tbx5 and Osr1 interact in atrial septation. OBJECTIVE To determine if and how Tbx5 and Osr1 interact in the posterior SHF for cardiac septation. METHODS AND RESULTS In the present study, genetic inducible fate mapping showed that Osr1-expressing cells contribute to atrial septum progenitors between E8.0 and E11.0. Osr1 expression in the pSHF was dependent on the level of Tbx5 at E8.5 and E9.5 but not E10.5, suggesting that the embryo stage before E10.5 is critical for Tbx5 interacting with Osr1 in atrial septation. Significantly more atrioventricular septal defects (AVSDs) were observed in embryos with compound haploinsufficiency for Tbx5 and Osr1. Conditional compound haploinsufficiency for Tbx5 and Osr1 resulted in a significant cell proliferation defect in the SHF, which was associated with fewer cells in the G2 and M phases and a decreased level of Cdk6 expression. Remarkably, genetically targeted disruption of Pten expression in atrial septum progenitors rescued AVSDs caused by Tbx5 and Osr1 compound haploinsufficiency. There was a significant decrease in Smo expression, which is a Hedgehog (Hh) signaling pathway modulator, in the pSHF of Osr1 knockout embryos at E9.5, implying a role for Osr1 in regulating Hh signaling. CONCLUSIONS Tbx5 and Osr1 interact to regulate posterior SHF cell cycle progression for cardiac septation.
Collapse
Affiliation(s)
- Lun Zhou
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; Department of Gerontology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jielin Liu
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Patrick Olson
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Ke Zhang
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Joshua Wynne
- Department of Internal Medicine, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Linglin Xie
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA.
| |
Collapse
|
42
|
A new heart for a new head in vertebrate cardiopharyngeal evolution. Nature 2015; 520:466-73. [PMID: 25903628 DOI: 10.1038/nature14435] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 11/25/2014] [Indexed: 12/22/2022]
Abstract
It has been more than 30 years since the publication of the new head hypothesis, which proposed that the vertebrate head is an evolutionary novelty resulting from the emergence of neural crest and cranial placodes. Neural crest generates the skull and associated connective tissues, whereas placodes produce sensory organs. However, neither crest nor placodes produce head muscles, which are a crucial component of the complex vertebrate head. We discuss emerging evidence for a surprising link between the evolution of head muscles and chambered hearts - both systems arise from a common pool of mesoderm progenitor cells within the cardiopharyngeal field of vertebrate embryos. We consider the origin of this field in non-vertebrate chordates and its evolution in vertebrates.
Collapse
|
43
|
Bosman A, Letourneau A, Sartiani L, Del Lungo M, Ronzoni F, Kuziakiv R, Tohonen V, Zucchelli M, Santoni F, Guipponi M, Dumevska B, Hovatta O, Antonarakis SE, Jaconi ME. Perturbations of Heart Development and Function in Cardiomyocytes from Human Embryonic Stem Cells with Trisomy 21. Stem Cells 2015; 33:1434-46. [DOI: 10.1002/stem.1961] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 12/19/2014] [Indexed: 12/31/2022]
Affiliation(s)
- Alexis Bosman
- Department of Pathology and Immunology; Faculty of Medicine; University of Geneva; Geneva Switzerland
- Victor Chang Cardiac Research Institute; Darlinghurst New South Wales Australia
| | - Audrey Letourneau
- Department of Genetic Medicine and Development; Faculty of Medicine, University of Geneva; Geneva Switzerland
| | - Laura Sartiani
- Department of Neuroscience; Psychology, Drug Research and Child Health, Center of Molecular Medicine, University of Florence; Florence Italy
| | - Martina Del Lungo
- Department of Neuroscience; Psychology, Drug Research and Child Health, Center of Molecular Medicine, University of Florence; Florence Italy
| | - Flavio Ronzoni
- Department of Pathology and Immunology; Faculty of Medicine; University of Geneva; Geneva Switzerland
| | - Rostyslav Kuziakiv
- Department of Pathology and Immunology; Faculty of Medicine; University of Geneva; Geneva Switzerland
| | - Virpi Tohonen
- Department of Biosciences and Nutrition; Karolinska Institute; Huddinge Sweden
| | - Marco Zucchelli
- Department of Biosciences and Nutrition; Karolinska Institute; Huddinge Sweden
| | - Federico Santoni
- Department of Genetic Medicine and Development; Faculty of Medicine, University of Geneva; Geneva Switzerland
| | - Michel Guipponi
- Department of Genetic Medicine and Development; Faculty of Medicine, University of Geneva; Geneva Switzerland
| | | | - Outi Hovatta
- Division of Obstetrics and Gynecology; Department of Clinical Science; Karolinska Institute; Huddinge Stockholm Sweden
| | - Stylianos E. Antonarakis
- Department of Genetic Medicine and Development; Faculty of Medicine, University of Geneva; Geneva Switzerland
- iGE3 Institute of Genetics and Genomics of Geneva; Geneva Switzerland
| | - Marisa E. Jaconi
- Department of Pathology and Immunology; Faculty of Medicine; University of Geneva; Geneva Switzerland
| |
Collapse
|
44
|
Regulation and evolution of cardiopharyngeal cell identity and behavior: insights from simple chordates. Curr Opin Genet Dev 2015; 32:119-28. [PMID: 25819888 DOI: 10.1016/j.gde.2015.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 02/16/2015] [Accepted: 02/23/2015] [Indexed: 01/08/2023]
Abstract
The vertebrate heart arises from distinct first and second heart fields. The latter also share a common origin with branchiomeric muscles in the pharyngeal mesoderm and transcription regulators, such as Nkx2-5, Tbx1 and Islet1. Despite significant progress, the complexity of vertebrate embryos has hindered the identification of multipotent cardiopharyngeal progenitors. Here, we summarize recent insights in cardiopharyngeal development gained from ascidian models, among the closest relatives to vertebrates. In a simplified cellular context, progressive fate specification of the ascidian cardiopharyngeal precursors presents striking similarities with their vertebrate counterparts. Multipotent cardiopharyngeal progenitors are primed to activate both the early cardiac and pharyngeal muscles programs, which segregate following asymmetric cells divisions as a result of regulatory cross-antagonisms involving Tbx1 and Nkx2-5 homologs. Activation of Ebf in pharyngeal muscle founder cells triggers both Myogenic Regulatory Factor-associated differentiation and Notch-mediated maintenance of an undifferentiated state in distinct precursors. Cross-species comparisons revealed the deep conservation of the cardiopharyngeal developmental sequence in spite of extreme genome sequence divergence, gene network rewiring and specific morphogenetic differences. Finally, analyses are beginning to uncover the influence of surrounding tissues in determining cardiopharyngeal cell identity and behavior. Thus, ascidian embryos offer a unique opportunity to study gene regulation and cell behaviors at the cellular level throughout cardiopharyngeal morphogenesis and evolution.
Collapse
|
45
|
Francou A, Saint-Michel E, Mesbah K, Kelly RG. TBX1 regulates epithelial polarity and dynamic basal filopodia in the second heart field. Development 2015; 141:4320-31. [PMID: 25371366 DOI: 10.1242/dev.115022] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Elongation of the vertebrate heart occurs by progressive addition of second heart field (SHF) cardiac progenitor cells from pharyngeal mesoderm to the poles of the heart tube. The importance of these cells in the etiology of congenital heart defects has led to extensive research into the regulation of SHF deployment by signaling pathways and transcription factors. However, the basic cellular features of these progenitor cells, including epithelial polarity, cell shape and cell dynamics, remain poorly characterized. Here, using immunofluorescence, live imaging and embryo culture, we demonstrate that SHF cells constitute an atypical, apicobasally polarized epithelium in the dorsal pericardial wall, characterized by apical monocilia and dynamic actin-rich basal filopodia. We identify the 22q11.2 deletion syndrome gene Tbx1, required in the SHF for outflow tract development, as a regulator of the epithelial properties of SHF cells. Cell shape changes in mutant embryos include increased circularity, a reduced basolateral membrane domain and impaired filopodial activity, and are associated with elevated aPKCζ levels. Activation of aPKCζ in embryo culture similarly impairs filopodia activity and phenocopies proliferative defects and ectopic differentiation observed in the SHF of Tbx1 null embryos. Our results reveal that epithelial and progenitor cell status are coupled in the SHF, identifying control of cell shape as a regulatory step in heart tube elongation and outflow tract morphogenesis.
Collapse
Affiliation(s)
- Alexandre Francou
- Aix Marseille Université, CNRS, IBDM UMR 7288, Marseille 13288, France
| | | | - Karim Mesbah
- Aix Marseille Université, CNRS, IBDM UMR 7288, Marseille 13288, France
| | - Robert G Kelly
- Aix Marseille Université, CNRS, IBDM UMR 7288, Marseille 13288, France
| |
Collapse
|
46
|
Sinha T, Li D, Théveniau-Ruissy M, Hutson MR, Kelly RG, Wang J. Loss of Wnt5a disrupts second heart field cell deployment and may contribute to OFT malformations in DiGeorge syndrome. Hum Mol Genet 2014; 24:1704-16. [PMID: 25410658 DOI: 10.1093/hmg/ddu584] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Outflow tract (OFT) malformation accounts for ∼30% of human congenital heart defects and manifests frequently in TBX1 haplo-insufficiency associated DiGeorge (22q11.2 deletion) syndrome. OFT myocardium originates from second heart field (SHF) progenitors in the pharyngeal and splanchnic mesoderm (SpM), but how these progenitors are deployed to the OFT is unclear. We find that SHF progenitors in the SpM gradually gain epithelial character and are deployed to the OFT as a cohesive sheet. Wnt5a, a non-canonical Wnt, is expressed specifically in the caudal SpM and may regulate oriented cell intercalation to incorporate SHF progenitors into an epithelial-like sheet, thereby generating the pushing force to deploy SHF cells rostrally into the OFT. Using enhancer trap and Cre transgenes, our lineage tracing experiments show that in Wnt5a null mice, SHF progenitors are trapped in the SpM and fail to be deployed to the OFT efficiently, resulting in a reduction in the inferior OFT myocardial wall and its derivative, subpulmonary myocardium. Concomitantly, the superior OFT and subaortic myocardium are expanded. Finally, in chick embryos, blocking the Wnt5a function in the caudal SpM perturbs polarized elongation of SHF progenitors, and compromises their deployment to the OFT. Collectively, our results highlight a critical role for Wnt5a in deploying SHF progenitors from the SpM to the OFT. Given that Wnt5a is a putative transcriptional target of Tbx1, and the similar reduction of subpulmonary myocardium in Tbx1 mutant mice, our results suggest that perturbing Wnt5a-mediated SHF deployment may be an important pathogenic mechanism contributing to OFT malformations in DiGeorge syndrome.
Collapse
Affiliation(s)
- Tanvi Sinha
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Alabama, USA
| | - Ding Li
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Alabama, USA
| | | | - Mary R Hutson
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina
| | - Robert G Kelly
- Aix Marseille Université, CNRS, IBDM UMR 7288, Marseille 13288, France
| | - Jianbo Wang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Alabama, USA,
| |
Collapse
|
47
|
Rana MS, Théveniau-Ruissy M, De Bono C, Mesbah K, Francou A, Rammah M, Domínguez JN, Roux M, Laforest B, Anderson RH, Mohun T, Zaffran S, Christoffels VM, Kelly RG. Tbx1 Coordinates Addition of Posterior Second Heart Field Progenitor Cells to the Arterial and Venous Poles of the Heart. Circ Res 2014; 115:790-9. [DOI: 10.1161/circresaha.115.305020] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- M. Sameer Rana
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| | - Magali Théveniau-Ruissy
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| | - Christopher De Bono
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| | - Karim Mesbah
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| | - Alexandre Francou
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| | - Mayyasa Rammah
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| | - Jorge N. Domínguez
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| | - Marine Roux
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| | - Brigitte Laforest
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| | - Robert H. Anderson
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| | - Timothy Mohun
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| | - Stephane Zaffran
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| | - Vincent M. Christoffels
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| | - Robert G. Kelly
- From the Department of Anatomy, Embryology and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (M.S.R., V.M.C.); CNRS, IBDM UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K.) and Inserm, GMGF UMR S910, Faculté de Médecine de la Timone (M.R., B.L., S.Z.), Aix Marseille Université, IBDM, CNRS UMR 7288 (M.T.-R., C.D.B., K.M., A.F., M.R., R.G.K) and GMGF Inserm URM S910 (M.R., B.L., S.Z.), Marseille, France; Department of
| |
Collapse
|
48
|
Kelly RG, Buckingham ME, Moorman AF. Heart fields and cardiac morphogenesis. Cold Spring Harb Perspect Med 2014; 4:4/10/a015750. [PMID: 25274757 DOI: 10.1101/cshperspect.a015750] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this review, we focus on two important steps in the formation of the embryonic heart: (i) the progressive addition of late differentiating progenitor cells from the second heart field that drives heart tube extension during looping morphogenesis, and (ii) the emergence of patterned proliferation within the embryonic myocardium that generates distinct cardiac chambers. During the transition between these steps, the major site of proliferation switches from progenitor cells outside the early heart to proliferation within the embryonic myocardium. The second heart field and ballooning morphogenesis concepts have major repercussions on our understanding of human heart development and disease. In particular, they provide a framework to dissect the origin of congenital heart defects and the regulation of myocardial proliferation and differentiation of relevance for cardiac repair.
Collapse
Affiliation(s)
- Robert G Kelly
- Aix Marseille University, CNRS, IBDM UMR 7288, 13288 Marseilles, France
| | - Margaret E Buckingham
- Department of Developmental and Stem Cell Biology, URA CNRS 2578, Pasteur Institute, 75015 Paris, France
| | - Antoon F Moorman
- Department of Anatomy, Embryology & Physiology, Academic Medical Centre, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
49
|
Sherif HM. Heterogeneity in the Segmental Development of the Aortic Tree: Impact on Management of Genetically Triggered Aortic Aneurysms. AORTA (STAMFORD, CONN.) 2014; 2:186-95. [PMID: 26798739 PMCID: PMC4686358 DOI: 10.12945/j.aorta.2014.14-032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/07/2014] [Indexed: 11/18/2022]
Abstract
An extensive search of the medical literature examining the development of the thoracic aortic tree reveals that the thoracic aorta does not develop as one unit or in one stage: the oldest part of the thoracic aorta is the descending aorta with the aortic arch being the second oldest, developing under influence from the neural crest cell. Following in chronological order are the proximal ascending aorta and aortic root, which develop from a conotruncal origin. Different areas of the thoracic aorta develop under the influence of different gene sets. These parts develop from different cell lineages: the aortic root (the conotruncus), developing from the mesoderm; the ascending aorta and aortic arch, developing from the neural crest cells; and the descending aorta from the mesoderm. Findings illustrate that the thoracic aorta is not a single entity, in developmental terms. It develops from three or four distinct areas, at different stages of embryonic life, and under different sets of genes and signaling pathways. Genetically triggered thoracic aortic aneurysms are not a monolithic group but rather share a multi-genetic origin. Identification of therapeutic targets should be based on the predilection of certain genes to cause aneurysmal disease in specific aortic segments.
Collapse
Affiliation(s)
- Hisham M.F. Sherif
- Department of Cardiac Surgery, Christiana Hospital, Christiana Care Health System, Newark, Delaware, USA
| |
Collapse
|
50
|
Xu YJ, Chen S, Zhang J, Fang SH, Guo QQ, Wang J, Fu QH, Li F, Xu R, Sun K. Novel TBX1 loss-of-function mutation causes isolated conotruncal heart defects in Chinese patients without 22q11.2 deletion. BMC MEDICAL GENETICS 2014; 15:78. [PMID: 24998776 PMCID: PMC4099205 DOI: 10.1186/1471-2350-15-78] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 06/24/2014] [Indexed: 12/18/2022]
Abstract
Background TBX1 and CRKL haploinsufficiency is thought to cause the cardiac phenotype of the 22q11.2 deletion syndrome. However, few unequivocal mutations of TBX1 and CRKL have been discovered in isolated conotrucal heart defects (CTDs) patients. The aim of the study was to screen the mutation of TBX1 and CRKL in isolated CTDs Chinese patients without 22q11.2 deletion and identify the pathomechanism of the missense mutations. Methods We enrolled 199 non-22q11.2 deletion patients with CTDs and 139 unrelated healthy controls. Gene sequencing were performed for all of them. The functional data of mutations were obtained by in vitro transfection and luciferase experiments and computer modelling. Results Screening of the TBX1 coding sequence identified a de novo missense mutation (c.385G → A; p.E129K) and a known polymorphism (c.928G → A; p.G310S). In vitro experiments demonstrate that the TBX1E129K variant almost lost transactivation activity. The TBX1G310S variant seems to affect the interaction of TBX1 with other factors. Computer molecular dynamics simulations showed the de novo missense mutation is likely to affect TBX1-DNA interaction. No mutation of CRKL gene was found. Conclusions These observations suggest that the TBX1 loss-of-function mutation may be involved in the pathogenesis of isolated CTDs. This is the first human missense mutation showing that TBX1 is a candidate causing isolated CTDs in Chinese patients without 22q11.2 deletion.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Rang Xu
- Department of Pediatric Cardiology, Xinhua hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China.
| | | |
Collapse
|