1
|
Wolton M, Davey MG, Dietrich S. At early stages of heart development, the first and second heart fields are a continuum of lateral head mesoderm-derived, cardiogenic cells. Dev Biol 2025; 520:200-223. [PMID: 39848483 DOI: 10.1016/j.ydbio.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/25/2025]
Abstract
Pioneering work in the chicken established that the initial development of the heart consists of two stages: the quick assembly of a beating heart, followed by the recruitment of cells from adjacent tissues to deliver the mature in-and outflow tract. Cells to build the primitive heart were dubbed the first heart field (FHF) cells, cells to be recruited later the second heart field (SHF) cells. The current view is that these cells represent distinct, maybe even pre-determined lineages. However, it is still unclear where exactly FHF and SHF are located at different stages of development, and whether there is a sharp boundary or rather an overlap between the two. It is also unclear whether both FHF cells and SHF cells originate from the lateral head mesoderm (LHM), whether the paraxial head mesoderm (PHM) contributes to the SHF, and where the LHM-PHM boundary might be. To investigate this problem, we exploited the size, ease of access and exquisite anatomy of the chicken embryo and used traditional strategies as well as newly developed transgenic lines to trace the location of cardiogenic fields and boundaries from the time the first heart-markers are expressed to the time SHF cell recruitment ceases. Our work shows that both FHF and SHF stem from the LHM. We also found that FHF and SHF lack a distinct anatomical boundary. Rather, FHF and SHF are a continuum, and the recruitment of cells into the heart is a chance event depending on morphogenetic movements, the position of cells within the moving tissues, the separation of the somatic and splanchnic LHM, and the separation of the heart from the splanchnic subpharyngeal mesoderm during heart-looping. Reconciling our and previous studies we propose that first and second heart field precursors are specified but not determined, thus relying on morphogenetic processes and local environments to realise their cardiogenic potential.
Collapse
Affiliation(s)
- Matthew Wolton
- Institute of Life Sciences and Health (ILSH), School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DT, UK
| | - Megan G Davey
- Functional Genetics, The Roslin Institute, The Royal (Dick) School of Veterinary Studies, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Susanne Dietrich
- Institute of Life Sciences and Health (ILSH), School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DT, UK.
| |
Collapse
|
2
|
Eisenberg LM, Eisenberg CA. Stem Cells Associated with Adult Skeletal Muscle Can Form Beating Cardiac Tissue In Vitro in Response to Media Containing Heparin, Dexamethasone, Growth Factors and Hydrogen Peroxide. Int J Mol Sci 2025; 26:2683. [PMID: 40141327 PMCID: PMC11942180 DOI: 10.3390/ijms26062683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/25/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Both cardiac and skeletal muscles originate from the mesoderm, although the two tissues develop from distinct primordia within the early embryo. The shared, albeit distinctive muscle phenotype of these two cell types have led many researchers to investigate whether stem cells from adult skeletal muscle have the capacity to generate cells with a contractile, cardiac phenotype. To date, most of those studies have relied on multistep protocols requiring tissue engineering, co-cultures or transplantation experimentation. In this report, we describe a simple, cell culture method for obtaining contractile, cardiogenic aggregates from skeletal muscle-derived stem cells (MDSCs). Combining in vitro conditions used for promoting the differentiation of cardiac progenitor cells and the long-term maintenance of heart tissue fragments, we have been able to convert MDSCs to myocardial cells that aggregate into beating myospheres. These selective and optimized culture conditions continued to support a contractile cardiogenic phenotype for over four months in vitro. This culture protocol provides a model for future insights into the pathways responsible for the divergence of skeletal and cardiac phenotypes, as well as a source of easily obtained myocardial tissue for subsequent scientific investigations into cardiac function and biology.
Collapse
Affiliation(s)
| | - Carol A. Eisenberg
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA;
| |
Collapse
|
3
|
Arriagada C, Lin E, Schonning M, Astrof S. Mesodermal fibronectin controls cell shape, polarity, and mechanotransduction in the second heart field during cardiac outflow tract development. Dev Cell 2025; 60:62-84.e7. [PMID: 39413783 PMCID: PMC11706711 DOI: 10.1016/j.devcel.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/06/2024] [Accepted: 09/13/2024] [Indexed: 10/18/2024]
Abstract
Failure in the elongation of the cardiac outflow tract (OFT) results in congenital heart disease due to the misalignment of the great arteries with the left and right ventricles. The OFT lengthens via the accretion of progenitors from the second heart field (SHF). SHF cells are exquisitely regionalized and organized into an epithelial-like layer, forming the dorsal pericardial wall (DPW). Tissue tension, cell polarity, and proliferation within the DPW are important for the addition of SHF-derived cells to the heart and OFT elongation. However, the genes controlling these processes are not completely characterized. Using conditional mutagenesis in the mouse, we show that fibronectin (FN1) synthesized by the mesoderm coordinates multiple cellular behaviors in the anterior DPW. FN1 is enriched in the anterior DPW and plays a role in OFT elongation by maintaining a balance between pro- and anti-adhesive cell-extracellular matrix (ECM) interactions and controlling DPW cell shape, polarity, cohesion, proliferation, and mechanotransduction.
Collapse
Affiliation(s)
- Cecilia Arriagada
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Evan Lin
- Princeton Day School, Princeton, NJ, USA
| | - Michael Schonning
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA.
| |
Collapse
|
4
|
Li Y, Du J, Deng S, Liu B, Jing X, Yan Y, Liu Y, Wang J, Zhou X, She Q. The molecular mechanisms of cardiac development and related diseases. Signal Transduct Target Ther 2024; 9:368. [PMID: 39715759 DOI: 10.1038/s41392-024-02069-8] [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: 06/19/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 12/25/2024] Open
Abstract
Cardiac development is a complex and intricate process involving numerous molecular signals and pathways. Researchers have explored cardiac development through a long journey, starting with early studies observing morphological changes and progressing to the exploration of molecular mechanisms using various molecular biology methods. Currently, advancements in stem cell technology and sequencing technology, such as the generation of human pluripotent stem cells and cardiac organoids, multi-omics sequencing, and artificial intelligence (AI) technology, have enabled researchers to understand the molecular mechanisms of cardiac development better. Many molecular signals regulate cardiac development, including various growth and transcription factors and signaling pathways, such as WNT signaling, retinoic acid signaling, and Notch signaling pathways. In addition, cilia, the extracellular matrix, epigenetic modifications, and hypoxia conditions also play important roles in cardiac development. These factors play crucial roles at one or even multiple stages of cardiac development. Recent studies have also identified roles for autophagy, metabolic transition, and macrophages in cardiac development. Deficiencies or abnormal expression of these factors can lead to various types of cardiac development abnormalities. Nowadays, congenital heart disease (CHD) management requires lifelong care, primarily involving surgical and pharmacological treatments. Advances in surgical techniques and the development of clinical genetic testing have enabled earlier diagnosis and treatment of CHD. However, these technologies still have significant limitations. The development of new technologies, such as sequencing and AI technologies, will help us better understand the molecular mechanisms of cardiac development and promote earlier prevention and treatment of CHD in the future.
Collapse
Affiliation(s)
- Yingrui Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Songbai Deng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Jing
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuling Yan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajie Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaobo Zhou
- Department of Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Germany; DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
5
|
Argiro L, Chevalier C, Choquet C, Nandkishore N, Ghata A, Baudot A, Zaffran S, Lescroart F. Gastruloids are competent to specify both cardiac and skeletal muscle lineages. Nat Commun 2024; 15:10172. [PMID: 39580459 PMCID: PMC11585638 DOI: 10.1038/s41467-024-54466-w] [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: 05/17/2023] [Accepted: 11/12/2024] [Indexed: 11/25/2024] Open
Abstract
Cardiopharyngeal mesoderm contributes to the formation of the heart and head muscles. However, the mechanisms governing cardiopharyngeal mesoderm specification remain unclear. Here, we reproduce cardiopharyngeal mesoderm specification towards cardiac and skeletal muscle lineages with gastruloids from mouse embryonic stem cells. By conducting a comprehensive temporal analysis of cardiopharyngeal mesoderm development and differentiation in gastruloids compared to mouse embryos, we present the evidence for skeletal myogenesis in gastruloids. We identify different subpopulations of cardiomyocytes and skeletal muscles, the latter of which most likely correspond to different states of myogenesis with "head-like" and "trunk-like" skeletal myoblasts. In this work, we unveil the potential of gastruloids to undergo specification into both cardiac and skeletal muscle lineages, allowing the investigation of the mechanisms of cardiopharyngeal mesoderm differentiation in development and how this could be affected in congenital diseases.
Collapse
Affiliation(s)
- Laurent Argiro
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Céline Chevalier
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Caroline Choquet
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Nitya Nandkishore
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
- Department of Biotechnology, SRM Institute of Science and Technology (SRMIST), Kattankulathur, Tamil Nadu, India
| | - Adeline Ghata
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Anaïs Baudot
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Stéphane Zaffran
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France.
| | - Fabienne Lescroart
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France.
| |
Collapse
|
6
|
Feng H, Yang S, Zhang L, Zhu J, Li J, Yang Z. A new Prdm1-Cre line is suitable for studying the second heart field development. Dev Biol 2024; 514:78-86. [PMID: 38880275 DOI: 10.1016/j.ydbio.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
The second heart field (SHF) plays a pivotal role in heart development, particularly in outflow tract (OFT) morphogenesis and septation, as well as in the expansion of the right ventricle (RV). Two mouse Cre lines, the Mef2c-AHF-Cre (Mef2c-Cre) and Isl1-Cre, have been widely used to study the SHF development. However, Cre activity is triggered not only in the SHF but also in the RV in the Mef2c-Cre mice, and in the Isl1-Cre mice, Cre activation is not SHF-specific. Therefore, a more suitable SHF-Cre line is desirable for better understanding SHF development. Here, we generated and characterized the Prdm1-Cre knock-in mice. In comparison with Mef2c-Cre mice, the Cre activity is similar in the pharyngeal and splanchnic mesoderm, and in the OFT of the Prdm1-Cre mice. Nonetheless, it was noticed that Cre expression is largely reduced in the RV of Prdm1-Cre mice compared to the Mef2c-Cre mice. Furthermore, we deleted Hand2, Nkx2-5, Pdk1 and Tbx20 using both Mef2c-Cre and Prdm1-Cre mice to study OFT morphogenesis and septation, making a comparison between these two Cre lines. New insights were obtained in understanding SHF development including differentiation into cardiomyocytes in the OFT using Prdm1-Cre mice. In conclusion, we found that Prdm1-Cre mouse line is a more appropriate tool to monitor SHF development, while the Mef2c-Cre mice are excellent in studying the role and function of the SHF in OFT morphogenesis and septation.
Collapse
Affiliation(s)
- Haiyue Feng
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Suming Yang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Lijun Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Jingai Zhu
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Jinsong Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
7
|
Zubrzycki M, Schramm R, Costard-Jäckle A, Grohmann J, Gummert JF, Zubrzycka M. Cardiac Development and Factors Influencing the Development of Congenital Heart Defects (CHDs): Part I. Int J Mol Sci 2024; 25:7117. [PMID: 39000221 PMCID: PMC11241401 DOI: 10.3390/ijms25137117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
The traditional description of cardiac development involves progression from a cardiac crescent to a linear heart tube, which in the phase of transformation into a mature heart forms a cardiac loop and is divided with the septa into individual cavities. Cardiac morphogenesis involves numerous types of cells originating outside the initial cardiac crescent, including neural crest cells, cells of the second heart field origin, and epicardial progenitor cells. The development of the fetal heart and circulatory system is subject to regulatation by both genetic and environmental processes. The etiology for cases with congenital heart defects (CHDs) is largely unknown, but several genetic anomalies, some maternal illnesses, and prenatal exposures to specific therapeutic and non-therapeutic drugs are generally accepted as risk factors. New techniques for studying heart development have revealed many aspects of cardiac morphogenesis that are important in the development of CHDs, in particular transposition of the great arteries.
Collapse
Affiliation(s)
- Marek Zubrzycki
- Department of Surgery for Congenital Heart Defects, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany;
| | - Rene Schramm
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany; (R.S.); (A.C.-J.); (J.F.G.)
| | - Angelika Costard-Jäckle
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany; (R.S.); (A.C.-J.); (J.F.G.)
| | - Jochen Grohmann
- Department of Congenital Heart Disease/Pediatric Cardiology, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany;
| | - Jan F. Gummert
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany; (R.S.); (A.C.-J.); (J.F.G.)
| | - Maria Zubrzycka
- Department of Clinical Physiology, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| |
Collapse
|
8
|
Aurigemma I, Lanzetta O, Cirino A, Allegretti S, Lania G, Ferrentino R, Poondi Krishnan V, Angelini C, Illingworth E, Baldini A. Endothelial gene regulatory elements associated with cardiopharyngeal lineage differentiation. Commun Biol 2024; 7:351. [PMID: 38514806 PMCID: PMC10957928 DOI: 10.1038/s42003-024-06017-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 03/06/2024] [Indexed: 03/23/2024] Open
Abstract
Endothelial cells (EC) differentiate from multiple sources, including the cardiopharyngeal mesoderm, which gives rise also to cardiac and branchiomeric muscles. The enhancers activated during endothelial differentiation within the cardiopharyngeal mesoderm are not completely known. Here, we use a cardiogenic mesoderm differentiation model that activates an endothelial transcription program to identify endothelial regulatory elements activated in early cardiogenic mesoderm. Integrating chromatin remodeling and gene expression data with available single-cell RNA-seq data from mouse embryos, we identify 101 putative regulatory elements of EC genes. We then apply a machine-learning strategy, trained on validated enhancers, to predict enhancers. Using this computational assay, we determine that 50% of these sequences are likely enhancers, some of which are already reported. We also identify a smaller set of regulatory elements of well-known EC genes and validate them using genetic and epigenetic perturbation. Finally, we integrate multiple data sources and computational tools to search for transcriptional factor binding motifs. In conclusion, we show EC regulatory sequences with a high likelihood to be enhancers, and we validate a subset of them using computational and cell culture models. Motif analyses show that the core EC transcription factors GATA/ETS/FOS is a likely driver of EC regulation in cardiopharyngeal mesoderm.
Collapse
Affiliation(s)
- Ilaria Aurigemma
- PhD program in Molecular Medicine and Medical Biotechnology, University Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II 132, 84084, Fisciano, Italy
| | - Olga Lanzetta
- Institute of Genetics and Biophysics, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Andrea Cirino
- Institute of Genetics and Biophysics, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Sara Allegretti
- PhD program in Molecular Medicine and Medical Biotechnology, University Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Gabriella Lania
- Institute of Genetics and Biophysics, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Rosa Ferrentino
- Institute of Genetics and Biophysics, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Varsha Poondi Krishnan
- Institute of Genetics and Biophysics, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Claudia Angelini
- Istituto Applicazioni del Calcolo, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Elizabeth Illingworth
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II 132, 84084, Fisciano, Italy
| | - Antonio Baldini
- PhD program in Molecular Medicine and Medical Biotechnology, University Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
9
|
Guijarro C, Kelly RG. On the involvement of the second heart field in congenital heart defects. C R Biol 2024; 347:9-18. [PMID: 38488639 DOI: 10.5802/crbiol.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 03/19/2024]
Abstract
Congenital heart defects (CHD) affect 1 in 100 live births and result from defects in cardiac development. Growth of the early heart tube occurs by the progressive addition of second heart field (SHF) progenitor cells to the cardiac poles. The SHF gives rise to ventricular septal, right ventricular and outflow tract myocardium at the arterial pole, and atrial, including atrial septal myocardium, at the venous pole. SHF deployment creates the template for subsequent cardiac septation and has been implicated in cardiac looping and in orchestrating outflow tract development with neural crest cells. Genetic or environmental perturbation of SHF deployment thus underlies a spectrum of common forms of CHD affecting conotruncal and septal morphogenesis. Here we review the major properties of SHF cells as well as recent insights into the developmental programs that drive normal cardiac progenitor cell addition and the origins of CHD.
Collapse
|
10
|
Buckingham M, Kelly RG. Cardiac Progenitor Cells of the First and Second Heart Fields. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:103-124. [PMID: 38884707 DOI: 10.1007/978-3-031-44087-8_5] [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
The heart forms from the first and second heart fields, which contribute to distinct regions of the myocardium. This is supported by clonal analyses, which identify corresponding first and second cardiac cell lineages in the heart. Progenitor cells of the second heart field and its sub-domains are controlled by a gene regulatory network and signaling pathways, which determine their behavior. Multipotent cells in this field can also contribute cardiac endothelial and smooth muscle cells. Furthermore, the skeletal muscles of the head and neck are clonally related to myocardial cells that form the arterial and venous poles of the heart. These lineage relationships, together with the genes that regulate the heart fields, have major implications for congenital heart disease.
Collapse
Affiliation(s)
- Margaret Buckingham
- Department of Developmental and Stem Cell Biology, CNRS UMR 3738, Institut Pasteur, Paris, France.
| | - Robert G Kelly
- Aix Marseille Université, Institut de Biologie du Dévelopment de Marseille, Marseille, France.
| |
Collapse
|
11
|
Bileckyj C, Blotz B, Cripps RM. Drosophila as a Model to Understand Second Heart Field Development. J Cardiovasc Dev Dis 2023; 10:494. [PMID: 38132661 PMCID: PMC10744189 DOI: 10.3390/jcdd10120494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
The genetic model system Drosophila has contributed fundamentally to our understanding of mammalian heart specification, development, and congenital heart disease. The relatively simple Drosophila heart is a linear muscular tube that is specified and develops in the embryo and persists throughout the life of the animal. It functions at all stages to circulate hemolymph within the open circulatory system of the body. During Drosophila metamorphosis, the cardiac tube is remodeled, and a new layer of muscle fibers spreads over the ventral surface of the heart to form the ventral longitudinal muscles. The formation of these fibers depends critically upon genes known to be necessary for mammalian second heart field (SHF) formation. Here, we review the prior contributions of the Drosophila system to the understanding of heart development and disease, discuss the importance of the SHF to mammalian heart development and disease, and then discuss how the ventral longitudinal adult cardiac muscles can serve as a novel model for understanding SHF development and disease.
Collapse
Affiliation(s)
| | | | - Richard M. Cripps
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| |
Collapse
|
12
|
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: 0.5] [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
|
13
|
Tyser RCV. Formation of the Heart: Defining Cardiomyocyte Progenitors at Single-Cell Resolution. Curr Cardiol Rep 2023; 25:495-503. [PMID: 37119451 PMCID: PMC10188409 DOI: 10.1007/s11886-023-01880-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/04/2023] [Indexed: 05/01/2023]
Abstract
PURPOSE OF REVIEW Formation of the heart requires the coordinated addition of multiple progenitor sources which have undergone different pathways of specification and differentiation. In this review, I aim to put into context how recent studies defining cardiac progenitor heterogeneity build on our understanding of early heart development and also discuss the questions raised by this new insight. RECENT FINDINGS With the development of sequencing technologies and imaging approaches, it has been possible to define, at high temporal resolution, the molecular profile and anatomical location of cardiac progenitors at the single-cell level, during the formation of the mammalian heart. Given the recent progress in our understanding of early heart development and technical advances in high-resolution time-lapse imaging and lineage analysis, we are now in a position of great potential, allowing us to resolve heart formation at previously impossible levels of detail. Understanding how this essential organ forms not only addresses questions of fundamental biological significance but also provides a blueprint for strategies to both treat and model heart disease.
Collapse
Affiliation(s)
- Richard C V Tyser
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge, CB2 0AW, UK.
| |
Collapse
|
14
|
Baral K, D'amato G, Kuschel B, Bogan F, Jones BW, Large CL, Whatley JD, Red-Horse K, Sharma B. APJ+ cells in the SHF contribute to the cells of aorta and pulmonary trunk through APJ signaling. Dev Biol 2023; 498:77-86. [PMID: 37037405 DOI: 10.1016/j.ydbio.2023.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 03/26/2023] [Accepted: 04/07/2023] [Indexed: 04/12/2023]
Abstract
Outflow tract develops from cardiac progenitor cells in the second heart field (SHF) domain. APJ, a G-Protein Coupled Receptor, is expressed by cardiac progenitor cells in the SHF. By lineage tracing APJ + SHF cells, we show that these cardiac progenitor cell contribute to the cells of outflow tract (OFT), which eventually give rise to aorta and pulmonary trunk/artery upon its morphogenesis. Furthermore, we show that early APJ + cells give rise to both aorta and pulmonary cells but late APJ + cells predominantly give rise to pulmonary cells. APJ is expressed by the outflow tract progenitors but its role in the SHF is unclear. We performed knockout studies to determine the role of APJ in SHF cell proliferation and survival. Our data suggested that APJ knockout in the SHF reduced the proliferation of SHF progenitors, while there was no significant impact on survival of the SHF progenitors. In addition, we show that ectopic overexpression of WNT in these cells disrupted aorta and pulmonary morphogenesis from outflow tract. Overall, our study have identified APJ + progenitor population within the SHF that give rise to aorta and pulmonary trunk/artery cells. Furthermore, we show that APJ signaling stimulate proliferation of these cells in the SHF.
Collapse
Affiliation(s)
- Kamal Baral
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Gaetano D'amato
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Bryce Kuschel
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Frank Bogan
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Brendan W Jones
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Colton L Large
- Department of Biology, Ball State University, Muncie, IN, USA
| | | | | | - Bikram Sharma
- Department of Biology, Ball State University, Muncie, IN, USA.
| |
Collapse
|
15
|
Yahya I, Brand-Saberi B, Morosan-Puopolo G. Chicken embryo as a model in second heart field development. Heliyon 2023; 9:e14230. [PMID: 36923876 PMCID: PMC10009738 DOI: 10.1016/j.heliyon.2023.e14230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/30/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Previously, a single source of progenitor cells was thought to be responsible for the formation of the cardiac muscle. However, the second heart field has recently been identified as an additional source of myocardial progenitor cells. The chicken embryo, which develops in the egg, outside the mother can easily be manipulated in vivo and in vitro. Hence, it was an excellent model for establishing the concept of the second heart field. Here, our review will focus on the chicken model, specifically its role in understanding the second heart field. In addition to discussing historical aspects, we provide an overview of recent findings that have helped to define the chicken second heart field progenitor cells. A better understanding of the second heart field development will provide important insights into the congenital malformations affecting cardiac muscle formation and function.
Collapse
Affiliation(s)
- Imadeldin Yahya
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801, Bochum, Germany
- Department of Anatomy, Faculty of Veterinary Medicine, University of Khartoum, Khartoum, 11115, Sudan
- Corresponding author. Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801 Bochum, Germany.
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801, Bochum, Germany
| | | |
Collapse
|
16
|
Restivo A, di Gioia C, Marino B, Putotto C. Transpositions of the great arteries versus aortic dextropositions. A review of some embryogenetic and morphological aspects. Anat Rec (Hoboken) 2023; 306:502-514. [PMID: 36426596 DOI: 10.1002/ar.25129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/15/2022] [Accepted: 10/18/2022] [Indexed: 11/27/2022]
Abstract
This review examines and discusses the morphology and embryology of two main groups of conotruncal cardiac malformations: (a) transposition of the great arteries (complete transposition and incomplete/partial transposition namely double outlet right ventricle), and (b) aortic dextroposition defects (tetralogy of Fallot and Eisenmenger malformation). In both groups, persistent truncus arteriosus was included because maldevelopment of the neural crest cell supply to the outflow tract, contributing to the production of the persistent truncus arteriosus, is shared by both groups of malformations. The potentially important role of the proximal conal cushions in the rotatory sequence of the conotruncus is emphasized. Most importantly, this study emphasizes the differentiation between the double-outlet right ventricle, which is a partial or incomplete transposition of the great arteries, and the Eisenmenger malformation, which is an aortic dextroposition. Special emphasis is also given to the leftward shift of the conoventricular junction, which covers an important morphogenetic role in both aortic dextropositions and transposition defects as well as in normal development, and whose molecular genetic regulation seems to remain unclear at present. Emphasis is placed on the distinct and overlapping roles of Tbx1 and Pitx2 transcription factors in modulating the development of the cardiac outflow tract.
Collapse
Affiliation(s)
- Angelo Restivo
- Pediatric Cardiology Unit, Department of Pediatrics, Obstetrics and Gynecology, Sapienza University of Rome, Rome, Italy.,Museum of Pathological Anatomy, Sapienza University of Rome, Rome, Italy
| | - Cira di Gioia
- Museum of Pathological Anatomy, Sapienza University of Rome, Rome, Italy.,Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Bruno Marino
- Pediatric Cardiology Unit, Department of Pediatrics, Obstetrics and Gynecology, Sapienza University of Rome, Rome, Italy
| | - Carolina Putotto
- Pediatric Cardiology Unit, Department of Pediatrics, Obstetrics and Gynecology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
17
|
Human Heart Morphogenesis: A New Vision Based on In Vivo Labeling and Cell Tracking. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010165. [PMID: 36676114 PMCID: PMC9861877 DOI: 10.3390/life13010165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/24/2022] [Accepted: 12/27/2022] [Indexed: 01/09/2023]
Abstract
Despite the extensive information available on the different genetic, epigenetic, and molecular features of cardiogenesis, the origin of congenital heart defects remains unknown. Most genetic and molecular studies have been conducted outside the context of the progressive anatomical and histological changes in the embryonic heart, which is one of the reasons for the limited knowledge of the origins of congenital heart diseases. We integrated the findings of descriptive studies on human embryos and experimental studies on chick, rat, and mouse embryos. This research is based on the new dynamic concept of heart development and the existence of two heart fields. The first field corresponds to the straight heart tube, into which splanchnic mesodermal cells from the second heart field are gradually recruited. The overall aim was to create a new vision for the analysis, diagnosis, and regionalized classification of congenital defects of the heart and great arteries. In addition to highlighting the importance of genetic factors in the development of congenital heart disease, this study provides new insights into the composition of the straight heart tube, the processes of twisting and folding, and the fate of the conus in the development of the right ventricle and its outflow tract. The new vision, based on in vivo labeling and cell tracking and enhanced by models such as gastruloids and organoids, has contributed to a better understanding of important errors in cardiac morphogenesis, which may lead to several congenital heart diseases.
Collapse
|
18
|
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
|
19
|
Ding S, Zhang X, Qiu H, Wo J, Zhang F, Na J. Non-cardiomyocytes in the heart in embryo development, health, and disease, a single-cell perspective. Front Cell Dev Biol 2022; 10:873264. [PMID: 36393852 PMCID: PMC9661523 DOI: 10.3389/fcell.2022.873264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 10/14/2022] [Indexed: 11/25/2022] Open
Abstract
Recent single-cell atlases of the heart gave unprecedented details about the diversity of cell types and states during heart development in health and disease conditions. Beyond a profiling tool, researchers also use single-cell analyses to dissect the mechanism of diseases in animal models. The new knowledge from these studies revealed that beating cardiomyocytes account for less than 50% of the total heart cell population. In contrast, non-cardiomyocytes (NCMs), such as cardiac fibroblasts, endothelial cells, and immune cells, make up the remaining proportion and have indispensable roles in structural support, homeostasis maintenance, and injury repair of the heart. In this review, we categorize the composition and characteristics of NCMs from the latest single-cell studies of the heart in various contexts and compare the findings from both human samples and mouse models. This information will enrich our understanding of the cellular basis of heart development and diseases and provide insights into the potential therapeutic targets in NCMs to repair the heart.
Collapse
Affiliation(s)
- Shuangyuan Ding
- School of Medicine, Tsinghua University, Beijing, China
- Center for Life Sciences, Tsinghua University and Peking University, Beijing, China
- *Correspondence: Shuangyuan Ding, ; Jie Na,
| | - Xingwu Zhang
- School of Medicine, Tsinghua University, Beijing, China
| | - Hui Qiu
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Jiaoyang Wo
- Center for Life Sciences, Tsinghua University and Peking University, Beijing, China
| | - Fengzhi Zhang
- Central Laboratory, First Hospital of Tsinghua University, Beijing, China
| | - Jie Na
- School of Medicine, Tsinghua University, Beijing, China
- *Correspondence: Shuangyuan Ding, ; Jie Na,
| |
Collapse
|
20
|
Rammah M, Théveniau-Ruissy M, Sturny R, Rochais F, Kelly RG. PPARγ and NOTCH Regulate Regional Identity in the Murine Cardiac Outflow Tract. Circ Res 2022; 131:842-858. [PMID: 36205127 DOI: 10.1161/circresaha.122.320766] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 09/14/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND The arterial pole of the heart is a hotspot for life-threatening forms of congenital heart defects (CHDs). Development of this cardiac region occurs by addition of Second Heart Field (SHF) progenitor cells to the embryonic outflow tract (OFT) and subsequently the base of the ascending aorta and pulmonary trunk. Understanding the cellular and genetic mechanisms driving arterial pole morphogenesis is essential to provide further insights into the cause of CHDs. METHODS A synergistic combination of bioinformatic analysis and mouse genetics as well as embryo and explant culture experiments were used to dissect the cross-regulatory transcriptional circuitry operating in future subaortic and subpulmonary OFT myocardium. RESULTS Here, we show that the lipid sensor PPARγ (peroxisome proliferator-activated receptor gamma) is expressed in future subpulmonary myocardium in the inferior wall of the OFT and that PPARγ signaling-related genes display regionalized OFT expression regulated by the transcription factor TBX1 (T-box transcription factor 1). Modulating PPARγ activity in ex vivo cultured embryos treated with a PPARγ agonist or antagonist or deleting Pparγ in cardiac progenitor cells using Mesp1-Cre reveals that Pparγ is required for addition of future subpulmonary myocardium and normal arterial pole development. Additionally, the non-canonical DLK1 (delta-like noncanonical Notch ligand 1)/NOTCH (Notch receptor 1)/HES1 (Hes family bHLH transcription factor 1) pathway negatively regulates Pparγ in future subaortic myocardium in the superior OFT wall. CONCLUSIONS Together these results identify Pparγ as a regulator of regional transcriptional identity in the developing heart, providing new insights into gene interactions involved in congenital heart defects.
Collapse
Affiliation(s)
- Mayyasa Rammah
- Aix Marseille University, CNRS UMR 7288, IBDM, Marseille, France (M.R., M.T.R., R.S., F.R., R.G.K.)
| | - Magali Théveniau-Ruissy
- Aix Marseille University, CNRS UMR 7288, IBDM, Marseille, France (M.R., M.T.R., R.S., F.R., R.G.K.)
- Aix Marseille Univ, INSERM, MMG, Marseille, France (M.T.R., F.R.)
| | - Rachel Sturny
- Aix Marseille University, CNRS UMR 7288, IBDM, Marseille, France (M.R., M.T.R., R.S., F.R., R.G.K.)
| | - Francesca Rochais
- Aix Marseille University, CNRS UMR 7288, IBDM, Marseille, France (M.R., M.T.R., R.S., F.R., R.G.K.)
- Aix Marseille Univ, INSERM, MMG, Marseille, France (M.T.R., F.R.)
| | - Robert G Kelly
- Aix Marseille University, CNRS UMR 7288, IBDM, Marseille, France (M.R., M.T.R., R.S., F.R., R.G.K.)
| |
Collapse
|
21
|
Nodal signaling regulates asymmetric cellular behaviors, driving clockwise rotation of the heart tube in zebrafish. Commun Biol 2022; 5:996. [PMID: 36131094 PMCID: PMC9492702 DOI: 10.1038/s42003-022-03826-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Abstract
Clockwise rotation of the primitive heart tube, a process regulated by restricted left-sided Nodal signaling, is the first morphological manifestation of left-right asymmetry. How Nodal regulates cell behaviors to drive asymmetric morphogenesis remains poorly understood. Here, using high-resolution live imaging of zebrafish embryos, we simultaneously visualized cellular dynamics underlying early heart morphogenesis and resulting changes in tissue shape, to identify two key cell behaviors: cell rearrangement and cell shape change, which convert initially flat heart primordia into a tube through convergent extension. Interestingly, left cells were more active in these behaviors than right cells, driving more rapid convergence of the left primordium, and thereby rotating the heart tube. Loss of Nodal signaling abolished the asymmetric cell behaviors as well as the asymmetric convergence of the left and right heart primordia. Collectively, our results demonstrate that Nodal signaling regulates the magnitude of morphological changes by acting on basic cellular behaviors underlying heart tube formation, driving asymmetric deformation and rotation of the heart tube.
Collapse
|
22
|
Yang D, Gomez-Garcia J, Funakoshi S, Tran T, Fernandes I, Bader GD, Laflamme MA, Keller GM. Modeling human multi-lineage heart field development with pluripotent stem cells. Cell Stem Cell 2022; 29:1382-1401.e8. [PMID: 36055193 DOI: 10.1016/j.stem.2022.08.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 07/01/2022] [Accepted: 08/11/2022] [Indexed: 12/27/2022]
Abstract
The cardiomyocyte (CM) subtypes in the mammalian heart derive from distinct lineages known as the first heart field (FHF), the anterior second heart field (aSHF), and the posterior second heart field (pSHF) lineages that are specified during gastrulation. We modeled human heart field development from human pluripotent stem cells (hPSCs) by using single-cell RNA-sequencing to delineate lineage specification and progression. Analyses of hPSC-derived and mouse mesoderm transcriptomes enabled the identification of distinct human FHF, aSHF, and pSHF mesoderm subpopulations. Through staged manipulation of signaling pathways identified from transcriptomics, we generated myocyte populations that display molecular characteristics of key CM subtypes. The developmental trajectory of the human cardiac lineages recapitulated that of the mouse, demonstrating conserved cardiovascular programs. These findings establish a comprehensive landscape of human embryonic cardiogenesis that provides access to a broad spectrum of cardiomyocytes for modeling congenital heart diseases and chamber-specific cardiomyopathies as well as for developing new therapies to treat them.
Collapse
Affiliation(s)
- Donghe Yang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
| | - Juliana Gomez-Garcia
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Biomedical Engineering, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Shunsuke Funakoshi
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Thinh Tran
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Ian Fernandes
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Gary D Bader
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Michael A Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gordon M Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
23
|
The negative regulation of gene expression by microRNAs as key driver of inducers and repressors of cardiomyocyte differentiation. Clin Sci (Lond) 2022; 136:1179-1203. [PMID: 35979890 PMCID: PMC9411751 DOI: 10.1042/cs20220391] [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: 06/09/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/28/2022]
Abstract
Cardiac muscle damage-induced loss of cardiomyocytes (CMs) and dysfunction of the remaining ones leads to heart failure, which nowadays is the number one killer worldwide. Therapies fostering effective cardiac regeneration are the holy grail of cardiovascular research to stop the heart failure epidemic. The main goal of most myocardial regeneration protocols is the generation of new functional CMs through the differentiation of endogenous or exogenous cardiomyogenic cells. Understanding the cellular and molecular basis of cardiomyocyte commitment, specification, differentiation and maturation is needed to devise innovative approaches to replace the CMs lost after injury in the adult heart. The transcriptional regulation of CM differentiation is a highly conserved process that require sequential activation and/or repression of different genetic programs. Therefore, CM differentiation and specification have been depicted as a step-wise specific chemical and mechanical stimuli inducing complete myogenic commitment and cell-cycle exit. Yet, the demonstration that some microRNAs are sufficient to direct ESC differentiation into CMs and that four specific miRNAs reprogram fibroblasts into CMs show that CM differentiation must also involve negative regulatory instructions. Here, we review the mechanisms of CM differentiation during development and from regenerative stem cells with a focus on the involvement of microRNAs in the process, putting in perspective their negative gene regulation as a main modifier of effective CM regeneration in the adult heart.
Collapse
|
24
|
Lu P, Wang P, Wu B, Wang Y, Liu Y, Cheng W, Feng X, Yuan X, Atteya MM, Ferro H, Sugi Y, Rydquist G, Esmaily M, Butcher JT, Chang CP, Lenz J, Zheng D, Zhou B. A SOX17-PDGFB signaling axis regulates aortic root development. Nat Commun 2022; 13:4065. [PMID: 35831318 PMCID: PMC9279414 DOI: 10.1038/s41467-022-31815-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 06/30/2022] [Indexed: 11/08/2022] Open
Abstract
Developmental etiologies causing complex congenital aortic root abnormalities are unknown. Here we show that deletion of Sox17 in aortic root endothelium in mice causes underdeveloped aortic root leading to a bicuspid aortic valve due to the absence of non-coronary leaflet and mispositioned left coronary ostium. The respective defects are associated with reduced proliferation of non-coronary leaflet mesenchyme and aortic root smooth muscle derived from the second heart field cardiomyocytes. Mechanistically, SOX17 occupies a Pdgfb transcriptional enhancer to promote its transcription and Sox17 deletion inhibits the endothelial Pdgfb transcription and PDGFB growth signaling to the non-coronary leaflet mesenchyme. Restoration of PDGFB in aortic root endothelium rescues the non-coronary leaflet and left coronary ostium defects in Sox17 nulls. These data support a SOX17-PDGFB axis underlying aortic root development that is critical for aortic valve and coronary ostium patterning, thereby informing a potential shared disease mechanism for concurrent anomalous aortic valve and coronary arteries.
Collapse
Affiliation(s)
- Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ping Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yidong Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Cardiovascular Research Center, School of Basic Medical Sciences, Jiaotong University, Xi'an, Shanxi, China
| | - Yang Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wei Cheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xuhui Feng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xinchun Yuan
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Miriam M Atteya
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Haleigh Ferro
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Yukiko Sugi
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Grant Rydquist
- School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Mahdi Esmaily
- School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | | | - Ching-Pin Chang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jack Lenz
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
- Departments of Neurology and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Bin Zhou
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
- Departments of Pediatrics and Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
25
|
Sun M, Zhang S, Li Y, Chen L, Diao J, Li J, Wei J, Song X, Liu Y, Shu J, Wang T, Huang P, Qin J. Effect of Maternal Antidepressant Use During the Pre-pregnancy/Early Pregnancy Period on Congenital Heart Disease: A Prospective Cohort Study in Central China. Front Cardiovasc Med 2022; 9:916882. [PMID: 35865384 PMCID: PMC9294218 DOI: 10.3389/fcvm.2022.916882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundWith the increase in maternal antidepressant prescribing before/during pregnancy, concerns about the safety of antidepressants have come into focus. The purpose of this study was to explore the association between maternal antidepressant use before pregnancy/in early pregnancy and the risk of congenital heart disease (CHD) in children, and to provide a scientific basis for clinical safety of antidepressant use.MethodsThe prospective cohort study ultimately included 34,104 singleton pregnancies. Modified Poisson regression model with robust error variances was used to evaluate RRs and 95% confidence intervals (CIs) for the risk of CHD in offspring exposed to maternal antidepressant in the 3 months before pregnancy and early pregnancy. In addition, sensitivity analysis was further performed to explore the robustness of the results.ResultsIn this study, the maternal antidepressant exposure rate was 2.83% in the 3 months before pregnancy, 2.42% in early pregnancy, and the incidence of CHD was 8.973 per 1,000 live births. We found that maternal antidepressant use in the 3 months before pregnancy and early pregnancy were all associated with an increased risk of CHD, ~2.54 times and 2.87 times, respectively, of non-use of antidepressants after adjusting for potential confounders. This association was also found in CHD specific phenotypic analysis. Of these, offspring whose mothers were exposed to antidepressants in the 3 months before pregnancy had the highest risk of transposition of the great arteries (aOR = 5.50, 95% CI: 1.91–15.88). The offspring of mothers exposed to antidepressants in early pregnancy had the highest risk of developing ventricular septal defect (aOR = 4.80, 95% CI: 2.50–9.24). Sensitivity analysis verified the stability of the results.ConclusionsMaternal antidepressant use in the 3 months before pregnancy and early pregnancy were all associated with an increased risk of CHD in their offspring. In order to reduce the risk of teratogenesis, we recommend that pregnant women prepare for pregnancy after their condition improves or receive the minimum effective dose of medication.
Collapse
Affiliation(s)
- Mengting Sun
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Senmao Zhang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Yihuan Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Letao Chen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Jingyi Diao
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Jinqi Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Jianhui Wei
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Xinli Song
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Yiping Liu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Jing Shu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Tingting Wang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
- *Correspondence: Tingting Wang ; orcid.org/0000-0003-1689-7257
| | - Peng Huang
- Department of Cardiothoracic Surgery, Hunan Children's Hospital, Changsha, China
- Peng Huang ; orcid.org/0000-0001-6015-6371
| | - Jiabi Qin
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
- Jiabi Qin ; orcid.org/0000-0002-9360-4991
| |
Collapse
|
26
|
Gonzalez DM, Schrode N, Ebrahim TAM, Broguiere N, Rossi G, Drakhlis L, Zweigerdt R, Lutolf MP, Beaumont KG, Sebra R, Dubois NC. Dissecting mechanisms of chamber-specific cardiac differentiation and its perturbation following retinoic acid exposure. Development 2022; 149:dev200557. [PMID: 35686629 PMCID: PMC9340554 DOI: 10.1242/dev.200557] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/26/2022] [Indexed: 05/30/2025]
Abstract
The specification of distinct cardiac lineages occurs before chamber formation and acquisition of bona fide atrial or ventricular identity. However, the mechanisms underlying these early specification events remain poorly understood. Here, we performed single cell analysis at the murine cardiac crescent, primitive heart tube and heart tube stages to uncover the transcriptional mechanisms underlying formation of atrial and ventricular cells. We find that progression towards differentiated cardiomyocytes occurs primarily based on heart field progenitor identity, and that progenitors contribute to ventricular or atrial identity through distinct differentiation mechanisms. We identify new candidate markers that define such differentiation processes and examine their expression dynamics using computational lineage trajectory methods. We further show that exposure to exogenous retinoic acid causes defects in ventricular chamber size, dysregulation in FGF signaling and a shunt in differentiation towards orthogonal lineages. Retinoic acid also causes defects in cell-cycle exit resulting in formation of hypomorphic ventricles. Collectively, our data identify, at a single cell level, distinct lineage trajectories during cardiac specification and differentiation, and the precise effects of manipulating cardiac progenitor patterning via retinoic acid signaling.
Collapse
Affiliation(s)
- David M. Gonzalez
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nadine Schrode
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Tasneem A. M. Ebrahim
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nicolas Broguiere
- Laboratory of Stem Cell Bioengineering, School of Life Sciences, EPFL, Lausanne CH-1015, Switzerland
| | - Giuliana Rossi
- Laboratory of Stem Cell Bioengineering, School of Life Sciences, EPFL, Lausanne CH-1015, Switzerland
| | - Lika Drakhlis
- Roche Institute for Translational Bioengineering, Roche Pharma Research and Early Development, Basel 4052, Switzerland
| | - Robert Zweigerdt
- Roche Institute for Translational Bioengineering, Roche Pharma Research and Early Development, Basel 4052, Switzerland
| | - Matthias P. Lutolf
- Laboratory of Stem Cell Bioengineering, School of Life Sciences, EPFL, Lausanne CH-1015, Switzerland
- Roche Institute for Translational Bioengineering, Roche Pharma Research and Early Development, Basel 4052, Switzerland
| | - Kristin G. Beaumont
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH–Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Robert Sebra
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Sema4, a Mount Sinai venture, Stamford, CT 06902, USA
| | - Nicole C. Dubois
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
27
|
Abrial M, Basu S, Huang M, Butty V, Schwertner A, Jeffrey S, Jordan D, Burns CE, Burns CG. Latent TGFβ-binding proteins 1 and 3 protect the larval zebrafish outflow tract from aneurysmal dilatation. Dis Model Mech 2022; 15:dmm046979. [PMID: 35098309 PMCID: PMC8990920 DOI: 10.1242/dmm.046979] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 01/13/2022] [Indexed: 11/20/2022] Open
Abstract
Aortic root aneurysm is a common cause of morbidity and mortality in Loeys-Dietz and Marfan syndromes, where perturbations in transforming growth factor beta (TGFβ) signaling play a causal or contributory role, respectively. Despite the advantages of cross-species disease modeling, animal models of aortic root aneurysm are largely restricted to genetically engineered mice. Here, we report that zebrafish devoid of the genes encoding latent-transforming growth factor beta-binding protein 1 and 3 (ltbp1 and ltbp3, respectively) develop rapid and severe aneurysm of the outflow tract (OFT), the aortic root equivalent. Similar to syndromic aneurysm tissue, the distended OFTs display evidence for paradoxical hyperactivated TGFβ signaling. RNA-sequencing revealed significant overlap between the molecular signatures of disease tissue from mutant zebrafish and a mouse model of Marfan syndrome. Moreover, chemical inhibition of TGFβ signaling in wild-type animals phenocopied mutants but chemical activation did not, demonstrating that TGFβ signaling is protective against aneurysm. Human relevance is supported by recent studies implicating genetic lesions in LTBP3 and, potentially, LTBP1 as heritable causes of aortic root aneurysm. Ultimately, our data demonstrate that zebrafish can now be leveraged to interrogate thoracic aneurysmal disease and identify novel lead compounds through small-molecule suppressor screens. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Maryline Abrial
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Sandeep Basu
- Harvard Medical School, Boston, MA 02115, USA
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Mengmeng Huang
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Vincent Butty
- BioMicroCenter, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Asya Schwertner
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Spencer Jeffrey
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Daniel Jordan
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Caroline E. Burns
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - C. Geoffrey Burns
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| |
Collapse
|
28
|
Bonet F, Inácio JM, Bover O, Añez SB, Belo JA. CCBE1 in Cardiac Development and Disease. Front Genet 2022; 13:836694. [PMID: 35222551 PMCID: PMC8864227 DOI: 10.3389/fgene.2022.836694] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/19/2022] [Indexed: 12/04/2022] Open
Abstract
The collagen- and calcium-binding EGF-like domains 1 (CCBE1) is a secreted protein extensively described as indispensable for lymphangiogenesis during development enhancing VEGF-C signaling. In human patients, mutations in CCBE1 have been found to cause Hennekam syndrome, an inherited disease characterized by malformation of the lymphatic system that presents a wide variety of symptoms such as primary lymphedema, lymphangiectasia, and heart defects. Importantly, over the last decade, an essential role for CCBE1 during heart development is being uncovered. In mice, Ccbe1 expression was initially detected in distinct cardiac progenitors such as first and second heart field, and the proepicardium. More recently, Ccbe1 expression was identified in the epicardium and sinus venosus (SV) myocardium at E11.5–E13.5, the stage when SV endocardium–derived (VEGF-C dependent) coronary vessels start to form. Concordantly, CCBE1 is required for the correct formation of the coronary vessels and the coronary artery stem in the mouse. Additionally, Ccbe1 was found to be enriched in mouse embryonic stem cells (ESC) and revealed as a new essential gene for the differentiation of ESC-derived early cardiac precursor cell lineages. Here, we bring an up-to-date review on the role of CCBE1 in cardiac development, function, and human disease implications. Finally, we envisage the potential of this molecule’s functions from a regenerative medicine perspective, particularly novel therapeutic strategies for heart disease.
Collapse
Affiliation(s)
- Fernando Bonet
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
- Medicine Department, School of Medicine, University of Cádiz (UCA), Cádiz, Spain
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, Cádiz, Spain
| | - José M. Inácio
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Oriol Bover
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Sabrina B. Añez
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
| | - José A. Belo
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
- *Correspondence: José A. Belo,
| |
Collapse
|
29
|
Ebrahimi N, Bradley C, Hunter P. An integrative multiscale view of early cardiac looping. WIREs Mech Dis 2022; 14:e1535. [PMID: 35023324 DOI: 10.1002/wsbm.1535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 11/12/2022]
Abstract
The heart is the first organ to form and function during the development of an embryo. Heart development consists of a series of events believed to be highly conserved in vertebrates. Development of heart begins with the formation of the cardiac fields followed by a linear heart tube formation. The straight heart tube then undergoes a ventral bending prior to further bending and helical torsion to form a looped heart. The looping phase is then followed by ballooning, septation, and valve formation giving rise to a four-chambered heart in avians and mammals. The looping phase plays a central role in heart development. Successful looping is essential for proper alignment of the future cardiac chambers and tracts. As aberrant looping results in various congenital heart diseases, the mechanisms of cardiac looping have been studied for several decades by various disciplines. Many groups have studied anatomy, biology, genetics, and mechanical processes during heart looping, and have proposed multiple mechanisms. Computational modeling approaches have been utilized to examine the proposed mechanisms of the looping process. Still, the exact underlying mechanism(s) controlling the looping phase remain poorly understood. Although further experimental measurements are obviously still required, the need for more integrative computational modeling approaches is also apparent in order to make sense of the vast amount of experimental data and the complexity of multiscale developmental systems. Indeed, there needs to be an iterative interaction between experimentation and modeling in order to properly find the gap in the existing data and to validate proposed hypotheses. This article is categorized under: Cardiovascular Diseases > Genetics/Genomics/Epigenetics Cardiovascular Diseases > Computational Models Cardiovascular Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Nazanin Ebrahimi
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Christopher Bradley
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Peter Hunter
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
30
|
Abstract
The application of next-generation sequencing to study congenital heart disease (CHD) is increasingly providing new insights into the causes and mechanisms of this prevalent birth anomaly. Whole-exome sequencing analysis identifies damaging gene variants altering single or contiguous nucleotides that are assigned pathogenicity based on statistical analyses of families and cohorts with CHD, high expression in the developing heart and depletion of damaging protein-coding variants in the general population. Gene classes fulfilling these criteria are enriched in patients with CHD and extracardiac abnormalities, evidencing shared pathways in organogenesis. Developmental single-cell transcriptomic data demonstrate the expression of CHD-associated genes in particular cell lineages, and emerging insights indicate that genetic variants perturb multicellular interactions that are crucial for cardiogenesis. Whole-genome sequencing analyses extend these observations, identifying non-coding variants that influence the expression of genes associated with CHD and contribute to the estimated ~55% of unexplained cases of CHD. These approaches combined with the assessment of common and mosaic genetic variants have provided a more complete knowledge of the causes and mechanisms of CHD. Such advances provide knowledge to inform the clinical care of patients with CHD or other birth defects and deepen our understanding of the complexity of human development. In this Review, we highlight known and candidate CHD-associated human genes and discuss how the integration of advances in developmental biology research can provide new insights into the genetic contributions to CHD.
Collapse
Affiliation(s)
- Sarah U Morton
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Daniel Quiat
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | | | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Howard Hughes Medical Institute, Harvard University, Boston, MA, USA.
| |
Collapse
|
31
|
Meuser M, Deuper L, Rudat C, Aydoğdu N, Thiesler H, Zarnovican P, Hildebrandt H, Trowe MO, Kispert A. FGFR2 signaling enhances the SHH-BMP4 signaling axis in early ureter development. Development 2022; 149:273983. [DOI: 10.1242/dev.200021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/09/2021] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The patterned array of basal, intermediate and superficial cells in the urothelium of the mature ureter arises from uncommitted epithelial progenitors of the distal ureteric bud. Urothelial development requires signaling input from surrounding mesenchymal cells, which, in turn, depend on cues from the epithelial primordium to form a layered fibro-muscular wall. Here, we have identified FGFR2 as a crucial component in this reciprocal signaling crosstalk in the murine ureter. Loss of Fgfr2 in the ureteric epithelium led to reduced proliferation, stratification, intermediate and basal cell differentiation in this tissue, and affected cell survival and smooth muscle cell differentiation in the surrounding mesenchyme. Loss of Fgfr2 impacted negatively on epithelial expression of Shh and its mesenchymal effector gene Bmp4. Activation of SHH or BMP4 signaling largely rescued the cellular defects of mutant ureters in explant cultures. Conversely, inhibition of SHH or BMP signaling in wild-type ureters recapitulated the mutant phenotype in a dose-dependent manner. Our study suggests that FGF signals from the mesenchyme enhance, via epithelial FGFR2, the SHH-BMP4 signaling axis to drive urothelial and mesenchymal development in the early ureter.
Collapse
Affiliation(s)
- Max Meuser
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Lena Deuper
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Carsten Rudat
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Nurullah Aydoğdu
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Hauke Thiesler
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Patricia Zarnovican
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| |
Collapse
|
32
|
Stutt N, Song M, Wilson MD, Scott IC. Cardiac specification during gastrulation - The Yellow Brick Road leading to Tinman. Semin Cell Dev Biol 2021; 127:46-58. [PMID: 34865988 DOI: 10.1016/j.semcdb.2021.11.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
The question of how the heart develops, and the genetic networks governing this process have become intense areas of research over the past several decades. This research is propelled by classical developmental studies and potential clinical applications to understand and treat congenital conditions in which cardiac development is disrupted. Discovery of the tinman gene in Drosophila, and examination of its vertebrate homolog Nkx2.5, along with other core cardiac transcription factors has revealed how cardiac progenitor differentiation and maturation drives heart development. Careful observation of cardiac morphogenesis along with lineage tracing approaches indicated that cardiac progenitors can be divided into two broad classes of cells, namely the first and second heart fields, that contribute to the heart in two distinct waves of differentiation. Ample evidence suggests that the fate of individual cardiac progenitors is restricted to distinct cardiac structures quite early in development, well before the expression of canonical cardiac progenitor markers like Nkx2.5. Here we review the initial specification of cardiac progenitors, discuss evidence for the early patterning of cardiac progenitors during gastrulation, and consider how early gene expression programs and epigenetic patterns can direct their development. A complete understanding of when and how the developmental potential of cardiac progenitors is determined, and their potential plasticity, is of great interest developmentally and also has important implications for both the study of congenital heart disease and therapeutic approaches based on cardiac stem cell programming.
Collapse
Affiliation(s)
- Nathan Stutt
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Mengyi Song
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Michael D Wilson
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Ian C Scott
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada.
| |
Collapse
|
33
|
Waight MC, Li AC, Leung LW, Wiles BM, Thomas GR, Gallagher MM, Behr ER, Sohal M, Restrepo AJ, Saba MM. Hourly variability in outflow tract ectopy as a predictor of its site of origin. J Cardiovasc Electrophysiol 2021; 33:7-16. [PMID: 34797600 DOI: 10.1111/jce.15295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/17/2021] [Accepted: 10/16/2021] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Before ablation, predicting the site of origin (SOO) of outflow tract ventricular arrhythmia (OTVA), can inform patient consent and facilitate appropriate procedural planning. We set out to determine if OTVA variability can accurately predict SOO. METHODS Consecutive patients with a clear SOO identified at OTVA ablation had their prior 24-h ambulatory ECGs retrospectively analysed (derivation cohort). Percentage ventricular ectopic (VE) burden, hourly VE values, episodes of trigeminy/bigeminy, and the variability in these parameters were evaluated for their ability to distinguish right from left-sided SOO. Effective parameters were then prospectively tested on a validation cohort of consecutive patients undergoing their first OTVA ablation. RESULTS High VE variability (coefficient of variation ≥0.7) and the presence of any hour with <50 VE, were found to accurately predict RVOT SOO in a derivation cohort of 40 patients. In a validation cohort of 29 patients, the correct SOO was prospectively identified in 23/29 patients (79.3%) using CoV, and 26/29 patients (89.7%) using VE < 50. Including current ECG algorithms, VE < 50 had the highest Youden Index (78), the highest positive predictive value (95.0%) and the highest negative predictive value (77.8%). CONCLUSION VE variability and the presence of a single hour where VE < 50 can be used to accurately predict SOO in patients with OTVA. Accuracy of these parameters compares favorably to existing ECG algorithms.
Collapse
Affiliation(s)
| | - Anthony C Li
- St George's University of London, Cranmer Terrace, London, UK.,St George's University Hospitals NHS Foundation Trust, London, UK
| | - Lisa W Leung
- St George's University of London, Cranmer Terrace, London, UK
| | - Benedict M Wiles
- St George's University Hospitals NHS Foundation Trust, London, UK
| | - Gareth R Thomas
- St George's University Hospitals NHS Foundation Trust, London, UK
| | - Mark M Gallagher
- St George's University Hospitals NHS Foundation Trust, London, UK
| | - Elijah R Behr
- St George's University of London, Cranmer Terrace, London, UK.,St George's University Hospitals NHS Foundation Trust, London, UK
| | - Manav Sohal
- St George's University Hospitals NHS Foundation Trust, London, UK
| | | | - Magdi M Saba
- St George's University of London, Cranmer Terrace, London, UK.,St George's University Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
34
|
Nomaru H, Liu Y, De Bono C, Righelli D, Cirino A, Wang W, Song H, Racedo SE, Dantas AG, Zhang L, Cai CL, Angelini C, Christiaen L, Kelly RG, Baldini A, Zheng D, Morrow BE. 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: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
Affiliation(s)
- Hiroko Nomaru
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yang Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Dario Righelli
- Institute for Applied Computing, National Research Council, Naples, Italy
- Department of Statistical Sciences, University of Padova, Padova, Italy
| | - Andrea Cirino
- Department of Molecular Medicine and Medical Biotechnology, University Federico II School of Medicine, Naples, Italy
- Institute of Genetics and Biophysics, National Research Council, Naples, Italy
| | - Wei Wang
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, USA
| | - Hansoo Song
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Silvia E Racedo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anelisa G Dantas
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Lu Zhang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chen-Leng Cai
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Claudia Angelini
- Institute for Applied Computing, National Research Council, Naples, Italy
| | - Lionel Christiaen
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, USA
| | - Robert G Kelly
- Aix-Marseille University, CNRS UMR 7288, IBDM, Marseille, France
| | - Antonio Baldini
- Department of Molecular Medicine and Medical Biotechnology, University Federico II School of Medicine, Naples, Italy
- Institute of Genetics and Biophysics, National Research Council, Naples, Italy
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
35
|
Lescroart F, Dumas CE, Adachi N, Kelly RG. Emergence of heart and branchiomeric muscles in cardiopharyngeal mesoderm. Exp Cell Res 2021; 410:112931. [PMID: 34798131 DOI: 10.1016/j.yexcr.2021.112931] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/27/2021] [Accepted: 11/14/2021] [Indexed: 12/17/2022]
Abstract
Branchiomeric muscles of the head and neck originate in a population of cranial mesoderm termed cardiopharyngeal mesoderm that also contains progenitor cells contributing to growth of the embryonic heart. Retrospective lineage analysis has shown that branchiomeric muscles share a clonal origin with parts of the heart, indicating the presence of common heart and head muscle progenitor cells in the early embryo. Genetic lineage tracing and functional studies in the mouse, as well as in Ciona and zebrafish, together with recent experiments using single cell transcriptomics and multipotent stem cells, have provided further support for the existence of bipotent head and heart muscle progenitor cells. Current challenges concern defining where and when such common progenitor cells exist in mammalian embryos and how alternative myogenic derivatives emerge in cardiopharyngeal mesoderm. Addressing these questions will provide insights into mechanisms of cell fate acquisition and the evolution of vertebrate musculature, as well as clinical insights into the origins of muscle restricted myopathies and congenital defects affecting craniofacial and cardiac development.
Collapse
Affiliation(s)
| | - Camille E Dumas
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009, Marseille, France
| | - Noritaka Adachi
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009, Marseille, France
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009, Marseille, France.
| |
Collapse
|
36
|
Shi L, Racedo SE, Diacou A, Park T, Zhou B, Morrow BE. Crk and Crkl have shared functions in neural crest cells for cardiac outflow tract septation and vascular smooth muscle differentiation. Hum Mol Genet 2021; 31:1197-1215. [PMID: 34686881 PMCID: PMC9029238 DOI: 10.1093/hmg/ddab313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/05/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
CRK and CRKL encode cytoplasmic adaptors that contribute to the etiology of congenital heart disease. Neural crest cells (NCCs) are required for cardiac outflow tract (OFT) septation and aortic arch formation. The roles of Crk/Crkl in NCCs during mouse cardiovascular development remains unknown. To test this, we inactivated Crk and/or Crkl in NCCs. We found that the loss of Crk, rather than Crkl, in NCCs resulted in double outlet right ventricle, while loss of both Crk/Crkl in NCCs resulted in severe defects with earlier lethality due to failed OFT septation and severe dilation of the pharyngeal arch arteries (PAAs). We found that these defects are due to altered cell morphology resulting in reduced localization of NCCs to the OFT and failed integrity of the PAAs, along with reduced expression of Integrin signaling genes. Further, molecular studies identified reduced differentiation of vascular smooth muscle cells that may in part be due to altered Notch signaling. Additionally, there is increased cellular stress that leads to modest increase in apoptosis. Overall, this explains the mechanism for the Crk/Crkl phenotype.
Collapse
Affiliation(s)
- Lijie Shi
- Department of Genetics, Albert Einstein college of Medicine, Bronx, NY, USA
| | - Silvia E Racedo
- Department of Genetics, Albert Einstein college of Medicine, Bronx, NY, USA
| | - Alexander Diacou
- Department of Genetics, Albert Einstein college of Medicine, Bronx, NY, USA
| | - Taeju Park
- Department of Pediatrics, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Bin Zhou
- Department of Genetics, Albert Einstein college of Medicine, Bronx, NY, USA
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein college of Medicine, Bronx, NY, USA
| |
Collapse
|
37
|
Derrick CJ, Pollitt EJG, Sanchez Sevilla Uruchurtu A, Hussein F, Grierson AJ, Noël ES. Lamb1a regulates atrial growth by limiting second heart field addition during zebrafish heart development. Development 2021; 148:272294. [PMID: 34568948 DOI: 10.1242/dev.199691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 09/19/2021] [Indexed: 12/20/2022]
Abstract
During early vertebrate heart development, the heart transitions from a linear tube to a complex asymmetric structure, a morphogenetic process that occurs simultaneously with growth of the heart. Cardiac growth during early heart morphogenesis is driven by deployment of cells from the second heart field (SHF) into both poles of the heart. Laminin is a core component of the extracellular matrix and, although mutations in laminin subunits are linked with cardiac abnormalities, no role for laminin has been identified in early vertebrate heart morphogenesis. We identified tissue-specific expression of laminin genes in the developing zebrafish heart, supporting a role for laminins in heart morphogenesis. Analysis of heart development in lamb1a zebrafish mutant embryos reveals mild morphogenetic defects and progressive cardiomegaly, and that Lamb1a functions to limit heart size during cardiac development by restricting SHF addition. lamb1a mutants exhibit hallmarks of altered haemodynamics, and blocking cardiac contractility in lamb1a mutants rescues heart size and atrial SHF addition. Together, these results suggest that laminin mediates interactions between SHF deployment and cardiac biomechanics during heart morphogenesis and growth in the developing embryo.
Collapse
Affiliation(s)
| | - Eric J G Pollitt
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | | | - Farah Hussein
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Andrew J Grierson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Emily S Noël
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
38
|
Rowton M, Guzzetta A, Rydeen AB, Moskowitz IP. Control of cardiomyocyte differentiation timing by intercellular signaling pathways. Semin Cell Dev Biol 2021; 118:94-106. [PMID: 34144893 PMCID: PMC8968240 DOI: 10.1016/j.semcdb.2021.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/19/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023]
Abstract
Congenital Heart Disease (CHD), malformations of the heart present at birth, is the most common class of life-threatening birth defect (Hoffman (1995) [1], Gelb (2004) [2], Gelb (2014) [3]). A major research challenge is to elucidate the genetic determinants of CHD and mechanistically link CHD ontogeny to a molecular understanding of heart development. Although the embryonic origins of CHD are unclear in most cases, dysregulation of cardiovascular lineage specification, patterning, proliferation, migration or differentiation have been described (Olson (2004) [4], Olson (2006) [5], Srivastava (2006) [6], Dunwoodie (2007) [7], Bruneau (2008) [8]). Cardiac differentiation is the process whereby cells become progressively more dedicated in a trajectory through the cardiac lineage towards mature cardiomyocytes. Defects in cardiac differentiation have been linked to CHD, although how the complex control of cardiac differentiation prevents CHD is just beginning to be understood. The stages of cardiac differentiation are highly stereotyped and have been well-characterized (Kattman et al. (2011) [9], Wamstad et al. (2012) [10], Luna-Zurita et al. (2016) [11], Loh et al. (2016) [12], DeLaughter et al. (2016) [13]); however, the developmental and molecular mechanisms that promote or delay the transition of a cell through these stages have not been as deeply investigated. Tight temporal control of progenitor differentiation is critically important for normal organ size, spatial organization, and cellular physiology and homeostasis of all organ systems (Raff et al. (1985) [14], Amthor et al. (1998) [15], Kopan et al. (2014) [16]). This review will focus on the action of signaling pathways in the control of cardiomyocyte differentiation timing. Numerous signaling pathways, including the Wnt, Fibroblast Growth Factor, Hedgehog, Bone Morphogenetic Protein, Insulin-like Growth Factor, Thyroid Hormone and Hippo pathways, have all been implicated in promoting or inhibiting transitions along the cardiac differentiation trajectory. Gaining a deeper understanding of the mechanisms controlling cardiac differentiation timing promises to yield insights into the etiology of CHD and to inform approaches to restore function to damaged hearts.
Collapse
|
39
|
Lewis-Israeli YR, Wasserman AH, Aguirre A. Heart Organoids and Engineered Heart Tissues: Novel Tools for Modeling Human Cardiac Biology and Disease. Biomolecules 2021; 11:1277. [PMID: 34572490 PMCID: PMC8468189 DOI: 10.3390/biom11091277] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/06/2021] [Accepted: 08/24/2021] [Indexed: 01/02/2023] Open
Abstract
Organoids are three-dimensional in vitro cell constructs that recapitulate organ properties and structure to a significant extent. They constitute particularly useful models to study unapproachable states in humans, such as embryonic and fetal development, or early disease progression in adults. In recent years organoids have been implemented to model a wide range of different organs and disease conditions. However, the technology for their fabrication and application to cardiovascular studies has been lagging significantly when compared to other organoid types (e.g., brain, pancreas, kidney, intestine). This is a surprising fact since cardiovascular disease (CVD) and congenital heart disease (CHD) constitute the leading cause of mortality and morbidity in the developed world, and the most common birth defect in humans, respectively, and collectively constitute one of the largest unmet medical needs in the modern world. There is a critical need to establish in vitro models of the human heart that faithfully recapitulate its biology and function, thus enabling basic and translational studies to develop new therapeutics. Generating heart organoids that truly resemble the heart has proven difficult due to its complexity, but significant progress has been made recently to overcome this obstacle. In this review, we will discuss progress in novel heart organoid generation methods, the advantages and disadvantages of each approach, and their translational applications for advancing cardiovascular studies and the treatment of heart disorders.
Collapse
Affiliation(s)
- Yonatan R. Lewis-Israeli
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48823, USA; (Y.R.L.-I.); (A.H.W.)
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48823, USA
| | - Aaron H. Wasserman
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48823, USA; (Y.R.L.-I.); (A.H.W.)
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48823, USA
| | - Aitor Aguirre
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48823, USA; (Y.R.L.-I.); (A.H.W.)
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48823, USA
| |
Collapse
|
40
|
The Spatiotemporal Expression of Notch1 and Numb and Their Functional Interaction during Cardiac Morphogenesis. Cells 2021; 10:cells10092192. [PMID: 34571841 PMCID: PMC8471136 DOI: 10.3390/cells10092192] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/18/2021] [Accepted: 08/21/2021] [Indexed: 12/13/2022] Open
Abstract
Numb family proteins (NFPs), including Numb and Numblike (Numbl), are commonly known for their role as cell fate determinants for multiple types of progenitor cells, mainly due to their function as Notch inhibitors. Previous studies have shown that myocardial NFP double knockout (MDKO) hearts display an up-regulated Notch activation and various defects in cardiac progenitor cell differentiation and cardiac morphogenesis. Whether enhanced Notch activation causes these defects in MDKO is not fully clear. To answer the question, we examined the spatiotemporal patterns of Notch1 expression, Notch activation, and Numb expression in the murine embryonic hearts using multiple approaches including RNAScope, and Numb and Notch reporter mouse lines. To further interrogate the interaction between NFPs and Notch signaling activation, we deleted both Notch1 or RBPJk alleles in the MDKO. We examined and compared the phenotypes of Notch1 knockout, NFPs double knockout, Notch1; Numb; Numbl and RBPJk; Numb; Numbl triple knockouts. Our study showed that Notch1 is expressed and activated in the myocardium at several stages, and Numb is enriched in the epicardium and did not show the asymmetric distribution in the myocardium. Cardiac-specific Notch1 deletion causes multiple structural defects and embryonic lethality. Notch1 or RBPJk deletion in MDKO did not rescue the structural defects in the MDKO but partially rescued the defects of cardiac progenitor cell differentiation, cardiomyocyte proliferation, and trabecular morphogenesis. Our study concludes that NFPs regulate progenitor cell differentiation, cardiomyocyte proliferation, and trabecular morphogenesis partially through Notch1 and play more roles than inhibiting Notch1 signaling during cardiac morphogenesis.
Collapse
|
41
|
Matos-Nieves A, Manivannan S, Majumdar U, McBride KL, White P, Garg V. A Multi-Omics Approach Using a Mouse Model of Cardiac Malformations for Prioritization of Human Congenital Heart Disease Contributing Genes. Front Cardiovasc Med 2021; 8:683074. [PMID: 34504875 PMCID: PMC8421733 DOI: 10.3389/fcvm.2021.683074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/22/2021] [Indexed: 01/22/2023] Open
Abstract
Congenital heart disease (CHD) is the most common type of birth defect, affecting ~1% of all live births. Malformations of the cardiac outflow tract (OFT) account for ~30% of all CHD and include a range of CHDs from bicuspid aortic valve (BAV) to tetralogy of Fallot (TOF). We hypothesized that transcriptomic profiling of a mouse model of CHD would highlight disease-contributing genes implicated in congenital cardiac malformations in humans. To test this hypothesis, we utilized global transcriptional profiling differences from a mouse model of OFT malformations to prioritize damaging, de novo variants identified from exome sequencing datasets from published cohorts of CHD patients. Notch1 +/- ; Nos3 -/- mice display a spectrum of cardiac OFT malformations ranging from BAV, semilunar valve (SLV) stenosis to TOF. Global transcriptional profiling of the E13.5 Notch1 +/- ; Nos3 -/- mutant mouse OFTs and wildtype controls was performed by RNA sequencing (RNA-Seq). Analysis of the RNA-Seq dataset demonstrated genes belonging to the Hif1α, Tgf-β, Hippo, and Wnt signaling pathways were differentially expressed in the mutant OFT. Mouse to human comparative analysis was then performed to determine if patients with TOF and SLV stenosis display an increased burden of damaging, genetic variants in gene homologs that were dysregulated in Notch1 +/- ; Nos3 -/- OFT. We found an enrichment of de novo variants in the TOF population among the 1,352 significantly differentially expressed genes in Notch1 +/- ; Nos3 -/- mouse OFT but not the SLV population. This association was not significant when comparing only highly expressed genes in the murine OFT to de novo variants in the TOF population. These results suggest that transcriptomic datasets generated from the appropriate temporal, anatomic and cellular tissues from murine models of CHD may provide a novel approach for the prioritization of disease-contributing genes in patients with CHD.
Collapse
Affiliation(s)
- Adrianna Matos-Nieves
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
| | - Sathiyanarayanan Manivannan
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
| | - Uddalak Majumdar
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
| | - Kim L. McBride
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, Ohio State University, Columbus, OH, United States
| | - Peter White
- Department of Pediatrics, Ohio State University, Columbus, OH, United States
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Vidu Garg
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, Ohio State University, Columbus, OH, United States
- Department of Molecular Genetics, Ohio State University, Columbus, OH, United States
| |
Collapse
|
42
|
Kern CB. Excess Provisional Extracellular Matrix: A Common Factor in Bicuspid Aortic Valve Formation. J Cardiovasc Dev Dis 2021; 8:92. [PMID: 34436234 PMCID: PMC8396938 DOI: 10.3390/jcdd8080092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 02/07/2023] Open
Abstract
A bicuspid aortic valve (BAV) is the most common cardiac malformation, found in 0.5% to 2% of the population. BAVs are present in approximately 50% of patients with severe aortic stenosis and are an independent risk factor for aortic aneurysms. Currently, there are no therapeutics to treat BAV, and the human mutations identified to date represent a relatively small number of BAV patients. However, the discovery of BAV in an increasing number of genetically modified mice is advancing our understanding of molecular pathways that contribute to BAV formation. In this study, we utilized the comparison of BAV phenotypic characteristics between murine models as a tool to advance our understanding of BAV formation. The collation of murine BAV data indicated that excess versican within the provisional extracellular matrix (P-ECM) is a common factor in BAV development. While the percentage of BAVs is low in many of the murine BAV models, the remaining mutant mice exhibit larger and more amorphous tricuspid AoVs, also with excess P-ECM compared to littermates. The identification of common molecular characteristics among murine BAV models may lead to BAV therapeutic targets and biomarkers of disease progression for this highly prevalent and heterogeneous cardiovascular malformation.
Collapse
Affiliation(s)
- Christine B Kern
- Department of Regenerative Medicine and Cell Biology, 171 Ashley Avenue, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
43
|
Wang Y, Fang Y, Lu P, Wu B, Zhou B. NOTCH Signaling in Aortic Valve Development and Calcific Aortic Valve Disease. Front Cardiovasc Med 2021; 8:682298. [PMID: 34239905 PMCID: PMC8259786 DOI: 10.3389/fcvm.2021.682298] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/14/2021] [Indexed: 01/05/2023] Open
Abstract
NOTCH intercellular signaling mediates the communications between adjacent cells involved in multiple biological processes essential for tissue morphogenesis and homeostasis. The NOTCH1 mutations are the first identified human genetic variants that cause congenital bicuspid aortic valve (BAV) and calcific aortic valve disease (CAVD). Genetic variants affecting other genes in the NOTCH signaling pathway may also contribute to the development of BAV and the pathogenesis of CAVD. While CAVD occurs commonly in the elderly population with tri-leaflet aortic valve, patients with BAV have a high risk of developing CAVD at a young age. This observation indicates an important role of NOTCH signaling in the postnatal homeostasis of the aortic valve, in addition to its prenatal functions during aortic valve development. Over the last decade, animal studies, especially with the mouse models, have revealed detailed information in the developmental etiology of congenital aortic valve defects. In this review, we will discuss the molecular and cellular aspects of aortic valve development and examine the embryonic pathogenesis of BAV. We will focus our discussions on the NOTCH signaling during the endocardial-to-mesenchymal transformation (EMT) and the post-EMT remodeling of the aortic valve. We will further examine the involvement of the NOTCH mutations in the postnatal development of CAVD. We will emphasize the deleterious impact of the embryonic valve defects on the homeostatic mechanisms of the adult aortic valve for the purpose of identifying the potential therapeutic targets for disease intervention.
Collapse
Affiliation(s)
- Yidong Wang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yuan Fang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Bin Zhou
- Departments of Genetics, Pediatrics (Pediatric Genetic Medicine), and Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States
- The Einstein Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
44
|
Kodo K, Uchida K, Yamagishi H. Genetic and Cellular Interaction During Cardiovascular Development Implicated in Congenital Heart Diseases. Front Cardiovasc Med 2021; 8:653244. [PMID: 33796576 PMCID: PMC8007765 DOI: 10.3389/fcvm.2021.653244] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/22/2021] [Indexed: 12/21/2022] Open
Abstract
Congenital heart disease (CHD) is the most common life-threatening congenital anomaly. CHD occurs due to defects in cardiovascular development, and the majority of CHDs are caused by a multifactorial inheritance mechanism, which refers to the interaction between genetic and environmental factors. During embryogenesis, the cardiovascular system is derived from at least four distinct cell lineages: the first heart field, second heart field, cardiac neural crest, and proepicardial organ. Understanding the genes involved in each lineage is essential to uncover the genomic architecture of CHD. Therefore, we provide an overview of recent research progress using animal models and mutation analyses to better understand the molecular mechanisms and pathways linking cardiovascular development and CHD. For example, we highlight our recent work on genes encoding three isoforms of inositol 1,4,5-trisphosphate receptors (IP3R1, 2, and 3) that regulate various vital and developmental processes, which have genetic redundancy during cardiovascular development. Specifically, IP3R1 and 2 have redundant roles in the atrioventricular cushion derived from the first heart field lineage, whereas IP3R1 and 3 exhibit redundancy in the right ventricle and the outflow tract derived from the second heart field lineage, respectively. Moreover, 22q11.2 deletion syndrome (22q11DS) is highly associated with CHD involving the outflow tract, characterized by defects of the cardiac neural crest lineage. However, our studies have shown that TBX1, a major genetic determinant of 22q11DS, was not expressed in the cardiac neural crest but rather in the second heart field, suggesting the importance of the cellular interaction between the cardiac neural crest and the second heart field. Comprehensive genetic analysis using the Japanese genome bank of CHD and mouse models revealed that a molecular regulatory network involving GATA6, FOXC1/2, TBX1, SEMA3C, and FGF8 was essential for reciprocal signaling between the cardiac neural crest and the second heart field during cardiovascular development. Elucidation of the genomic architecture of CHD using induced pluripotent stem cells and next-generation sequencing technology, in addition to genetically modified animal models and human mutation analyses, would facilitate the development of regenerative medicine and/or preventive medicine for CHD in the near future.
Collapse
Affiliation(s)
- Kazuki Kodo
- Division of Pediatric Cardiology, Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Keiko Uchida
- Division of Pediatric Cardiology, Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Yamagishi
- Division of Pediatric Cardiology, Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
45
|
Yahya I, Al Haj A, Brand-Saberi B, Morosan-Puopolo G. Chicken Second Branchial Arch Progenitor Cells Contribute to Heart Musculature in vitro and in vivo. Cells Tissues Organs 2021; 209:165-176. [PMID: 33423027 DOI: 10.1159/000511686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/15/2020] [Indexed: 11/19/2022] Open
Abstract
In the past, the heart muscle was thought to originate from a single source of myocardial progenitor cells. More recently, however, an additional source of myocardial progenitors has been revealed to be the second heart field, and chicken embryos were important for establishing this concept. However, there have been few studies in chicken on how this field contributes to heart muscles in vitro. We have developed an ex vivo experimental system from chicken embryos between stages HH17-20 to investigate how mesodermal progenitors in the second branchial arch (BA2) differentiate into cardiac muscles. Using this method, we presented evidence that the progenitor cells within the BA2 arch differentiated into beating cardiomyocytes in vitro. The beating explant cells were positive for cardiac actin, Nkx2.5, and ventricular myosin heavy chain. In addition, we performed a time course for the expression of second heart field markers (Isl1 and Nkx2.5) in the BA2 from stage HH16 to stage HH21 using in situ hybridization. Accordingly, using EGFP-based cell labeling techniques and quail-chicken cell injection, we demonstrated that mesodermal cells from the BA2 contributed to the outflow tract and ventricular myocardium in vivo. Thus, our findings highlight the cardiogenic potential of chicken BA2 mesodermal cells in vitro and in vivo.
Collapse
Affiliation(s)
- Imadeldin Yahya
- Institute of Anatomy, Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany.,Department of Anatomy, Faculty of Veterinary Medicine, Khartoum University, Khartoum, Sudan
| | - Abdulatif Al Haj
- Institute of Anatomy, Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| | - Beate Brand-Saberi
- Institute of Anatomy, Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| | - Gabriela Morosan-Puopolo
- Institute of Anatomy, Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany,
| |
Collapse
|
46
|
Mef2c factors are required for early but not late addition of cardiomyocytes to the ventricle. Dev Biol 2020; 470:95-107. [PMID: 33245870 PMCID: PMC7819464 DOI: 10.1016/j.ydbio.2020.11.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022]
Abstract
During heart formation, the heart grows and undergoes dramatic morphogenesis to achieve efficient embryonic function. Both in fish and amniotes, much of the growth occurring after initial heart tube formation arises from second heart field (SHF)-derived progenitor cell addition to the arterial pole, allowing chamber formation. In zebrafish, this process has been extensively studied during embryonic life, but it is unclear how larval cardiac growth occurs beyond 3 days post-fertilisation (dpf). By quantifying zebrafish myocardial growth using live imaging of GFP-labelled myocardium we show that the heart grows extensively between 3 and 5 dpf. Using methods to assess cell division, cellular development timing assay and Kaede photoconversion, we demonstrate that proliferation, CM addition, and hypertrophy contribute to ventricle growth. Mechanistically, we show that reduction in Mef2c activity (mef2ca+/-;mef2cb-/-), downstream or in parallel with Nkx2.5 and upstream of Ltbp3, prevents some CM addition and differentiation, resulting in a significantly smaller ventricle by 3 dpf. After 3 dpf, however, CM addition in mef2ca+/-;mef2cb-/- mutants recovers to a normal pace, and the heart size gap between mutants and their siblings diminishes into adulthood. Thus, as in mice, there is an early time window when SHF contribution to the myocardium is particularly sensitive to loss of Mef2c activity.
Collapse
|
47
|
van den Hoff MJB, Wessels A. Muscularization of the Mesenchymal Outlet Septum during Cardiac Development. J Cardiovasc Dev Dis 2020; 7:jcdd7040051. [PMID: 33158304 PMCID: PMC7711588 DOI: 10.3390/jcdd7040051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 12/11/2022] Open
Abstract
After the formation of the linear heart tube, it becomes divided into right and left components by the process of septation. Relatively late during this process, within the developing outflow tract, the initially mesenchymal outlet septum becomes muscularized as the result of myocardialization. Myocardialization is defined as the process in which existing cardiomyocytes migrate into flanking mesenchyme. Studies using genetically modified mice, as well as experimental approaches using in vitro models, demonstrate that Wnt and TGFβ signaling play an essential role in the regulation of myocardialization. They also show the significance of the interaction between cardiomyocytes, endocardial derived cells, neural crest cells, and the extracellular matrix. Interestingly, Wnt-mediated non-canonical planar cell polarity signaling was found to be a crucial regulator of myocardialization in the outlet septum and Wnt-mediated canonical β-catenin signaling is an essential regulator of the expansion of mesenchymal cells populating the outflow tract cushions.
Collapse
Affiliation(s)
- Maurice J. B. van den Hoff
- Department of Medical Biology, AmsterdamUMC, Location AMC, 1105AZ Amsterdam, The Netherlands
- Correspondence: ; Tel.: +1-3120-5665-405
| | - Andy Wessels
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA;
| |
Collapse
|
48
|
Deepe R, Fitzgerald E, Wolters R, Drummond J, Guzman KD, van den Hoff MJ, Wessels A. The Mesenchymal Cap of the Atrial Septum and Atrial and Atrioventricular Septation. J Cardiovasc Dev Dis 2020; 7:jcdd7040050. [PMID: 33158164 PMCID: PMC7712865 DOI: 10.3390/jcdd7040050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/28/2020] [Accepted: 11/02/2020] [Indexed: 12/26/2022] Open
Abstract
In this publication, dedicated to Professor Robert H. Anderson and his contributions to the field of cardiac development, anatomy, and congenital heart disease, we will review some of our earlier collaborative studies. The focus of this paper is on our work on the development of the atrioventricular mesenchymal complex, studies in which Professor Anderson has played a significant role. We will revisit a number of events relevant to atrial and atrioventricular septation and present new data on the development of the mesenchymal cap of the atrial septum, a component of the atrioventricular mesenchymal complex which, thus far, has received only moderate attention.
Collapse
Affiliation(s)
- Ray Deepe
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (R.D.); (E.F.); (R.W.); (J.D.); (K.D.G.)
| | - Emily Fitzgerald
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (R.D.); (E.F.); (R.W.); (J.D.); (K.D.G.)
| | - Renélyn Wolters
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (R.D.); (E.F.); (R.W.); (J.D.); (K.D.G.)
| | - Jenna Drummond
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (R.D.); (E.F.); (R.W.); (J.D.); (K.D.G.)
| | - Karen De Guzman
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (R.D.); (E.F.); (R.W.); (J.D.); (K.D.G.)
| | - Maurice J.B. van den Hoff
- Amsterdam UMC, Academic Medical Center, Department of Medical Biology, Meibergdreef 15, 1105AZ Amsterdam, The Netherlands;
| | - Andy Wessels
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (R.D.); (E.F.); (R.W.); (J.D.); (K.D.G.)
- Correspondence: ; Tel.: +1-843-792-8183
| |
Collapse
|
49
|
Boezio GL, Bensimon-Brito A, Piesker J, Guenther S, Helker CS, Stainier DY. Endothelial TGF-β signaling instructs smooth muscle cell development in the cardiac outflow tract. eLife 2020; 9:57603. [PMID: 32990594 PMCID: PMC7524555 DOI: 10.7554/elife.57603] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022] Open
Abstract
The development of the cardiac outflow tract (OFT), which connects the heart to the great arteries, relies on a complex crosstalk between endothelial (ECs) and smooth muscle (SMCs) cells. Defects in OFT development can lead to severe malformations, including aortic aneurysms, which are frequently associated with impaired TGF-β signaling. To better understand the role of TGF-β signaling in OFT formation, we generated zebrafish lacking the TGF-β receptor Alk5 and found a strikingly specific dilation of the OFT: alk5-/- OFTs exhibit increased EC numbers as well as extracellular matrix (ECM) and SMC disorganization. Surprisingly, endothelial-specific alk5 overexpression in alk5-/- rescues the EC, ECM, and SMC defects. Transcriptomic analyses reveal downregulation of the ECM gene fibulin-5, which when overexpressed in ECs ameliorates OFT morphology and function. These findings reveal a new requirement for endothelial TGF-β signaling in OFT morphogenesis and suggest an important role for the endothelium in the etiology of aortic malformations.
Collapse
Affiliation(s)
- Giulia Lm Boezio
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Anabela Bensimon-Brito
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Janett Piesker
- Scientific Service Group Microscopy, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Guenther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Christian Sm Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Didier Yr Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
50
|
De Zoysa P, Liu J, Toubat O, Choi J, Moon A, Gill PS, Duarte A, Sucov HM, Kumar SR. Delta-like ligand 4-mediated Notch signaling controls proliferation of second heart field progenitor cells by regulating Fgf8 expression. Development 2020; 147:dev185249. [PMID: 32778568 PMCID: PMC7502602 DOI: 10.1242/dev.185249] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 07/27/2020] [Indexed: 12/15/2022]
Abstract
The role played by the Notch pathway in cardiac progenitor cell biology remains to be elucidated. Delta-like ligand 4 (Dll4), the arterial-specific Notch ligand, is expressed by second heart field (SHF) progenitors at time-points that are crucial in SHF biology. Dll4-mediated Notch signaling is required for maintaining an adequate pool of SHF progenitors, such that Dll4 knockout results in a reduction in proliferation and an increase in apoptosis. A reduced SHF progenitor pool leads to an underdeveloped right ventricle (RV) and outflow tract (OFT). In its most severe form, there is severe RV hypoplasia and poorly developed OFT resulting in early embryonic lethality. In its milder form, the OFT is foreshortened and misaligned, resulting in a double outlet right ventricle. Dll4-mediated Notch signaling maintains Fgf8 expression by transcriptional regulation at the promoter level. Combined heterozygous knockout of Dll4 and Fgf8 demonstrates genetic synergy in OFT alignment. Exogenous supplemental Fgf8 rescues proliferation in Dll4 mutants in ex-vivo culture. Our results establish a novel role for Dll4-mediated Notch signaling in SHF biology. More broadly, our model provides a platform for understanding oligogenic inheritance that results in clinically relevant OFT malformations.
Collapse
Affiliation(s)
- Prashan De Zoysa
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Jiang Liu
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Omar Toubat
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Jongkyu Choi
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Anne Moon
- Department of Molecular and Functional Genomics, Geisinger Clinic, PA 17822, USA
| | - Parkash S Gill
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Antonio Duarte
- Centro Interdisciplinar de Investigação em Sanidade Animal, University of Lisbon, Department of Physiology, 1300-477 Lisboa, Portugal
| | - Henry M Sucov
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29403, USA
| | - S Ram Kumar
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| |
Collapse
|