1
|
Angom RS, Singh M, Muhammad H, Varanasi SM, Mukhopadhyay D. Zebrafish as a Versatile Model for Cardiovascular Research: Peering into the Heart of the Matter. Cells 2025; 14:531. [PMID: 40214485 PMCID: PMC11988917 DOI: 10.3390/cells14070531] [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: 02/25/2025] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in the world. A total of 17.5 million people died of CVDs in the year 2012, accounting for 31% of all deaths globally. Vertebrate animal models have been used to understand cardiac disease biology, as the cellular, molecular, and physiological aspects of human CVDs can be replicated closely in these organisms. Zebrafish is a popular model organism offering an arsenal of genetic tools that allow the rapid in vivo analysis of vertebrate gene function and disease conditions. It has a short breeding cycle, high fecundity, optically transparent embryos, rapid internal organ development, and easy maintenance. This review aims to give readers an overview of zebrafish cardiac biology and a detailed account of heart development in zebrafish and its comparison with humans and the conserved genetic circuitry. We also discuss the contributions made in CVD research using the zebrafish model. The first part of this review focuses on detailed information on the morphogenetic and differentiation processes in early cardiac development. The overlap and divergence of the human heart's genetic circuitry, structure, and physiology are emphasized wherever applicable. In the second part of the review, we overview the molecular tools and techniques available to dissect gene function and expression in zebrafish, with special mention of the use of these tools in cardiac biology.
Collapse
Affiliation(s)
- Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| | - Meghna Singh
- Department of Pathology and Lab Medicine, University of California, Los Angeles, CA 92093, USA;
| | - Huzaifa Muhammad
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Sai Manasa Varanasi
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| |
Collapse
|
2
|
Albu M, Affolter E, Gentile A, Xu Y, Kikhi K, Howard S, Kuenne C, Priya R, Gunawan F, Stainier DYR. Distinct mechanisms regulate ventricular and atrial chamber wall formation. Nat Commun 2024; 15:8159. [PMID: 39289341 PMCID: PMC11408654 DOI: 10.1038/s41467-024-52340-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024] Open
Abstract
Tissues undergo distinct morphogenetic processes to achieve similarly shaped structures. In the heart, cardiomyocytes in both the ventricle and atrium build internal structures for efficient contraction. Ventricular wall formation (trabeculation) is initiated by cardiomyocyte delamination. How cardiomyocytes build the atrial wall is poorly understood. Using longitudinal imaging in zebrafish, we found that at least 25% of the atrial cardiomyocytes elongate along the long axis of the heart. These cell shape changes result in cell intercalation and convergent thickening, leading to the formation of the internal muscle network. We tested factors important for ventricular trabeculation including Nrg/ErbB and Notch signaling and found no evidence for their role in atrial muscle network formation. Instead, our data suggest that atrial cardiomyocyte elongation is regulated by Yap, which has not been implicated in trabeculation. Altogether, these data indicate that distinct cellular and molecular mechanisms build the internal muscle structures in the atrium and ventricle.
Collapse
Affiliation(s)
- Marga Albu
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Eileen Affolter
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Alessandra Gentile
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- MRC Centre for Neurodevelopmental Disorders, King's College, London, UK
| | - Yanli Xu
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Khrievono Kikhi
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Flow Cytometry Service Group, Max Planck for Heart and Lung Research, Bad Nauheim, Germany
| | - Sarah Howard
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Carsten Kuenne
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rashmi Priya
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Francis Crick Institute, London, UK
| | - Felix Gunawan
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Institute of Cell Biology, University of Münster, Münster, Germany
| | - Didier Y R Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany.
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.
| |
Collapse
|
3
|
Yang D, Jian Z, Tang C, Chen Z, Zhou Z, Zheng L, Peng X. Zebrafish Congenital Heart Disease Models: Opportunities and Challenges. Int J Mol Sci 2024; 25:5943. [PMID: 38892128 PMCID: PMC11172925 DOI: 10.3390/ijms25115943] [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: 04/14/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Congenital heart defects (CHDs) are common human birth defects. Genetic mutations potentially cause the exhibition of various pathological phenotypes associated with CHDs, occurring alone or as part of certain syndromes. Zebrafish, a model organism with a strong molecular conservation similar to humans, is commonly used in studies on cardiovascular diseases owing to its advantageous features, such as a similarity to human electrophysiology, transparent embryos and larvae for observation, and suitability for forward and reverse genetics technology, to create various economical and easily controlled zebrafish CHD models. In this review, we outline the pros and cons of zebrafish CHD models created by genetic mutations associated with single defects and syndromes and the underlying pathogenic mechanism of CHDs discovered in these models. The challenges of zebrafish CHD models generated through gene editing are also discussed, since the cardiac phenotypes resulting from a single-candidate pathological gene mutation in zebrafish might not mirror the corresponding human phenotypes. The comprehensive review of these zebrafish CHD models will facilitate the understanding of the pathogenic mechanisms of CHDs and offer new opportunities for their treatments and intervention strategies.
Collapse
|
4
|
Nakanishi-Koakutsu M, Miki K, Naka Y, Sasaki M, Wakimizu T, Napier SC, Okubo C, Narita M, Nishikawa M, Hata R, Chonabayashi K, Hotta A, Imahashi K, Nishimoto T, Yoshida Y. CD151 expression marks atrial- and ventricular- differentiation from human induced pluripotent stem cells. Commun Biol 2024; 7:231. [PMID: 38418926 PMCID: PMC10901864 DOI: 10.1038/s42003-024-05809-2] [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: 09/03/2022] [Accepted: 01/11/2024] [Indexed: 03/02/2024] Open
Abstract
Current differentiation protocols for human induced pluripotent stem cells (hiPSCs) produce heterogeneous cardiomyocytes (CMs). Although chamber-specific CM selection using cell surface antigens enhances biomedical applications, a cell surface marker that accurately distinguishes between hiPSC-derived atrial CMs (ACMs) and ventricular CMs (VCMs) has not yet been identified. We have developed an approach for obtaining functional hiPSC-ACMs and -VCMs based on CD151 expression. For ACM differentiation, we found that ACMs are enriched in the CD151low population and that CD151 expression is correlated with the expression of Notch4 and its ligands. Furthermore, Notch signaling inhibition followed by selecting the CD151low population during atrial differentiation leads to the highly efficient generation of ACMs as evidenced by gene expression and electrophysiology. In contrast, for VCM differentiation, VCMs exhibiting a ventricular-related gene signature and uniform action potentials are enriched in the CD151high population. Our findings enable the production of high-quality ACMs and VCMs appropriate for hiPSC-derived chamber-specific disease models and other applications.
Collapse
Affiliation(s)
- Misato Nakanishi-Koakutsu
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Kenji Miki
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan.
- Center for Organ Engineering, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, 02114, USA.
- Premium Research Institute for Human Metaverse Medicine, Osaka University, Suita, 565-0871, Japan.
| | - Yuki Naka
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
| | - Masako Sasaki
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
| | - Takayuki Wakimizu
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
| | - Stephanie C Napier
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
- Global Advanced Platform, Takeda Pharmaceutical Company Limited, Fujisawa, 251-8555, Japan
| | - Chikako Okubo
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Megumi Narita
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Misato Nishikawa
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Reo Hata
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Kazuhisa Chonabayashi
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
| | - Kenichi Imahashi
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
- Global Advanced Platform, Takeda Pharmaceutical Company Limited, Fujisawa, 251-8555, Japan
| | - Tomoyuki Nishimoto
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
- Orizuru Therapeutics Incorporated, Fujisawa, 251-8555, Japan
| | - Yoshinori Yoshida
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan.
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan.
| |
Collapse
|
5
|
Yao Y, Gupta D, Yelon D. The MEK-ERK signaling pathway promotes maintenance of cardiac chamber identity. Development 2024; 151:dev202183. [PMID: 38293792 PMCID: PMC10911121 DOI: 10.1242/dev.202183] [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: 07/17/2023] [Accepted: 01/21/2024] [Indexed: 02/01/2024]
Abstract
Ventricular and atrial cardiac chambers have unique structural and contractile characteristics that underlie their distinct functions. The maintenance of chamber-specific features requires active reinforcement, even in differentiated cardiomyocytes. Previous studies in zebrafish have shown that sustained FGF signaling acts upstream of Nkx factors to maintain ventricular identity, but the rest of this maintenance pathway remains unclear. Here, we show that MEK1/2-ERK1/2 signaling acts downstream of FGF and upstream of Nkx factors to promote ventricular maintenance. Inhibition of MEK signaling, like inhibition of FGF signaling, results in ectopic atrial gene expression and reduced ventricular gene expression in ventricular cardiomyocytes. FGF and MEK signaling both influence ventricular maintenance over a similar timeframe, when phosphorylated ERK (pERK) is present in the myocardium. However, the role of FGF-MEK activity appears to be context-dependent: some ventricular regions are more sensitive than others to inhibition of FGF-MEK signaling. Additionally, in the atrium, although endogenous pERK does not induce ventricular traits, heightened MEK signaling can provoke ectopic ventricular gene expression. Together, our data reveal chamber-specific roles of MEK-ERK signaling in the maintenance of ventricular and atrial identities.
Collapse
Affiliation(s)
- Yao Yao
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Deepam Gupta
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Deborah Yelon
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
6
|
Shafi O, Siddiqui G, Jaffry HA. The benign nature and rare occurrence of cardiac myxoma as a possible consequence of the limited cardiac proliferative/ regenerative potential: a systematic review. BMC Cancer 2023; 23:1245. [PMID: 38110859 PMCID: PMC10726542 DOI: 10.1186/s12885-023-11723-3] [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: 08/08/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Cardiac Myxoma is a primary tumor of heart. Its origins, rarity of the occurrence of primary cardiac tumors and how it may be related to limited cardiac regenerative potential, are not yet entirely known. This study investigates the key cardiac genes/ transcription factors (TFs) and signaling pathways to understand these important questions. METHODS Databases including PubMed, MEDLINE, and Google Scholar were searched for published articles without any date restrictions, involving cardiac myxoma, cardiac genes/TFs/signaling pathways and their roles in cardiogenesis, proliferation, differentiation, key interactions and tumorigenesis, with focus on cardiomyocytes. RESULTS The cardiac genetic landscape is governed by a very tight control between proliferation and differentiation-related genes/TFs/pathways. Cardiac myxoma originates possibly as a consequence of dysregulations in the gene expression of differentiation regulators including Tbx5, GATA4, HAND1/2, MYOCD, HOPX, BMPs. Such dysregulations switch the expression of cardiomyocytes into progenitor-like state in cardiac myxoma development by dysregulating Isl1, Baf60 complex, Wnt, FGF, Notch, Mef2c and others. The Nkx2-5 and MSX2 contribute predominantly to both proliferation and differentiation of Cardiac Progenitor Cells (CPCs), may possibly serve roles based on the microenvironment and the direction of cell circuitry in cardiac tumorigenesis. The Nkx2-5 in cardiac myxoma may serve to limit progression of tumorigenesis as it has massive control over the proliferation of CPCs. The cardiac cell type-specific genetic programming plays governing role in controlling the tumorigenesis and regenerative potential. CONCLUSION The cardiomyocytes have very limited proliferative and regenerative potential. They survive for long periods of time and tightly maintain the gene expression of differentiation genes such as Tbx5, GATA4 that interact with tumor suppressors (TS) and exert TS like effect. The total effect such gene expression exerts is responsible for the rare occurrence and benign nature of primary cardiac tumors. This prevents the progression of tumorigenesis. But this also limits the regenerative and proliferative potential of cardiomyocytes. Cardiac Myxoma develops as a consequence of dysregulations in these key genes which revert the cells towards progenitor-like state, hallmark of CM. The CM development in carney complex also signifies the role of TS in cardiac cells.
Collapse
Affiliation(s)
- Ovais Shafi
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan.
| | - Ghazia Siddiqui
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan
| | - Hassam A Jaffry
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
7
|
Gauvrit S, Bossaer J, Lee J, Collins MM. Modeling Human Cardiac Arrhythmias: Insights from Zebrafish. J Cardiovasc Dev Dis 2022; 9:jcdd9010013. [PMID: 35050223 PMCID: PMC8779270 DOI: 10.3390/jcdd9010013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/23/2021] [Accepted: 12/25/2021] [Indexed: 12/13/2022] Open
Abstract
Cardiac arrhythmia, or irregular heart rhythm, is associated with morbidity and mortality and is described as one of the most important future public health challenges. Therefore, developing new models of cardiac arrhythmia is critical for understanding disease mechanisms, determining genetic underpinnings, and developing new therapeutic strategies. In the last few decades, the zebrafish has emerged as an attractive model to reproduce in vivo human cardiac pathologies, including arrhythmias. Here, we highlight the contribution of zebrafish to the field and discuss the available cardiac arrhythmia models. Further, we outline techniques to assess potential heart rhythm defects in larval and adult zebrafish. As genetic tools in zebrafish continue to bloom, this model will be crucial for functional genomics studies and to develop personalized anti-arrhythmic therapies.
Collapse
|
8
|
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
|
9
|
Wang W, Hu YF, Pang M, Chang N, Yu C, Li Q, Xiong JW, Peng Y, Zhang R. BMP and Notch Signaling Pathways differentially regulate Cardiomyocyte Proliferation during Ventricle Regeneration. Int J Biol Sci 2021; 17:2157-2166. [PMID: 34239346 PMCID: PMC8241734 DOI: 10.7150/ijbs.59648] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/14/2021] [Indexed: 01/15/2023] Open
Abstract
Adult mammalian hearts show limited capacity to proliferate after injury, while zebrafish are capable to completely regenerate injured hearts through the proliferation of spared cardiomyocytes. BMP and Notch signaling pathways have been implicated in cardiomyocyte proliferation during zebrafish heart regeneration. However, the molecular mechanism underneath this process as well as the interaction between these two pathways remains to be further explored. In this study we showed BMP signaling was activated after ventricle ablation and acted epistatic downstream of Notch signaling. Inhibition of both signaling pathways differentially influenced ventricle regeneration and cardiomyocyte proliferation, as revealed by time-lapse analysis using a cardiomyocyte-specific FUCCI (fluorescent ubiquitylation-based cell cycle indicator) system. Further experiments revealed that inhibition of BMP and Notch signaling led to cell-cycle arrest at different phases. Overall, our results shed light on the interaction between BMP and Notch signaling pathways and their functions in cardiomyocyte proliferation during cardiac regeneration.
Collapse
Affiliation(s)
- Wenyuan Wang
- School of Life Sciences, Fudan University, Shanghai, China
| | - Ye-Fan Hu
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Meijun Pang
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Nannan Chang
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Chunxiao Yu
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Qi Li
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jing-Wei Xiong
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Yuanyuan Peng
- School of Life Sciences, Fudan University, Shanghai, China
| | - Ruilin Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Rufaihah AJ, Chen CK, Yap CH, Mattar CNZ. Mending a broken heart: In vitro, in vivo and in silico models of congenital heart disease. Dis Model Mech 2021; 14:dmm047522. [PMID: 33787508 PMCID: PMC8033415 DOI: 10.1242/dmm.047522] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Birth defects contribute to ∼0.3% of global infant mortality in the first month of life, and congenital heart disease (CHD) is the most common birth defect among newborns worldwide. Despite the significant impact on human health, most treatments available for this heterogenous group of disorders are palliative at best. For this reason, the complex process of cardiogenesis, governed by multiple interlinked and dose-dependent pathways, is well investigated. Tissue, animal and, more recently, computerized models of the developing heart have facilitated important discoveries that are helping us to understand the genetic, epigenetic and mechanobiological contributors to CHD aetiology. In this Review, we discuss the strengths and limitations of different models of normal and abnormal cardiogenesis, ranging from single-cell systems and 3D cardiac organoids, to small and large animals and organ-level computational models. These investigative tools have revealed a diversity of pathogenic mechanisms that contribute to CHD, including genetic pathways, epigenetic regulators and shear wall stresses, paving the way for new strategies for screening and non-surgical treatment of CHD. As we discuss in this Review, one of the most-valuable advances in recent years has been the creation of highly personalized platforms with which to study individual diseases in clinically relevant settings.
Collapse
Affiliation(s)
- Abdul Jalil Rufaihah
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Ching Kit Chen
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Choon Hwai Yap
- Division of Cardiology, Department of Paediatrics, Khoo Teck Puat -National University Children's Medical Institute, National University Health System, Singapore 119228
- Department of Bioengineering, Imperial College London, London, UK
| | - Citra N Z Mattar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
- Department of Obstetrics and Gynaecology, National University Health System, Singapore 119228
| |
Collapse
|
11
|
Martin KE, Waxman JS. Atrial and Sinoatrial Node Development in the Zebrafish Heart. J Cardiovasc Dev Dis 2021; 8:jcdd8020015. [PMID: 33572147 PMCID: PMC7914448 DOI: 10.3390/jcdd8020015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
Proper development and function of the vertebrate heart is vital for embryonic and postnatal life. Many congenital heart defects in humans are associated with disruption of genes that direct the formation or maintenance of atrial and pacemaker cardiomyocytes at the venous pole of the heart. Zebrafish are an outstanding model for studying vertebrate cardiogenesis, due to the conservation of molecular mechanisms underlying early heart development, external development, and ease of genetic manipulation. Here, we discuss early developmental mechanisms that instruct appropriate formation of the venous pole in zebrafish embryos. We primarily focus on signals that determine atrial chamber size and the specialized pacemaker cells of the sinoatrial node through directing proper specification and differentiation, as well as contemporary insights into the plasticity and maintenance of cardiomyocyte identity in embryonic zebrafish hearts. Finally, we integrate how these insights into zebrafish cardiogenesis can serve as models for human atrial defects and arrhythmias.
Collapse
Affiliation(s)
- Kendall E. Martin
- Molecular Genetics, Biochemistry, and Microbiology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Joshua S. Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Correspondence:
| |
Collapse
|
12
|
Yao Y, Marra AN, Yelon D. Pathways Regulating Establishment and Maintenance of Cardiac Chamber Identity in Zebrafish. J Cardiovasc Dev Dis 2021; 8:13. [PMID: 33572830 PMCID: PMC7912383 DOI: 10.3390/jcdd8020013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 02/07/2023] Open
Abstract
The vertebrate heart is comprised of two types of chambers-ventricles and atria-that have unique morphological and physiological properties. Effective cardiac function depends upon the distinct characteristics of ventricular and atrial cardiomyocytes, raising interest in the genetic pathways that regulate chamber-specific traits. Chamber identity seems to be specified in the early embryo by signals that establish ventricular and atrial progenitor populations and trigger distinct differentiation pathways. Intriguingly, chamber-specific features appear to require active reinforcement, even after myocardial differentiation is underway, suggesting plasticity of chamber identity within the developing heart. Here, we review the utility of the zebrafish as a model organism for studying the mechanisms that establish and maintain cardiac chamber identity. By combining genetic and embryological approaches, work in zebrafish has revealed multiple players with potent influences on chamber fate specification and commitment. Going forward, analysis of cardiomyocyte identity at the single-cell level is likely to yield a high-resolution understanding of the pathways that link the relevant players together, and these insights will have the potential to inform future strategies in cardiac tissue engineering.
Collapse
Affiliation(s)
| | | | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; (Y.Y.); (A.N.M.)
| |
Collapse
|
13
|
Pawlak M, Kedzierska KZ, Migdal M, Karim AN, Ramilowski JA, Bugajski L, Hashimoto K, Marconi A, Piwocka K, Carninci P, Winata CL. Dynamics of cardiomyocyte transcriptome and chromatin landscape demarcates key events of heart development. Genome Res 2019; 29:506-519. [PMID: 30760547 PMCID: PMC6396412 DOI: 10.1101/gr.244491.118] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/09/2019] [Indexed: 12/19/2022]
Abstract
Organogenesis involves dynamic regulation of gene transcription and complex multipathway interactions. Despite our knowledge of key factors regulating various steps of heart morphogenesis, considerable challenges in understanding its mechanism still exist because little is known about their downstream targets and interactive regulatory network. To better understand transcriptional regulatory mechanism driving heart development and the consequences of its disruption in vivo, we performed time-series analyses of the transcriptome and genome-wide chromatin accessibility in isolated cardiomyocytes (CMs) from wild-type zebrafish embryos at developmental stages corresponding to heart tube morphogenesis, looping, and maturation. We identified genetic regulatory modules driving crucial events of heart development that contained key cardiac TFs and are associated with open chromatin regions enriched for DNA sequence motifs belonging to the family of the corresponding TFs. Loss of function of cardiac TFs Gata5, Tbx5a, and Hand2 affected the cardiac regulatory networks and caused global changes in chromatin accessibility profile, indicating their role in heart development. Among regions with differential chromatin accessibility in mutants were highly conserved noncoding elements that represent putative enhancers driving heart development. The most prominent gene expression changes, which correlated with chromatin accessibility modifications within their proximal promoter regions, occurred between heart tube morphogenesis and looping, and were associated with metabolic shift and hematopoietic/cardiac fate switch during CM maturation. Our results revealed the dynamic regulatory landscape throughout heart development and identified interactive molecular networks driving key events of heart morphogenesis.
Collapse
Affiliation(s)
- Michal Pawlak
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, 02-109 Warsaw, Poland
| | - Katarzyna Z Kedzierska
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, 02-109 Warsaw, Poland
| | - Maciej Migdal
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, 02-109 Warsaw, Poland
| | - Abu Nahia Karim
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, 02-109 Warsaw, Poland
| | | | - Lukasz Bugajski
- Nencki Institute of Experimental Biology, Laboratory of Cytometry, 02-093 Warsaw, Poland
| | - Kosuke Hashimoto
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045 Japan
| | - Aleksandra Marconi
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, 02-109 Warsaw, Poland
| | - Katarzyna Piwocka
- Nencki Institute of Experimental Biology, Laboratory of Cytometry, 02-093 Warsaw, Poland
| | - Piero Carninci
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045 Japan
| | - Cecilia L Winata
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, 02-109 Warsaw, Poland
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
14
|
Fukui H, Chiba A, Miyazaki T, Takano H, Ishikawa H, Omori T, Mochiuzki N. Spatial Allocation and Specification of Cardiomyocytes during Zebrafish Embryogenesis. Korean Circ J 2017; 47:160-167. [PMID: 28382067 PMCID: PMC5378018 DOI: 10.4070/kcj.2016.0280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/05/2016] [Accepted: 10/13/2016] [Indexed: 12/21/2022] Open
Abstract
Incomplete development and severe malformation of the heart result in miscarriage of embryos because of its malfunction as a pump for circulation. During cardiogenesis, development of the heart is precisely coordinated by the genetically-primed program that is revealed by the sequential expression of transcription factors. It is important to investigate how spatial allocation of the heart containing cardiomyocytes and other mesoderm-derived cells is determined. In addition, the molecular mechanism underlying cardiomyocyte differentiation still remains elusive. The location of ectoderm-, mesoderm-, and endoderm-derived organs is determined by their initial allocation and subsequent mutual cell-cell interactions or paracrine-based regulation. In the present work, we provide an overview of cardiac development controlled by the germ layers and discuss the points that should be uncovered in future for understanding cardiogenesis.
Collapse
Affiliation(s)
- Hajime Fukui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Ayano Chiba
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Takahiro Miyazaki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Haruko Takano
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Hiroyuki Ishikawa
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Toyonori Omori
- Management office, National Center for Child Health and Development, Tokyo, Japan
| | - Naoki Mochiuzki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan.; AMED-CREST, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| |
Collapse
|
15
|
Bylund JB, Trinh LT, Awgulewitsch CP, Paik DT, Jetter C, Jha R, Zhang J, Nolan K, Xu C, Thompson TB, Kamp TJ, Hatzopoulos AK. Coordinated Proliferation and Differentiation of Human-Induced Pluripotent Stem Cell-Derived Cardiac Progenitor Cells Depend on Bone Morphogenetic Protein Signaling Regulation by GREMLIN 2. Stem Cells Dev 2017; 26:678-693. [PMID: 28125926 DOI: 10.1089/scd.2016.0226] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Heart development depends on coordinated proliferation and differentiation of cardiac progenitor cells (CPCs), but how the two processes are synchronized is not well understood. Here, we show that the secreted Bone Morphogenetic Protein (BMP) antagonist GREMLIN 2 (GREM2) is induced in CPCs shortly after cardiac mesoderm specification during differentiation of human pluripotent stem cells. GREM2 expression follows cardiac lineage differentiation independently of the differentiation method used, or the origin of the pluripotent stem cells, suggesting that GREM2 is linked to cardiogenesis. Addition of GREM2 protein strongly increases cardiomyocyte output compared to established procardiogenic differentiation methods. Our data show that inhibition of canonical BMP signaling by GREM2 is necessary to promote proliferation of CPCs. However, canonical BMP signaling inhibition alone is not sufficient to induce cardiac differentiation, which depends on subsequent JNK pathway activation specifically by GREM2. These findings may have broader implications in the design of approaches to orchestrate growth and differentiation of pluripotent stem cell-derived lineages that depend on precise regulation of BMP signaling.
Collapse
Affiliation(s)
- Jeffery B Bylund
- 1 Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee.,2 Department of Pharmacology, Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Linh T Trinh
- 1 Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Cassandra P Awgulewitsch
- 1 Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - David T Paik
- 1 Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee.,3 Department of Cell and Developmental Biology, Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Christopher Jetter
- 1 Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Rajneesh Jha
- 4 Department of Pediatrics, Emory University School of Medicine , Atlanta, Georgia
| | - Jianhua Zhang
- 5 Stem Cell and Regenerative Medicine Center, University of Wisconsin School of Medicine and Public Health , Madison, Wisconsin
| | - Kristof Nolan
- 6 Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati , Cincinnati, Ohio
| | - Chunhui Xu
- 4 Department of Pediatrics, Emory University School of Medicine , Atlanta, Georgia
| | - Thomas B Thompson
- 6 Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati , Cincinnati, Ohio
| | - Timothy J Kamp
- 5 Stem Cell and Regenerative Medicine Center, University of Wisconsin School of Medicine and Public Health , Madison, Wisconsin
| | - Antonis K Hatzopoulos
- 1 Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee.,3 Department of Cell and Developmental Biology, Vanderbilt University School of Medicine , Nashville, Tennessee
| |
Collapse
|
16
|
Calderon D, Bardot E, Dubois N. Probing early heart development to instruct stem cell differentiation strategies. Dev Dyn 2016; 245:1130-1144. [PMID: 27580352 DOI: 10.1002/dvdy.24441] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 08/20/2016] [Accepted: 08/20/2016] [Indexed: 12/19/2022] Open
Abstract
Scientists have studied organs and their development for centuries and, along that path, described models and mechanisms explaining the developmental principles of organogenesis. In particular, with respect to the heart, new fundamental discoveries are reported continuously that keep changing the way we think about early cardiac development. These discoveries are driven by the need to answer long-standing questions regarding the origin of the earliest cells specified to the cardiac lineage, the differentiation potential of distinct cardiac progenitor cells, and, very importantly, the molecular mechanisms underlying these specification events. As evidenced by numerous examples, the wealth of developmental knowledge collected over the years has had an invaluable impact on establishing efficient strategies to generate cardiovascular cell types ex vivo, from either pluripotent stem cells or via direct reprogramming approaches. The ability to generate functional cardiovascular cells in an efficient and reliable manner will contribute to therapeutic strategies aimed at alleviating the increasing burden of cardiovascular disease and morbidity. Here we will discuss the recent discoveries in the field of cardiac progenitor biology and their translation to the pluripotent stem cell model to illustrate how developmental concepts have instructed regenerative model systems in the past and promise to do so in the future. Developmental Dynamics 245:1130-1144, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Damelys Calderon
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Evan Bardot
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Nicole Dubois
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| |
Collapse
|
17
|
Sanders LN, Schoenhard JA, Saleh MA, Mukherjee A, Ryzhov S, McMaster WG, Nolan K, Gumina RJ, Thompson TB, Magnuson MA, Harrison DG, Hatzopoulos AK. BMP Antagonist Gremlin 2 Limits Inflammation After Myocardial Infarction. Circ Res 2016; 119:434-49. [PMID: 27283840 DOI: 10.1161/circresaha.116.308700] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/09/2016] [Indexed: 11/16/2022]
Abstract
RATIONALE We have recently shown that the bone morphogenetic protein (BMP) antagonist Gremlin 2 (Grem2) is required for early cardiac development and cardiomyocyte differentiation. Our initial studies discovered that Grem2 is strongly induced in the adult heart after experimental myocardial infarction (MI). However, the function of Grem2 and BMP-signaling inhibitors after cardiac injury is currently unknown. OBJECTIVE To investigate the role of Grem2 during cardiac repair and assess its potential to improve ventricular function after injury. METHODS AND RESULTS Our data show that Grem2 is transiently induced after MI in peri-infarct area cardiomyocytes during the inflammatory phase of cardiac tissue repair. By engineering loss- (Grem2(-/-)) and gain- (TG(Grem2)) of-Grem2-function mice, we discovered that Grem2 controls the magnitude of the inflammatory response and limits infiltration of inflammatory cells in peri-infarct ventricular tissue, improving cardiac function. Excessive inflammation in Grem2(-/-) mice after MI was because of overactivation of canonical BMP signaling, as proven by the rescue of the inflammatory phenotype through administration of the canonical BMP inhibitor, DMH1. Furthermore, intraperitoneal administration of Grem2 protein in wild-type mice was sufficient to reduce inflammation after MI. Cellular analyses showed that BMP2 acts with TNFα to induce expression of proinflammatory proteins in endothelial cells and promote adhesion of leukocytes, whereas Grem2 specifically inhibits the BMP2 effect. CONCLUSIONS Our results indicate that Grem2 provides a molecular barrier that controls the magnitude and extent of inflammatory cell infiltration by suppressing canonical BMP signaling, thereby providing a novel mechanism for limiting the adverse effects of excessive inflammation after MI.
Collapse
Affiliation(s)
- Lehanna N Sanders
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - John A Schoenhard
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Mohamed A Saleh
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Amrita Mukherjee
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Sergey Ryzhov
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - William G McMaster
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Kristof Nolan
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Richard J Gumina
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Thomas B Thompson
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Mark A Magnuson
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - David G Harrison
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Antonis K Hatzopoulos
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.).
| |
Collapse
|
18
|
Crottès D, Rapetti-Mauss R, Alcaraz-Perez F, Tichet M, Gariano G, Martial S, Guizouarn H, Pellissier B, Loubat A, Popa A, Paquet A, Presta M, Tartare-Deckert S, Cayuela ML, Martin P, Borgese F, Soriani O. SIGMAR1 Regulates Membrane Electrical Activity in Response to Extracellular Matrix Stimulation to Drive Cancer Cell Invasiveness. Cancer Res 2016; 76:607-18. [PMID: 26645564 DOI: 10.1158/0008-5472.can-15-1465] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 10/27/2015] [Indexed: 01/12/2023]
Abstract
The sigma 1 receptor (Sig1R) is a stress-activated chaperone that regulates ion channels and is associated with pathologic conditions, such as stroke, neurodegenerative diseases, and addiction. Aberrant expression levels of ion channels and Sig1R have been detected in tumors and cancer cells, such as myeloid leukemia and colorectal cancer, but the link between ion channel regulation and Sig1R overexpression during malignancy has not been established. In this study, we found that Sig1R dynamically controls the membrane expression of the human voltage-dependent K(+) channel human ether-à-go-go-related gene (hERG) in myeloid leukemia and colorectal cancer cell lines. Sig1R promoted the formation of hERG/β1-integrin signaling complexes upon extracellular matrix stimulation, triggering the activation of the PI3K/AKT pathway. Consequently, the presence of Sig1R in cancer cells increased motility and VEGF secretion. In vivo, Sig1R expression enhanced the aggressiveness of tumor cells by potentiating invasion and angiogenesis, leading to poor survival. Collectively, our findings highlight a novel function for Sig1R in mediating cross-talk between cancer cells and their microenvironment, thus driving oncogenesis by shaping cellular electrical activity in response to extracellular signals. Given the involvement of ion channels in promoting several hallmarks of cancer, our study also offers a potential strategy to therapeutically target ion channel function through Sig1R inhibition.
Collapse
Affiliation(s)
- David Crottès
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France. Department of Physiology, University of California, San Francisco, San Francisco, California
| | - Raphael Rapetti-Mauss
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Francisca Alcaraz-Perez
- Telomerase, Aging and Cancer Group, Research Unit, Department of Surgery, CIBERehd, University Hospital "Virgen de la Arrixaca", Murcia, Spain. Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Mélanie Tichet
- Université Nice Sophia Antipolis, C3M, Inserm U1065, Nice, France
| | - Giuseppina Gariano
- Unit of Oncology and Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sonia Martial
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Hélène Guizouarn
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Bernard Pellissier
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Agnès Loubat
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Alexandra Popa
- Université Nice Sophia Antipolis, IPMC, CNRS UMR7275, Sophia Antipolis, France
| | - Agnès Paquet
- Université Nice Sophia Antipolis, IPMC, CNRS UMR7275, Sophia Antipolis, France
| | - Marco Presta
- Unit of Oncology and Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Maria Luisa Cayuela
- Telomerase, Aging and Cancer Group, Research Unit, Department of Surgery, CIBERehd, University Hospital "Virgen de la Arrixaca", Murcia, Spain. Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Patrick Martin
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Franck Borgese
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Olivier Soriani
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France.
| |
Collapse
|
19
|
Caporilli S, Latinkic BV. Ventricular cell fate can be specified until the onset of myocardial differentiation. Mech Dev 2016; 139:31-41. [PMID: 26776863 PMCID: PMC4798847 DOI: 10.1016/j.mod.2016.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/24/2015] [Accepted: 01/07/2016] [Indexed: 02/03/2023]
Abstract
The mechanisms that govern specification of various cell types that constitute vertebrate heart are not fully understood. Whilst most studies of heart development have utilised the mouse embryo, we have used an alternative model, embryos of the frog Xenopus laevis, which permits direct experimental manipulation of a non-essential heart. We show that in this model pluripotent animal cap explants injected with cardiogenic factor GATA4 mRNA express pan-myocardial as well as ventricular and proepicardial markers. We found that cardiac cell fate diversification, as assessed by ventricular and proepicardial markers, critically depends on tissue integrity, as it is disrupted by dissociation but can be fully restored by inhibition of the BMP pathway and partially by Dkk-1. Ventricular and proepicardial cell fates can also be restored in reaggregated GATA4-expressing cells upon transplantation into a host embryo. The competence of the host embryo to induce ventricular and proepicardial markers gradually decreases with the age of the transplant and is lost by the onset of myocardial differentiation at the late tailbud stage (st. 28). The influence of the host on the transplant was not limited to diversification of cardiac cell fates, but also included induction of growth and rhythmic beating, resulting in generation of a secondary heart-like structure. Our results additionally show that efficient generation of secondary heart requires normal axial patterning of the host embryo. Furthermore, secondary hearts can be induced in a wide range of locations within the host, arguing that the host embryo provides a permissive environment for development of cardiac patterning, growth and physiological maturation. Our results have implications for a major goal of cardiac regenerative medicine, differentiation of ventricular myocardium. Ventricular and proepicardial fate can be induced by cardiogenic factor GATA4. This process requires tissue integrity. Ventricular and proepicardial cell fate can be restored by BMP inhibition. A secondary heart-like structure can be induced from GATA4-expressing cells.
Collapse
Affiliation(s)
- Simona Caporilli
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, Wales, UK
| | - Branko V Latinkic
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, Wales, UK.
| |
Collapse
|
20
|
Affiliation(s)
- Dennis Schade
- Department
of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse
6, 44227 Dortmund, Germany
| | - Alleyn T. Plowright
- Department
of Medicinal Chemistry, Cardiovascular and Metabolic Diseases Innovative
Medicines, AstraZeneca, Pepparedsleden 1, Mölndal, 43183, Sweden
| |
Collapse
|
21
|
Palencia-Desai S, Rost MS, Schumacher JA, Ton QV, Craig MP, Baltrunaite K, Koenig AL, Wang J, Poss KD, Chi NC, Stainier DYR, Sumanas S. Myocardium and BMP signaling are required for endocardial differentiation. Development 2015; 142:2304-15. [PMID: 26092845 DOI: 10.1242/dev.118687] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 05/12/2015] [Indexed: 01/09/2023]
Abstract
Endocardial and myocardial progenitors originate in distinct regions of the anterior lateral plate mesoderm and migrate to the midline where they coalesce to form the cardiac tube. Endocardial progenitors acquire a molecular identity distinct from other vascular endothelial cells and initiate expression of specific genes such as nfatc1. Yet the molecular pathways and tissue interactions involved in establishing endocardial identity are poorly understood. The endocardium develops in tight association with cardiomyocytes. To test for a potential role of the myocardium in endocardial morphogenesis, we used two different zebrafish models deficient in cardiomyocytes: the hand2 mutant and a myocardial-specific genetic ablation method. We show that in hand2 mutants endocardial progenitors migrate to the midline but fail to assemble into a cardiac cone and do not express markers of differentiated endocardium. Endocardial differentiation defects were rescued by myocardial but not endocardial-specific expression of hand2. In metronidazole-treated myl7:nitroreductase embryos, myocardial cells were targeted for apoptosis, which resulted in the loss of endocardial nfatc1 expression. However, endocardial cells were present and retained expression of general vascular endothelial markers. We further identified bone morphogenetic protein (BMP) as a candidate myocardium-derived signal required for endocardial differentiation. Chemical and genetic inhibition of BMP signaling at the tailbud stage resulted in severe inhibition of endocardial differentiation while there was little effect on myocardial development. Heat-shock-induced bmp2b expression rescued endocardial nfatc1 expression in hand2 mutants and in myocardium-depleted embryos. Our results indicate that the myocardium is crucial for endocardial morphogenesis and differentiation, and identify BMP as a signal involved in endocardial differentiation.
Collapse
Affiliation(s)
- Sharina Palencia-Desai
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Megan S Rost
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jennifer A Schumacher
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Quynh V Ton
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michael P Craig
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kristina Baltrunaite
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Andrew L Koenig
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jinhu Wang
- Department of Cell Biology and Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Kenneth D Poss
- Department of Cell Biology and Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Neil C Chi
- Department of Biochemistry and Biophysics, UCSF, San Francisco, CA 94158, USA
| | - Didier Y R Stainier
- Department of Biochemistry and Biophysics, UCSF, San Francisco, CA 94158, USA
| | - Saulius Sumanas
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
22
|
Birket MJ, Mummery CL. Pluripotent stem cell derived cardiovascular progenitors--a developmental perspective. Dev Biol 2015; 400:169-79. [PMID: 25624264 DOI: 10.1016/j.ydbio.2015.01.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 01/12/2015] [Accepted: 01/14/2015] [Indexed: 12/15/2022]
Abstract
Human pluripotent stem cells can now be routinely differentiated into cardiac cell types including contractile cardiomyocytes, enabling the study of heart development and disease in vitro, and creating opportunities for the development of novel therapeutic interventions for patients. Our grasp of the system, however, remains partial, and a significant reason for this has been our inability to effectively purify and expand the intermediate cardiovascular progenitor cells (CPCs) equivalent to those studied in heart development. Doing so could facilitate the construction of a cardiac lineage cell fate map, boosting our capacity to more finely control stem cell lineage commitment to functionally distinct cardiac identities, as well as providing a model for identifying which genes confer cardiac potential on CPCs. This review offers a perspective on CPC development as understood from model organisms and pluripotent stem cell systems, focusing on issues of identity as well as the signalling implicated in inducing, expanding and patterning these cells.
Collapse
Affiliation(s)
- Matthew J Birket
- Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | | |
Collapse
|
23
|
Targoff KL, Colombo S, George V, Schell T, Kim SH, Solnica-Krezel L, Yelon D. Nkx genes are essential for maintenance of ventricular identity. Development 2013; 140:4203-13. [PMID: 24026123 DOI: 10.1242/dev.095562] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Establishment of specific characteristics of each embryonic cardiac chamber is crucial for development of a fully functional adult heart. Despite the importance of defining and maintaining unique features in ventricular and atrial cardiomyocytes, the regulatory mechanisms guiding these processes are poorly understood. Here, we show that the homeodomain transcription factors Nkx2.5 and Nkx2.7 are necessary to sustain ventricular chamber attributes through repression of atrial chamber identity. Mutation of nkx2.5 in zebrafish yields embryos with diminutive ventricular and bulbous atrial chambers. These chamber deformities emerge gradually during development, with a severe collapse in the number of ventricular cardiomyocytes and an accumulation of excess atrial cardiomyocytes as the heart matures. Removal of nkx2.7 function from nkx2.5 mutants exacerbates the loss of ventricular cells and the gain of atrial cells. Moreover, in these Nkx-deficient embryos, expression of vmhc, a ventricular gene, fades, whereas expression of amhc, an atrial gene, expands. Cell-labeling experiments suggest that ventricular cardiomyocytes can transform into atrial cardiomyocytes in the absence of Nkx gene function. Through suggestion of transdifferentiation from ventricular to atrial fate, our data reveal a pivotal role for Nkx genes in maintaining ventricular identity and highlight remarkable plasticity in differentiated myocardium. Thus, our results are relevant to the etiologies of fetal and neonatal cardiac pathology and could direct future innovations in cardiac regenerative medicine.
Collapse
Affiliation(s)
- Kimara L Targoff
- Developmental Genetics Program and Department of Cell Biology, Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Staudt D, Stainier D. Uncovering the molecular and cellular mechanisms of heart development using the zebrafish. Annu Rev Genet 2012; 46:397-418. [PMID: 22974299 DOI: 10.1146/annurev-genet-110711-155646] [Citation(s) in RCA: 205] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Over the past 20 years, the zebrafish has emerged as a powerful model organism for studying cardiac development. Its ability to survive without an active circulation and amenability to forward genetics has led to the identification of numerous mutants whose study has helped elucidate new mechanisms in cardiac development. Furthermore, its transparent, externally developing embryos have allowed detailed cellular analyses of heart development. In this review, we discuss the molecular and cellular processes involved in zebrafish heart development from progenitor specification to development of the valve and the conduction system. We focus on imaging studies that have uncovered the cellular bases of heart development and on zebrafish mutants with cardiac abnormalities whose study has revealed novel molecular pathways in cardiac cell specification and tissue morphogenesis.
Collapse
Affiliation(s)
- David Staudt
- Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158, USA
| | | |
Collapse
|
25
|
Ao A, Hao J, Hopkins CR, Hong CC. DMH1, a novel BMP small molecule inhibitor, increases cardiomyocyte progenitors and promotes cardiac differentiation in mouse embryonic stem cells. PLoS One 2012; 7:e41627. [PMID: 22848549 PMCID: PMC3407188 DOI: 10.1371/journal.pone.0041627] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 06/22/2012] [Indexed: 01/24/2023] Open
Abstract
The possibility of using cell-based therapeutics to treat cardiac failure has generated significant interest since the initial introduction of stem cell-based technologies. However, the methods to quickly and robustly direct stem cell differentiation towards cardiac cell types have been limited by a reliance on recombinant growth factors to provide necessary biological cues. We report here the use of dorsomorphin homologue 1 (DMH1), a second-generation small molecule BMP inhibitor based on dorsomorphin, to efficiently induce beating cardiomyocyte formation in mouse embryonic stem cells (ESCs) and to specifically upregulate canonical transcriptional markers associated with cardiac development. DMH1 differs significantly from its predecessor by its ability to enrich for pro-cardiac progenitor cells that respond to late-stage Wnt inhibition using XAV939 and produce secondary beating cardiomyocytes. Our study demonstrates the utility of small molecules to complement existing in vitro cardiac differentiation protocols and highlights the role of transient BMP inhibition in cardiomyogenesis.
Collapse
Affiliation(s)
- Ada Ao
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- * E-mail: (AA); (CCH)
| | - Jijun Hao
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Corey R. Hopkins
- Vanderbilt Institute of Chemical Biology, Department of Pharmacology, Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Charles C. Hong
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Vanderbilt Institute of Chemical Biology, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, United States of America
- * E-mail: (AA); (CCH)
| |
Collapse
|
26
|
Tu S, Chi NC. Zebrafish models in cardiac development and congenital heart birth defects. Differentiation 2012; 84:4-16. [PMID: 22704690 DOI: 10.1016/j.diff.2012.05.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 05/02/2012] [Accepted: 05/21/2012] [Indexed: 12/31/2022]
Abstract
The zebrafish has become an ideal vertebrate animal system for investigating cardiac development due to its genetic tractability, external fertilization, early optical clarity and ability to survive without a functional cardiovascular system during development. In particular, recent advances in imaging techniques and the creation of zebrafish transgenics now permit the in vivo analysis of the dynamic cellular events that transpire during cardiac morphogenesis. As a result, the combination of these salient features provides detailed insight as to how specific genes may influence cardiac development at the cellular level. In this review, we will highlight how the zebrafish has been utilized to elucidate not only the underlying mechanisms of cardiac development and human congenital heart diseases (CHDs), but also potential pathways that may modulate cardiac regeneration. Thus, we have organized this review based on the major categories of CHDs-structural heart, functional heart, and vascular/great vessel defects, and will conclude with how the zebrafish may be further used to contribute to our understanding of specific human CHDs in the future.
Collapse
Affiliation(s)
- Shu Tu
- Department of Medicine, Division of Cardiology, University of California, San Diego, CA 92093-0613J, USA
| | | |
Collapse
|
27
|
Chernyavskaya Y, Ebert AM, Milligan E, Garrity DM. Voltage-gated calcium channel CACNB2 (β2.1) protein is required in the heart for control of cell proliferation and heart tube integrity. Dev Dyn 2012; 241:648-62. [DOI: 10.1002/dvdy.23746] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2012] [Indexed: 01/11/2023] Open
|
28
|
de Pater E, Ciampricotti M, Priller F, Veerkamp J, Strate I, Smith K, Lagendijk AK, Schilling TF, Herzog W, Abdelilah-Seyfried S, Hammerschmidt M, Bakkers J. Bmp signaling exerts opposite effects on cardiac differentiation. Circ Res 2012; 110:578-87. [PMID: 22247485 DOI: 10.1161/circresaha.111.261172] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
RATIONALE The importance for Bmp signaling during embryonic stem cell differentiation into myocardial cells has been recognized. The question when and where Bmp signaling in vivo regulates myocardial differentiation has remained largely unanswered. OBJECTIVE To identify when and where Bmp signaling regulates cardiogenic differentiation. METHODS AND RESULTS Here we have observed that in zebrafish embryos, Bmp signaling is active in cardiac progenitor cells prior to their differentiation into cardiomyocytes. Bmp signaling is continuously required during somitogenesis within the anterior lateral plate mesoderm to induce myocardial differentiation. Surprisingly, Bmp signaling is actively repressed in differentiating myocardial cells. We identified the inhibitory Smad6a, which is expressed in the cardiac tissue, to be required to inhibit Bmp signaling and thereby promote expansion of the ventricular myocardium. CONCLUSION Bmp signaling exerts opposing effects on myocardial differentiation in the embryo by promoting as well as inhibiting cardiac growth.
Collapse
Affiliation(s)
- Emma de Pater
- Cardiac development and genetics group, Hubrecht Institute for Developmental Biology and Stem Cell Research, Uppsalalaan 8, Utrecht, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ni TT, Rellinger EJ, Mukherjee A, Stephens L, Thorne CA, Kim K, Hu J, Xie S, Lee E, Marnett L, Hatzopoulos AK, Zhong TP. Discovering small molecules that promote cardiomyocyte generation by modulating Wnt signaling. CHEMISTRY & BIOLOGY 2011; 18:1658-68. [PMID: 22195568 PMCID: PMC3645312 DOI: 10.1016/j.chembiol.2011.09.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 09/18/2011] [Accepted: 09/19/2011] [Indexed: 01/12/2023]
Abstract
We have developed a robust in vivo small-molecule screen that modulates heart size and cardiomyocyte generation in zebrafish. Three structurally related compounds (Cardionogen-1 to Cardionogen-3) identified from our screen enlarge the size of the developing heart via myocardial hyperplasia. Increased cardiomyocyte number in Cardionogen-treated embryos is due to expansion of cardiac progenitor cells. In zebrafish embryos and murine embryonic stem (ES) cells, Cardionogen treatment promotes cardiogenesis during and after gastrulation, whereas it inhibits heart formation before gastrulation. Cardionogen-induced effects can be antagonized by increasing Wnt/β-catenin signaling activity. We demonstrate that Cardionogen inhibits Wnt/β-catenin-dependent transcription in murine ES cells and zebrafish embryos. Cardionogen can rescue Wnt8-induced cardiomyocyte deficiency and heart-specific phenotypes during development. These findings demonstrate that in vivo small-molecule screens targeting heart size can reveal compounds with cardiomyogenic effects and identify underlying target pathways.
Collapse
Affiliation(s)
- Terri T. Ni
- State Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 20043, China
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Eric J. Rellinger
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Amrita Mukherjee
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Lauren Stephens
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Cutris A Thorne
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Kwangho Kim
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Jiangyong Hu
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Shuying Xie
- State Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 20043, China
| | - Ethan Lee
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Larry Marnett
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Antonis K. Hatzopoulos
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Tao P. Zhong
- State Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 20043, China
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| |
Collapse
|
30
|
Distinct phases of Wnt/β-catenin signaling direct cardiomyocyte formation in zebrafish. Dev Biol 2011; 361:364-76. [PMID: 22094017 DOI: 10.1016/j.ydbio.2011.10.032] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 10/25/2011] [Accepted: 10/27/2011] [Indexed: 02/02/2023]
Abstract
Normal heart formation requires reiterative phases of canonical Wnt/β-catenin (Wnt) signaling. Understanding the mechanisms by which Wnt signaling directs cardiomyocyte (CM) formation in vivo is critical to being able to precisely direct differentiated CMs from stem cells in vitro. Here, we investigate the roles of Wnt signaling in zebrafish CM formation using heat-shock inducible transgenes that increase and decrease Wnt signaling. We find that there are three phases during which CM formation is sensitive to modulation of Wnt signaling through the first 24 h of development. In addition to the previously recognized roles for Wnt signaling during mesoderm specification and in the pre-cardiac mesoderm, we find a previously unrecognized role during CM differentiation where Wnt signaling is necessary and sufficient to promote the differentiation of additional atrial cells. We also extend the previous studies of the roles of Wnt signaling during mesoderm specification and in pre-cardiac mesoderm. Importantly, in pre-cardiac mesoderm we define a new mechanism where Wnt signaling is sufficient to prevent CM differentiation, in contrast to a proposed role in inhibiting cardiac progenitor (CP) specification. The inability of the CPs to differentiate appears to lead to cell death through a p53/Caspase-3 independent mechanism. Together with a report for an even later role for Wnt signaling in restricting proliferation of differentiated ventricular CMs, our results indicate that during the first 3days of development in zebrafish there are four distinct phases during which CMs are sensitive to Wnt signaling.
Collapse
|
31
|
Abstract
Over the last decade, the zebrafish has entered the field of cardiovascular research as a new model organism. This is largely due to a number of highly successful small- and large-scale forward genetic screens, which have led to the identification of zebrafish mutants with cardiovascular defects. Genetic mapping and identification of the affected genes have resulted in novel insights into the molecular regulation of vertebrate cardiac development. More recently, the zebrafish has become an attractive model to study the effect of genetic variations identified in patients with cardiovascular defects by candidate gene or whole-genome-association studies. Thanks to an almost entirely sequenced genome and high conservation of gene function compared with humans, the zebrafish has proved highly informative to express and study human disease-related gene variants, providing novel insights into human cardiovascular disease mechanisms, and highlighting the suitability of the zebrafish as an excellent model to study human cardiovascular diseases. In this review, I discuss recent discoveries in the field of cardiac development and specific cases in which the zebrafish has been used to model human congenital and acquired cardiac diseases.
Collapse
Affiliation(s)
- Jeroen Bakkers
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Interuniversity Cardiology Institute of The Netherlands, 3584 CT Utrecht, The Netherlands.
| |
Collapse
|
32
|
Wang J, Greene SB, Martin JF. BMP signaling in congenital heart disease: new developments and future directions. ACTA ACUST UNITED AC 2011; 91:441-8. [PMID: 21384533 DOI: 10.1002/bdra.20785] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 12/22/2010] [Accepted: 01/03/2011] [Indexed: 01/07/2023]
Abstract
Congenital heart malformations are the most common of all congenital human birth anomalies. During the past decade, research with zebrafish, chick, and mouse models have elucidated many fundamental genetic pathways that govern early cardiac patterning and differentiation. This review highlights the roles of the bone morphogenetic protein (BMP) signaling pathway in cardiogenesis and how defective BMP signals can disrupt the intricate steps of cardiac formation and cause congenital heart defects.
Collapse
Affiliation(s)
- Jun Wang
- Institute of Biosciences and Technology, Texas A&M System Health Science Center, 2121 W. Holcombe Blvd., Houston, TX 77030, USA
| | | | | |
Collapse
|
33
|
Laux DW, Febbo JA, Roman BL. Dynamic analysis of BMP-responsive smad activity in live zebrafish embryos. Dev Dyn 2011; 240:682-94. [PMID: 21337466 DOI: 10.1002/dvdy.22558] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2010] [Indexed: 11/06/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are critical players in development and disease, regulating such diverse processes as dorsoventral patterning, palate formation, and ossification. These ligands are classically considered to signal via BMP receptor-specific Smad proteins 1, 5, and 8. To determine the spatiotemporal pattern of Smad1/5/8 activity and thus canonical BMP signaling in the developing zebrafish embryo, we generated a transgenic line expressing EGFP under the control of a BMP-responsive element. EGFP is expressed in many established BMP signaling domains and is responsive to alterations in BMP type I receptor activity and smad1 and smad5 expression. This transgenic Smad1/5/8 reporter line will be useful for determining ligand and receptor requirements for specific domains of BMP activity, as well as for genetic and pharmacological screens aimed at identifying enhancers or suppressors of canonical BMP signaling.
Collapse
|
34
|
Abstract
The myocardium of the heart is composed of multiple highly specialized myocardial lineages, including those of the ventricular and atrial myocardium, and the specialized conduction system. Specification and maturation of each of these lineages during heart development is a highly ordered, ongoing process involving multiple signaling pathways and their intersection with transcriptional regulatory networks. Here, we attempt to summarize and compare much of what we know about specification and maturation of myocardial lineages from studies in several different vertebrate model systems. To date, most research has focused on early specification, and although there is still more to learn about early specification, less is known about factors that promote subsequent maturation of myocardial lineages required to build the functioning adult heart.
Collapse
Affiliation(s)
- Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Medicine, University of California San Diego, 9500 Gilman Dr, La Jolla CA 92093, USA.
| | | | | | | |
Collapse
|
35
|
Affiliation(s)
- Michela Noseda
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Tessa Peterkin
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Filipa C. Simões
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Roger Patient
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Michael D. Schneider
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| |
Collapse
|
36
|
Young DA, DeQuach JA, Christman KL. Human cardiomyogenesis and the need for systems biology analysis. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2010; 3:666-80. [PMID: 21197666 DOI: 10.1002/wsbm.141] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease remains the leading cause of death in the Western world and myocardial infarction is one of the primary facets of this disease. The limited natural self-renewal of cardiac muscle following injury and restricted supply of heart transplants has encouraged researchers to investigate other means to stimulate regeneration of damaged myocardium. The plasticity of stem cells toward multiple lineages offers the potential to repair the heart following injury. Embryonic stem cells have been extensively studied for their ability to differentiate into early cardiomyocytes, however, the pathway has only been partially defined and inadequate efficiency limits their clinical applicability. Some studies have shown cardiomyogenesis from adult mesenchymal stem cells, from both bone marrow and adipose tissue, but their differentiation pathway remains poorly detailed and these results remain controversial. Despite promising results using stem cells in animal models of cardiac injury, the driving mechanisms behind their differentiation down a cardiomyogenic pathway have yet to be determined. Currently, there is a paucity of information regarding cardiomyogenesis on the systemic level. Stem cell differentiation results from multiple signaling parameters operating in a tightly regulated spatiotemporal pattern. Investigating this phenomenon from a systems biology perspective could unveil the abstruse mechanisms controlling cardiomyogenesis that would otherwise require extensive in vitro testing.
Collapse
Affiliation(s)
- D Adam Young
- Department of Bioengineering, University of California, San Diego, CA, USA
| | | | | |
Collapse
|
37
|
Mandel EM, Kaltenbrun E, Callis TE, Zeng XXI, Marques SR, Yelon D, Wang DZ, Conlon FL. The BMP pathway acts to directly regulate Tbx20 in the developing heart. Development 2010; 137:1919-29. [PMID: 20460370 DOI: 10.1242/dev.043588] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
TBX20 has been shown to be essential for vertebrate heart development. Mutations within the TBX20 coding region are associated with human congenital heart disease, and the loss of Tbx20 in a wide variety of model systems leads to cardiac defects and eventually heart failure. Despite the crucial role of TBX20 in a range of cardiac cellular processes, the signal transduction pathways that act upstream of Tbx20 remain unknown. Here, we have identified and characterized a conserved 334 bp Tbx20 cardiac regulatory element that is directly activated by the BMP/SMAD1 signaling pathway. We demonstrate that this element is both necessary and sufficient to drive cardiac-specific expression of Tbx20 in Xenopus, and that blocking SMAD1 signaling in vivo specifically abolishes transcription of Tbx20, but not that of other cardiac factors, such as Tbx5 and MHC, in the developing heart. We further demonstrate that activation of Tbx20 by SMAD1 is mediated by a set of novel, non-canonical, high-affinity SMAD-binding sites located within this regulatory element and that phospho-SMAD1 directly binds a non-canonical SMAD1 site in vivo. Finally, we show that these non-canonical sites are necessary and sufficient for Tbx20 expression in Xenopus, and that reporter constructs containing these sites are expressed in a cardiac-specific manner in zebrafish and mouse. Collectively, our findings define Tbx20 as a direct transcriptional target of the BMP/SMAD1 signaling pathway during cardiac maturation.
Collapse
Affiliation(s)
- Elizabeth M Mandel
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | |
Collapse
|