1
|
Shi J, Jin Y, Lin S, Li X, Zhang D, Wu J, Qi Y, Li Y. Mitochondrial non-energetic function and embryonic cardiac development. Front Cell Dev Biol 2024; 12:1475603. [PMID: 39435335 PMCID: PMC11491369 DOI: 10.3389/fcell.2024.1475603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 09/20/2024] [Indexed: 10/23/2024] Open
Abstract
The initial contraction of the heart during the embryonic stage necessitates a substantial energy supply, predominantly derived from mitochondrial function. However, during embryonic heart development, mitochondria influence beyond energy supplementation. Increasing evidence suggests that mitochondrial permeability transition pore opening and closing, mitochondrial fusion and fission, mitophagy, reactive oxygen species production, apoptosis regulation, Ca2+ homeostasis, and cellular redox state also play critical roles in early cardiac development. Therefore, this review aims to describe the essential roles of mitochondrial non-energetic function embryonic cardiac development.
Collapse
Affiliation(s)
- Jingxian Shi
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuxi Jin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sha Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xing Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Jinlin Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Qi
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Li S, Tao G. Perish in the Attempt: Regulated Cell Death in Regenerative and Nonregenerative Tissue. Antioxid Redox Signal 2023; 39:1053-1069. [PMID: 37218435 PMCID: PMC10715443 DOI: 10.1089/ars.2022.0166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/24/2023]
Abstract
Significance: A cell plays its roles throughout its life span, even during its demise. Regulated cell death (RCD) is one of the key topics in modern biomedical studies. It is considered the main approach for removing stressed and/or damaged cells. Research during the past two decades revealed more roles of RCD, such as coordinating tissue development and driving compensatory proliferation during tissue repair. Recent Advances: Compensatory proliferation, initially identified in primitive organisms during the regeneration of lost tissue, is an evolutionarily conserved process that also functions in mammals. Among various types of RCD, apoptosis is considered the top candidate to induce compensatory proliferation in damaged tissue. Critical Issues: The roles of apoptosis in the recovery of nonregenerative tissue are still vague. The roles of other types of RCD, such as necroptosis and ferroptosis, have not been well characterized in the context of tissue regeneration. Future Directions: In this review article, we attempt to summarize the recent insights on the role of RCD in tissue repair. We focus on apoptosis, with expansion to ferroptosis and necroptosis, in primitive organisms with significant regenerative capacity as well as common mammalian research models. After gathering hints from regenerative tissue, in the second half of the review, we take a notoriously nonregenerative tissue, the myocardium, as an example to discuss the role of RCD in terminally differentiated quiescent cells. Antioxid. Redox Signal. 39, 1053-1069.
Collapse
Affiliation(s)
- Shuang Li
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
3
|
Boulet F, Odelin G, Harrington A, Moore-Morris T. Nipbl Haploinsufficiency Leads to Delayed Outflow Tract Septation and Aortic Valve Thickening. Int J Mol Sci 2023; 24:15564. [PMID: 37958548 PMCID: PMC10648932 DOI: 10.3390/ijms242115564] [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: 08/31/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023] Open
Abstract
Cornelia de Lange Syndrome (CdLS) patients, who frequently carry a mutation in NIPBL, present an increased incidence of outflow tract (OFT)-related congenital heart defects (CHDs). Nipbl+/- mice recapitulate a number of phenotypic traits of CdLS patients, including a small body size and cardiac defects, but no study has specifically focused on the valves. Here, we show that adult Nipbl+/- mice present aortic valve thickening, a condition that has been associated with stenosis. During development, we observed that OFT septation and neural crest cell condensation was delayed in Nipbl+/- embryos. However, we did not observe defects in the deployment of the main lineages contributing to the semilunar valves. Indeed, endocardial endothelial-to-mesenchymal transition (EndMT), analysed via outflow tract explants, and neural crest migration, analysed via genetic lineage tracing, did not significantly differ in Nipbl+/- mice and their wild-type littermates. Our study provides the first direct evidence for valve formation defects in Nipbl+/- mice and points to specific developmental defects as an origin for valve disease in patients.
Collapse
Affiliation(s)
- Fanny Boulet
- Institut de Génomique Fonctionnelle, University of Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, 34094 Montpellier, France
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Gaelle Odelin
- Aix Marseille University, INSERM, MMG, 13005 Marseille, France
| | - Alenca Harrington
- Institut de Génomique Fonctionnelle, University of Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, 34094 Montpellier, France
| | - Thomas Moore-Morris
- Institut de Génomique Fonctionnelle, University of Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, 34094 Montpellier, France
| |
Collapse
|
4
|
First-Trimester Maternal Folic Acid Supplementation Modifies the Effects of Risk Factors Exposures on Congenital Heart Disease in Offspring. Life (Basel) 2021; 11:life11080724. [PMID: 34440469 PMCID: PMC8399198 DOI: 10.3390/life11080724] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/14/2021] [Accepted: 07/18/2021] [Indexed: 02/06/2023] Open
Abstract
This study aimed to examine effect modification of maternal risk factor exposures and congenital heart disease (CHD) by maternal folic acid supplementation (FAS)/non-FAS. We included 8379 CHD cases and 6918 CHD-free controls from 40 clinical centers in Guangdong Province, Southern China, 2004-2016. Controls were randomly chosen from malformation-free fetuses and infants and frequency matched to the echocardiogram-confirmed cases by enrollment hospital and year of birth. We used multiple regression models to evaluate interactions between FAS/non-FAS and risk factors on CHDs and major CHD categories, adjusted for confounding variables. We detected statistically significant additive and multiplicative interactions between maternal FAS/non-FAS and first-trimester fever, viral infection, and threatened abortion on CHDs. An additive interaction on CHDs was also identified between non-FAS and living in a newly renovated home. We observed a statistically significant dose-response relationship between non-FAS and a greater number of maternal risk factors on CHDs. Non-FAS and maternal risk factors interacted additively on multiple critical CHDs, conotruncal defects, and right ventricular outflow tract obstruction. Maternal risk factor exposures may have differential associations with CHD risk in offspring, according to FAS. These findings may inform the design of targeted interventions to prevent CHDs in highly susceptible population groups.
Collapse
|
5
|
Abstract
For more than 2000 years, the avian embryo has helped scientists understand questions of developmental and cell biology. As early as 350 BC Aristotle described embryonic development inside a chicken egg (Aristotle, Generation of animals. Loeb Classical Library (translated), vol. 8, 1943). In the seventeenth century, Marcello Malpighi, referred to as the father of embryology, first diagramed the microscopic morphogenesis of the chick embryo, including extensive characterization of the cardiovascular system (Pearce Eur Neurol 58(4):253-255, 2007; West, Am J Physiol Lung Cell Mol Physiol 304(6):L383-L390, 2016). The ease of accessibility to the embryo and similarity to mammalian development have made avians a powerful system among model organisms. Currently, a unique combination of classical and modern techniques is employed for investigation of the vascular system in the avian embryo. Here, we will introduce the essential techniques of embryonic manipulation for experimental study in vascular biology.
Collapse
Affiliation(s)
- Rieko Asai
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Michael Bressan
- Department of Cell Biology and Physiology, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Takashi Mikawa
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
6
|
Llurba Olive E, Xiao E, Natale DR, Fisher SA. Oxygen and lack of oxygen in fetal and placental development, feto-placental coupling, and congenital heart defects. Birth Defects Res 2019; 110:1517-1530. [PMID: 30576091 DOI: 10.1002/bdr2.1430] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022]
Abstract
Low oxygen concentration (hypoxia) is part of normal embryonic development, yet the situation is complex. Oxygen (O2 ) is a janus gas with low levels signaling through hypoxia-inducible transcription factor (HIF) that are required for development of fetal and placental vasculature and fetal red blood cells. This results in coupling of fetus and mother around midgestation as a functional feto-placental unit (FPU) for O2 transport, which is required for continued growth and development of the fetus. Defects in these processes may leave the developing fetus vulnerable to O2 deprivation or other stressors during this critical midgestational transition when common septal and conotruncal heart defects (CHDs) are likely to arise. Recent human epidemiological and case-control studies support an association between placental dysfunction, manifest as early onset pre-eclampsia (PE) and increased serum bio-markers, and CHD. Animal studies support this association, in particular those using gene inactivation in the mouse. Sophisticated methods for gene inactivation, cell fate mapping, and a quantitative bio-reporter of O2 concentration support the premise that hypoxic stress at critical stages of development leads to CHD. The secondary heart field contributing to the cardiac outlet is a key target, with activation of the un-folded protein response and abrogation of FGF signaling or precocious activation of a cardiomyocyte transcriptional program for differentiation, suggested as mechanisms. These studies provide a strong foundation for further study of feto-placental coupling and hypoxic stress in the genesis of human CHD.
Collapse
Affiliation(s)
- Elisa Llurba Olive
- Director of the Obstetrics and Gynecology Department, Sant Pau University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain.,Maternal and Child Health and Development Network II (SAMID II) RD16/0022, Institute of Health Carlos III, Madrid, Spain
| | - Emily Xiao
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland
| | - David R Natale
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California San Diego, San Diego, California
| | - Steven A Fisher
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Physiology and Biophysics, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
7
|
Yang Q, Wu F, Wang F, Cai K, Zhang Y, Sun Q, Zhao X, Gui Y, Li Q. Impact of DNA methyltransferase inhibitor 5-azacytidine on cardiac development of zebrafish in vivo and cardiomyocyte proliferation, apoptosis, and the homeostasis of gene expression in vitro. J Cell Biochem 2019; 120:17459-17471. [PMID: 31271227 DOI: 10.1002/jcb.29010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 12/15/2022]
Abstract
Cardiac development is a peculiar process involving coordinated cellular differentiation, migration, proliferation, and apoptosis. DNA methylation plays a key role in genomic stability, tissue-specific gene expression, cell proliferation, and apoptosis. Hypomethylation in the global genome has been reported in cardiovascular diseases. However, little is known about the impact and specific mechanism of global hypomethylation on cardiomyocytes. In the present study, we explored the impact of DNA methyltransferase inhibitors 5-azacytidine on cardiac development. In vivo experiment showed that hypomethylation of zebrafish embryos with 5-azacytidine exposure significantly reduced survival, induced malformations, and delayed general development process. Furthermore, zebrafish embryos injected with 5-azacytidine developed pericardial edema, ventricular volume reduction, looping deformity, and reduction in heart rate and ventricular shortening fraction. Cardiomyocytes treated with 5-azacytidine in vitro decreased proliferation and induced apoptosis in a concentration-dependent manner. Furthermore, 5-azacytidine treatment in cardiomyocytes resulted in 20 downregulated genes expression and two upregulated genes expression in 45 candidate genes, which indicated that DNA methylation functions as a bidirectional modulator in regulating gene expression. In conclusion, these results show the regulative effects of the epigenetic modifier 5-azacytidine in cardiac development of zebrafish embryos in vivo and cardiomyocyte proliferation and apoptosis and the homeostasis of gene expression in vitro, which offer a novel understanding of aberrant DNA methylation in the etiology of cardiovascular disease.
Collapse
Affiliation(s)
- Qian Yang
- Shanghai Key Laboratory of Birth Defect, Translational Medical Center for Development and Disease, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Fang Wu
- Shanghai Key Laboratory of Birth Defect, Translational Medical Center for Development and Disease, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Feng Wang
- Shanghai Key Laboratory of Birth Defect, Translational Medical Center for Development and Disease, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Ke Cai
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Yawen Zhang
- Shanghai Key Laboratory of Birth Defect, Translational Medical Center for Development and Disease, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Quanya Sun
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaolong Zhao
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yonghao Gui
- Shanghai Key Laboratory of Birth Defect, Translational Medical Center for Development and Disease, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Qiang Li
- Shanghai Key Laboratory of Birth Defect, Translational Medical Center for Development and Disease, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
8
|
Zhao Q, Sun Q, Zhou L, Liu K, Jiao K. Complex Regulation of Mitochondrial Function During Cardiac Development. J Am Heart Assoc 2019; 8:e012731. [PMID: 31215339 PMCID: PMC6662350 DOI: 10.1161/jaha.119.012731] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Qiancong Zhao
- Department of Cardiovascular SurgeryThe Second Hospital of Jilin UniversityChangchunChina
- Department of GeneticsThe University of Alabama at BirminghamAL
| | - Qianchuang Sun
- Department of AnesthesiologyThe Second Hospital of Jilin UniversityChangchunChina
- Department of GeneticsThe University of Alabama at BirminghamAL
| | - Lufang Zhou
- Department of MedicineThe University of Alabama at BirminghamAL
| | - Kexiang Liu
- Department of Cardiovascular SurgeryThe Second Hospital of Jilin UniversityChangchunChina
| | - Kai Jiao
- Department of GeneticsThe University of Alabama at BirminghamAL
| |
Collapse
|
9
|
Lai T, Xiang L, Liu Z, Mu Y, Li X, Li N, Li S, Chen X, Yang J, Tao J, Zhu J. Association of maternal disease and medication use with the risk of congenital heart defects in offspring: a case-control study using logistic regression with a random-effects model. J Perinat Med 2019; 47:455-463. [PMID: 30794526 DOI: 10.1515/jpm-2018-0281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/21/2019] [Indexed: 01/29/2023]
Abstract
Objective To examine the association between maternal diseases and congenital heart defects (CHDs) and to evaluate whether those associations vary with corresponding medication use. Methods A multi-hospital case-control study conducted from February 2010 to December 2014 analysed 916 controls and 1236 cases. Participating mothers were asked whether they suffered from influenza, common cold, herpes and threatened abortion or had used corresponding medication during the periconception period or the early pregnancy period. We used a random-effects logistic regression model to compute the odds ratios (ORs), adjusted odds ratios (AORs) and 95% confidence intervals (CIs) while controlling for potential confounders. Results Compared with the results for mothers with no exposure, there were significant associations between maternal diseases with medication non-use and CHDs in the aggregate, including influenza (AOR, 1.83; 95% CI, 1.13-2.95), common cold (AOR, 2.05; 95% CI, 1.60-2.64) and herpes (AOR, 7.00; 95% CI, 2.15-22.84). There was no significant association between medication users and offspring with any subtype of CHDs, except that maternal common cold with medication use slightly increased the risk of the specific subtype, namely, isolated cardiac defects. However, an association was observed between maternal threatened abortion and medication and isolated cardiac defects (AOR, 1.33; 95% CI, 1.01-1.75). Conclusion Maternal influenza, common cold, herpes and threatened abortion from 3 months before pregnancy through the first trimester were associated with an increased risk of congenital heart disease in offspring. The teratogenic effect of these conditions may be attenuated by medication use, except for threatened abortion.
Collapse
Affiliation(s)
- Ting Lai
- West China School of Public Health, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Liangcheng Xiang
- National Center for Birth Defect Monitoring, Department of Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Zhen Liu
- National Center for Birth Defect Monitoring, Department of Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sec. 3 No. 17, South RenMin Road, Chengdu, Sichuan, P.R. China
| | - Yi Mu
- National Center for Birth Defect Monitoring, Department of Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Xiaohong Li
- National Center for Birth Defect Monitoring, Department of Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Nana Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sec. 3 No. 17, South RenMin Road, Chengdu, Sichuan, P.R. China
| | - Shengli Li
- Department of Ultrasound, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, P.R. China
| | - Xinlin Chen
- Department of Ultrasound, Hubei Maternity and Child Healthcare Hospital, Wuhan, Hubei, P.R. China
| | - Jiaxiang Yang
- Department of Ultrasound, Sichuan Maternity and Child Healthcare Hospital, Chengdu, P.R. China
| | - Jing Tao
- National Center for Birth Defect Monitoring, Department of Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China, Tel.: +86-028-85501362
| | - Jun Zhu
- National Center for Birth Defect Monitoring, Department of Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sec. 3 No. 17, South RenMin Road, Chengdu, Sichuan 610041, P.R. China, Tel.: +86-028-85503121
| |
Collapse
|
10
|
Lorda-Diez CI, Solis-Mancilla ME, Sanchez-Fernandez C, Garcia-Porrero JA, Hurle JM, Montero JA. Cell senescence, apoptosis and DNA damage cooperate in the remodeling processes accounting for heart morphogenesis. J Anat 2019; 234:815-829. [PMID: 30875434 PMCID: PMC6539749 DOI: 10.1111/joa.12972] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2019] [Indexed: 01/10/2023] Open
Abstract
During embryonic development, organ morphogenesis requires major tissue rearrangements that are tightly regulated at the genetic level. A large number of studies performed in recent decades assigned a central role to programmed cell death for such morphogenetic tissue rearrangements that often sculpt the shape of embryonic organs. However, accumulating evidence indicates that far from being the only factor responsible for sculpting organ morphology, programmed cell death is accompanied by other tissue remodeling events that ensure the outcome of morphogenesis. In this regard, cell senescence has been recently associated with morphogenetic degenerative embryonic processes as an early tissue remodeling event in development of the limbs, kidney and inner ear. Here, we have explored cell senescence by monitoring β‐galactosidase activity during embryonic heart development where programmed cell death is believed to exert an important morphogenetic function. We report the occurrence of extensive cell senescence foci during heart morphogenesis. These foci overlap spatially and temporally with the areas of programmed cell death that are associated with remodeling of the outflow tract to build the roots of the great arteries and with the septation of cardiac cavities. qPCR analysis allowed us to identify a gene expression profile characteristic of the so‐called senescence secretory associated phenotype in the remodeling outflow tract of the embryonic heart. In addition, we confirmed local upregulation of numerous tumor suppressor genes including p21, p53, p63, p73 and Btg2. Interestingly, the areas of cell senescence were also accompanied by intense lysosomal activation and non‐apoptotic DNA damage revealed by γH2AX immunolabeling. Considering the importance of sustained DNA damage as a triggering factor for cell senescence and apoptosis, we propose the coordinated contribution of DNA damage, senescence and apoptotic cell death to assure tissue remodeling in the developing vertebrate heart.
Collapse
Affiliation(s)
- Carlos I Lorda-Diez
- Facultad de Medicina, Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Michelle E Solis-Mancilla
- Facultad de Medicina, Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Cristina Sanchez-Fernandez
- Facultad de Medicina, Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Juan A Garcia-Porrero
- Facultad de Medicina, Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Juan M Hurle
- Facultad de Medicina, Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Juan A Montero
- Facultad de Medicina, Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, Santander, Spain
| |
Collapse
|
11
|
Lazzarini R, Gómez-Quiroz LE, González-Márquez H, Villavicencio-Guzmán L, Salazar-García M, Sánchez-Gómez C. The proximal segment of the embryonic outflow (conus) does not participate in aortic vestibule development. PLoS One 2018; 13:e0209930. [PMID: 30596770 PMCID: PMC6312233 DOI: 10.1371/journal.pone.0209930] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/13/2018] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE There is no consensus on the embryonic components or morphogenetic processes involved in mature ventricular outflow tract development. Our goal was to use in vivo labelling to investigate the prospective fate of the myocardium of each conal wall. The conal and atrioventricular cushion mesenchyme changes during transformation into mature structures and their role in apoptosis were also investigated. METHODS Plastic labels were placed at the cephalic and caudal conal limits of chicken embryo hearts (stage 22HH) and traced up to stage 36HH. Histological analyses, scanning electron microscopy and apoptotic detection using Lysotracker-Red were performed. The conal longitudinal length and medial displacement were registered. Muscle myosin was identified by immunofluorescence. RESULTS Labels positioned in the myocardium of each conal wall moved to the right ventricle (RV), shifting from the arterial subvalvular myocardial zone to the apex. No labels were found in the aortic vestibule. At stage 22HH, the conus was a tubular structure composed of myocardium and endocardium with scarce mesenchyme. The dorso-left conal myocardial wall gradually lost continuity and the free ends separated, while the myocardium was distributed to the RV free wall (24HH-28HH). At stage 22HH, conal crests were not observed, but they were apparent at the dorsal zone of the conus at stage 26HH; towards stage 30HH, they fused to form the supraventricular crest, and the pulmonary infundibulum was evident. The ventro-superior cushion of the AV canal was reorganized into the fibrous and muscular structures lined the aortic vestibule. CONCLUSIONS The posterior conus is an erroneous concept. The conal myocardium is reorganized in the free wall of the RV. Internally, the conal lumen is transformed into the pulmonary infundibulum. The aortic vestibule is formed from the ventro-superior cushion of the AV canal. Thus, the ventricular outflow tracts have different embryonic origins.
Collapse
Affiliation(s)
- Roberto Lazzarini
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México, México
| | - Luis Enrique Gómez-Quiroz
- Departamento Ciencias de la Salud, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México, México
| | - Humberto González-Márquez
- Departamento Ciencias de la Salud, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México, México
| | - Laura Villavicencio-Guzmán
- Laboratorio de Investigación en Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México, Federico Gómez, Ciudad de México, México
| | - Marcela Salazar-García
- Laboratorio de Investigación en Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México, Federico Gómez, Ciudad de México, México
| | - Concepción Sánchez-Gómez
- Laboratorio de Investigación en Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México, Federico Gómez, Ciudad de México, México
| |
Collapse
|
12
|
Paffett-Lugassy N, Novikov N, Jeffrey S, Abrial M, Guner-Ataman B, Sakthivel S, Burns CE, Burns CG. Unique developmental trajectories and genetic regulation of ventricular and outflow tract progenitors in the zebrafish second heart field. Development 2017; 144:4616-4624. [PMID: 29061637 DOI: 10.1242/dev.153411] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 10/11/2017] [Indexed: 02/03/2023]
Abstract
During mammalian embryogenesis, cardiac progenitor cells constituting the second heart field (SHF) give rise to the right ventricle and primitive outflow tract (OFT). In zebrafish, previous lineage-tracing and mutant analyses suggested that SHF ventricular and OFT progenitors co-migrate to the arterial pole of the zebrafish heart tube soon after their specification in the nkx2.5+ field of anterior lateral plate mesoderm (ALPM). Using additional prospective lineage tracing, we demonstrate that while SHF ventricular progenitors migrate directly to the arterial pole, OFT progenitors become temporarily sequestered in the mesodermal cores of pharyngeal arch 2 (PA2), where they downregulate nkx2.5 expression. While there, they intermingle with precursors for PA2-derived head muscles (HMs) and hypobranchial artery endothelium, which we demonstrate are co-specified with SHF progenitors in the nkx2.5+ ALPM. Soon after their sequestration in PA2, OFT progenitors migrate to the arterial pole of the heart and differentiate into OFT lineages. Lastly, we demonstrate that SHF ventricular and OFT progenitors exhibit unique sensitivities to a mutation in fgf8a Our data highlight novel aspects of SHF, OFT and HM development in zebrafish that will inform mechanistic interpretations of cardiopharyngeal phenotypes in zebrafish models of human congenital disorders.
Collapse
Affiliation(s)
- Noelle Paffett-Lugassy
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA.,Harvard Medical School, Boston, MA 02115, USA
| | - Natasha Novikov
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA.,Harvard Medical School, Boston, MA 02115, USA
| | - Spencer Jeffrey
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Maryline Abrial
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA.,Harvard Medical School, Boston, MA 02115, USA
| | - Burcu Guner-Ataman
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA.,Harvard Medical School, Boston, MA 02115, USA
| | - Srinivasan Sakthivel
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA.,Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200N, Denmark
| | - Caroline E Burns
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA .,Harvard Medical School, Boston, MA 02115, USA.,Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - C Geoffrey Burns
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA .,Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
13
|
Signaling Pathways in Cardiac Myocyte Apoptosis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9583268. [PMID: 28101515 PMCID: PMC5215135 DOI: 10.1155/2016/9583268] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/20/2016] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases, the number 1 cause of death worldwide, are frequently associated with apoptotic death of cardiac myocytes. Since cardiomyocyte apoptosis is a highly regulated process, pharmacological intervention of apoptosis pathways may represent a promising therapeutic strategy for a number of cardiovascular diseases and disorders including myocardial infarction, ischemia/reperfusion injury, chemotherapy cardiotoxicity, and end-stage heart failure. Despite rapid growth of our knowledge in apoptosis signaling pathways, a clinically applicable treatment targeting this cellular process is currently unavailable. To help identify potential innovative directions for future research, it is necessary to have a full understanding of the apoptotic pathways currently known to be functional in cardiac myocytes. Here, we summarize recent progress in the regulation of cardiomyocyte apoptosis by multiple signaling molecules and pathways, with a focus on the involvement of these pathways in the pathogenesis of heart disease. In addition, we provide an update regarding bench to bedside translation of this knowledge and discuss unanswered questions that need further investigation.
Collapse
|
14
|
Ta-Shma A, Zhang K, Salimova E, Zernecke A, Sieiro-Mosti D, Stegner D, Furtado M, Shaag A, Perles Z, Nieswandt B, Rein AJJT, Rosenthal N, Neiman AM, Elpeleg O. Congenital valvular defects associated with deleterious mutations in the PLD1 gene. J Med Genet 2016; 54:278-286. [PMID: 27799408 DOI: 10.1136/jmedgenet-2016-104259] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 09/28/2016] [Accepted: 09/29/2016] [Indexed: 11/04/2022]
Abstract
BACKGROUND The underlying molecular aetiology of congenital heart defects is largely unknown. The aim of this study was to explore the genetic basis of non-syndromic severe congenital valve malformations in two unrelated families. METHODS Whole-exome analysis was used to identify the mutations in five patients who suffered from severe valvular malformations involving the pulmonic, tricuspid and mitral valves. The significance of the findings was assessed by studying sporulation of yeast carrying a homologous Phospholipase D (PLD1) mutation, in situ hybridisation in chick embryo and echocardiography and histological examination of hearts of PLD1 knockout mice. RESULTS Three mutations, p.His442Pro, p.Thr495fs32* and c.2882+2T>C, were identified in the PLD1 gene. The mutations affected highly conserved sites in the PLD1 protein and the p.His442Pro mutation produced a strong loss of function phenotype in yeast homologous mutant strain. Here we show that in chick embryos PLD1 expression is confined to the forming heart (E2-E8) and homogeneously expressed all over the heart during days E2-E3. Thereafter its expression decreases, remaining only adjacent to the atrioventricular valves and the right ventricular outflow tract. This pattern of expression follows the known dynamic patterning of apoptosis in the developing heart, consistent with the known role of PLD1 in the promotion of apoptosis. In hearts of PLD1 knockout mice, we detected marked tricuspid regurgitation, right atrial enlargement, and increased flow velocity, narrowing and thickened leaflets of the pulmonic valve. CONCLUSIONS The findings support a role for PLD1 in normal heart valvulogenesis.
Collapse
Affiliation(s)
- Asaf Ta-Shma
- Department of Pediatric Cardiology, Hadassah, Hebrew University Medical Center, Jerusalem, Israel.,Monique and Jacques Roboh Department of Genetic Research, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Kai Zhang
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA
| | - Ekaterina Salimova
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Daniel Sieiro-Mosti
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - David Stegner
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Milena Furtado
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Avraham Shaag
- Monique and Jacques Roboh Department of Genetic Research, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Zeev Perles
- Department of Pediatric Cardiology, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Azaria J J T Rein
- Department of Pediatric Cardiology, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Nadia Rosenthal
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Aaron M Neiman
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA
| | - Orly Elpeleg
- Monique and Jacques Roboh Department of Genetic Research, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
15
|
Wu Y, Zhou X, Huang X, Xia Q, Chen Z, Zhang X, Yang D, Geng YJ. Pax8 plays a pivotal role in regulation of cardiomyocyte growth and senescence. J Cell Mol Med 2016; 20:644-54. [PMID: 26781745 PMCID: PMC5125375 DOI: 10.1111/jcmm.12779] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 12/05/2015] [Indexed: 12/18/2022] Open
Abstract
Congenital heart disease (CHD) is a worldwide health problem, particularly in young populations. In spite of the advancement and progress in medical research and technology, the underlying causative factors and mechanisms of CHD still remain unclear. Bone morphogenetic protein receptor IA (ALK3) mediates the development of ventricular septal defect (VSD). We have recently found that paired box gene 8 (Pax8) may be the downstream molecule of ALK3. Paired box gene 8 plays an essential role in VSD, and apoptosis and proliferation imbalance leads to septal dysplasia. Recent studies have also disclosed that cellular senescence also participates in embryonic development. Whether programmed senescence exists in cardiac organogenesis has not ever been reported. We hypothesized that together with various biological processes, such as apoptosis, enhanced cellular senescence may occur actively in the development of Pax8 null mice murine hearts. In H9C2 myogenic cells, Pax8 overexpression can rescue caspase‐dependent apoptosis induced by ALK3 silencing. Senescent cells and senescence‐associated mediators in Pax8 knockout hearts increased compared with the wild‐type ones in an age‐dependent manner. These results suggest that Pax8 maybe the downstream molecule of ALK3, it mediates the murine heart development perhaps via cellular senescence, which may serve as a mechanism that compensates for the cell loss via apoptosis in heart development.
Collapse
Affiliation(s)
- Yihao Wu
- Division of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xi Zhou
- Division of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoyan Huang
- Division of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Quan Xia
- Division of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Zhe Chen
- Division of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Xingwei Zhang
- Division of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Deye Yang
- Division of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Division of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Yong-jian Geng
- The University of Texas School of Medicine at Houston, Houston, TX, USA
| |
Collapse
|
16
|
Abstract
For more than 2,000 years, philosophers and scientists have turned to the avian embryo with questions of how life begins (Aristotle and Peck Generations of Animals. Loeb Classics, vol. XIII. Harvard University Press, Cambridge, 1943; Needham, A history of embryology. Abelard-Schuman, New York, 1959). Then, as now, the unique accessibility of the embryo both in terms of acquisition of eggs from domesticated fowl and ease at which the embryo can be visualized by simply opening the shell has made avians an appealing and powerful model system for the study of development. Thus, as the field of embryology has evolved through observational, comparative, and experimental embryology into its current iteration as the cellular and molecular biology of development, avians have remained a useful and practical system of study.
Collapse
Affiliation(s)
- Michael Bressan
- Cardiovascular Research Institute, University of California, San Francisco, 555 Mission Bay Blvd South, MC3120, San Francisco, CA, 94143-3120, USA,
| | | |
Collapse
|
17
|
Abdelwahid E. Getting the heart into shape by the influence of cell death machinery. Am J Med Genet A 2014; 167A:243-5. [PMID: 25339610 DOI: 10.1002/ajmg.a.36791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 09/03/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Eltyeb Abdelwahid
- Feinberg School of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, Illinois
| |
Collapse
|
18
|
Kowalski WJ, Dur O, Wang Y, Patrick MJ, Tinney JP, Keller BB, Pekkan K. Critical transitions in early embryonic aortic arch patterning and hemodynamics. PLoS One 2013; 8:e60271. [PMID: 23555940 PMCID: PMC3605337 DOI: 10.1371/journal.pone.0060271] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 02/25/2013] [Indexed: 02/01/2023] Open
Abstract
Transformation from the bilaterally symmetric embryonic aortic arches to the mature great vessels is a complex morphogenetic process, requiring both vasculogenic and angiogenic mechanisms. Early aortic arch development occurs simultaneously with rapid changes in pulsatile blood flow, ventricular function, and downstream impedance in both invertebrate and vertebrate species. These dynamic biomechanical environmental landscapes provide critical epigenetic cues for vascular growth and remodeling. In our previous work, we examined hemodynamic loading and aortic arch growth in the chick embryo at Hamburger-Hamilton stages 18 and 24. We provided the first quantitative correlation between wall shear stress (WSS) and aortic arch diameter in the developing embryo, and observed that these two stages contained different aortic arch patterns with no inter-embryo variation. In the present study, we investigate these biomechanical events in the intermediate stage 21 to determine insights into this critical transition. We performed fluorescent dye microinjections to identify aortic arch patterns and measured diameters using both injection recordings and high-resolution optical coherence tomography. Flow and WSS were quantified with 3D computational fluid dynamics (CFD). Dye injections revealed that the transition in aortic arch pattern is not a uniform process and multiple configurations were documented at stage 21. CFD analysis showed that WSS is substantially elevated compared to both the previous (stage 18) and subsequent (stage 24) developmental time-points. These results demonstrate that acute increases in WSS are followed by a period of vascular remodeling to restore normative hemodynamic loading. Fluctuations in blood flow are one possible mechanism that impacts the timing of events such as aortic arch regression and generation, leading to the variable configurations at stage 21. Aortic arch variations noted during normal rapid vascular remodeling at stage 21 identify a temporal window of increased vulnerability to aberrant aortic arch morphogenesis with the potential for profound effects on subsequent cardiovascular morphogenesis.
Collapse
Affiliation(s)
- William J. Kowalski
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Onur Dur
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Yajuan Wang
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Michael J. Patrick
- Molecular Biosensor and Imaging Center, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Joseph P. Tinney
- Department of Pediatrics, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
| | - Bradley B. Keller
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- Department of Pediatrics, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
| | - Kerem Pekkan
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
19
|
Scherptong RWC, Jongbloed MRM, Wisse LJ, Vicente-Steijn R, Bartelings MM, Poelmann RE, Schalij MJ, Gittenberger-De Groot AC. Morphogenesis of outflow tract rotation during cardiac development: the pulmonary push concept. Dev Dyn 2012; 241:1413-22. [PMID: 22826212 DOI: 10.1002/dvdy.23833] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2012] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Understanding of cardiac outflow tract (OFT) remodeling is essential to explain repositioning of the aorta and pulmonary orifice. In wild type embryos (E9.5-14.5), second heart field contribution (SHF) to the OFT was studied using expression patterns of Islet 1, Nkx2.5, MLC-2a, WT-1, and 3D-reconstructions. Abnormal remodeling was studied in VEGF120/120 embryos. RESULTS In wild type, Islet 1 and Nkx2.5 positive myocardial precursors formed an asymmetric elongated column almost exclusively at the pulmonary side of the OFT up to the pulmonary orifice. In VEGF120/120 embryos, the Nkx2.5-positive mesenchymal population was disorganized with a short extension along the pulmonary OFT. CONCLUSIONS We postulate that normally the pulmonary trunk and orifice are pushed in a higher and more frontal position relative to the aortic orifice by asymmetric addition of SHF-myocardium. Deficient or disorganized right ventricular OFT expansion might explain cardiac malformations with abnormal position of the great arteries, such as double outlet right ventricle.
Collapse
|
20
|
Abstract
Ten years ago, a population of cardiac progenitor cells was identified in pharyngeal mesoderm that gives rise to a major part of the amniote heart. These multipotent progenitor cells, termed the second heart field (SHF), contribute progressively to the poles of the elongating heart tube during looping morphogenesis, giving rise to myocardium, smooth muscle, and endothelial cells. Research into the mechanisms of SHF development has contributed significantly to our understanding of the properties of cardiac progenitor cells and the origins of congenital heart defects. Here recent data concerning the regulation, clinically relevant subpopulations, evolution and lineage relationships of the SHF are reviewed. Proliferation and differentiation of SHF cells are controlled by multiple intercellular signaling pathways and a transcriptional regulatory network that is beginning to be elucidated. Perturbation of SHF development results in common forms of congenital heart defects and particular progenitor cell subpopulations are highly relevant clinically, including cells giving rise to myocardium at the base of the pulmonary trunk and the interatrial septum. A SHF has recently been identified in amphibian, fish, and agnathan embryos, highlighting the important contribution of these cells to the evolution of the vertebrate heart. Finally, SHF-derived parts of the heart share a lineage relationship with craniofacial skeletal muscles revealing that these progenitor cells belong to a broad cardiocraniofacial field of pharyngeal mesoderm. Investigation of the mechanisms underlying the dynamic process of SHF deployment is likely to yield further insights into cardiac development and pathology.
Collapse
Affiliation(s)
- Robert G Kelly
- Developmental Biology Institute of Marseilles-Luminy, Aix-Marseille Université, CNRS UMR 7288, Marseilles, France
| |
Collapse
|
21
|
Parisot P, Mesbah K, Théveniau-Ruissy M, Kelly RG. Tbx1, subpulmonary myocardium and conotruncal congenital heart defects. ACTA ACUST UNITED AC 2011; 91:477-84. [PMID: 21591244 DOI: 10.1002/bdra.20803] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 01/25/2011] [Accepted: 02/09/2011] [Indexed: 12/22/2022]
Abstract
Conotruncal congenital heart defects, including defects in septation and alignment of the ventricular outlets, account for approximately a third of all congenital heart defects. Failure of the left ventricle to obtain an independent outlet results in incomplete separation of systemic and pulmonary circulation at birth. The embryonic outflow tract, a transient cylinder of myocardium connecting the embryonic ventricles to the aortic sac, plays a critical role in this process during normal development. The outflow tract (OFT) is derived from a population of cardiac progenitor cells called the second heart field that contributes to the arterial pole of the heart tube during cardiac looping. During septation, the OFT is remodeled to form the base of the ascending aorta and pulmonary trunk. Tbx1, the major candidate gene for DiGeorge syndrome, is a critical transcriptional regulator of second heart field development. DiGeorge syndrome patients are haploinsufficient for Tbx1 and present a spectrum of conotruncal anomalies including tetralogy of Fallot, pulmonary atresia, and common arterial trunk. In this review, we focus on the role of Tbx1 in the regulation of second heart field deployment and, in particular, in the development of a specific population of myocardial cells at the base of the pulmonary trunk. Recent data characterizing additional properties and regulators of development of this region of the heart, including the retinoic acid, hedgehog, and semaphorin signaling pathways, are discussed. These findings identify future subpulmonary myocardium as the clinically relevant component of the second heart field and provide new mechanistic insight into a spectrum of common conotruncal congenital heart defects.
Collapse
Affiliation(s)
- Pauline Parisot
- Developmental Biology Institute of Marseilles-Luminy, UMR 6216/CNRS, Université de la Méditerranée, Marseilles, France
| | | | | | | |
Collapse
|
22
|
Overexpression of Jazf1 induces cardiac malformation through the upregulation of pro-apoptotic genes in mice. Transgenic Res 2011; 20:1019-31. [DOI: 10.1007/s11248-010-9476-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Accepted: 12/15/2010] [Indexed: 11/25/2022]
|
23
|
Yang D, Lai D, Huang X, Shi X, Gao Z, Huang F, Zhou X, Geng YJ. The defects in development and apoptosis of cardiomyocytes in mice lacking the transcriptional factor Pax-8. Int J Cardiol 2010; 154:43-51. [PMID: 20851479 DOI: 10.1016/j.ijcard.2010.08.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Revised: 05/31/2010] [Accepted: 08/19/2010] [Indexed: 11/29/2022]
Abstract
BACKGROUND Cardiac-specific deletion of ALK3 is lethal in mid-gestation with ventricular septum malformations (VSM). This study was designed to define the Pax-8's role in heart development and cardiomyocyte apoptosis. METHODS Pathologic changes in the hearts of Pax-8 or ALK3 knockout and wild type control mice were determined by light and electron microscopy. Analysis of cardiomyocyte apoptosis was performed by TUNEL. The effect of Pax-8 gene deficiency on caspase-3 activity was examined after transfecting Pax-8 siRNA into cultured myoblast cell line. RESULTS Mice with ALK3 or Pax-8 gene knockout but not wild type control animals showed the development of VSM. Increased cardiomyocyte apoptosis was found in homozygotes. Echocardiography showed that Pax-8 homozygote mice developed malfunction of the heart. Furthermore, the caspase-3 activity was significantly higher in the cells treated with Pax-8 siRNA as compared to those treated with negative control siRNA in H9C2 (2-1) cell line. CONCLUSIONS The Pax-8 gene may play a crucial role in heart development and regulating cardiocyte apoptosis. Knockout of Pax-8 may exert a similar effect on myocardial morphology and apoptosis as those seen in ALK3 knockouts. Furthermore, the ventricular septum malformations could be partially attributed to accelerated cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Deye Yang
- The First Affiliated Hospital of Wenzhou Medical College, Wenzhou 325000, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Okamoto N, Akimoto N, Hidaka N, Shoji S, Sumida H. Formal genesis of the outflow tracts of the heart revisited: previous works in the light of recent observations. Congenit Anom (Kyoto) 2010; 50:141-58. [PMID: 20608949 DOI: 10.1111/j.1741-4520.2010.00286.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The formal genesis of the great arteries continues to be controversial due to the lack of consensus of septation of the developing outflow tract. In order to make it clear how the great arteries are generated, we have re-examined our previous papers which emphasized the formation of the aorta and pulmonary trunk, concept of the aorticopulmonary septum, formation of the leaflets of semilunar valves, morphogenesis of the crista supraventricularis, programmed cell death and rotation of the outflow tract. In the present paper, we compare outcomes gained from the re-examination of our previous papers with prevalent interpretations of the arterial trunk. We obtained conclusions as follows: (i) The elongation of the fourth and sixth aortic arch arteries, which sprout from the wall of the aortic sac at the expense of the distal truncus, contributes to the formation of the aorta and pulmonary trunk; (ii) Smooth muscle cells of the tunica media of the arterial trunks do not arise from the transformation of the myocardial cells of the truncus wall (not 'arterialization'); (iii) Truncus swellings are divided into two parts: distal and proximal. The former contributes to the separation of the orifices of arterial trunks ('aorticopulmonary septum'). The latter contributes to the formation of the leaflets of the semilunar valves of the aorta and pulmonary trunk; (iv) The origin of the myocardial cells of the crista supraventricularis is a wall of the conus originated from secondary/anterior heart fields; and (v) There has been no acceptable proof that rotation and counterclockwise rotation are involved.
Collapse
Affiliation(s)
- Naomasa Okamoto
- Hiroshima University and Miyazaki University, Hiroshima, Japan
| | | | | | | | | |
Collapse
|
25
|
Uriu-Adams JY, Scherr RE, Lanoue L, Keen CL. Influence of copper on early development: prenatal and postnatal considerations. Biofactors 2010; 36:136-52. [PMID: 20232410 DOI: 10.1002/biof.85] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Copper (Cu) is an essential nutrient whose requirement is increased during pregnancy and lactation. These represent times of critical growth and development, and the fetus and neonate are particularly vulnerable to deficiencies of this nutrient. Genetic mutations that predispose the offspring to inadequate stores of Cu can be life threatening as is observed in children with Menkes disease. During the last decade, severe Cu deficiency, once thought to be a rare condition, has been reported in the literature at an increasing frequency. Secondary Cu deficiencies can be induced by a variety of ways such as excessive zinc or iron intake, certain drugs, and bariatric surgery. Premature and low birth weight infants can be born with low Cu stores. A number of mechanisms can contribute to the teratogenicity of Cu including decreased activity of select cuproenzymes, increased oxidative stress, decreased nitric oxide availability, altered iron metabolism, abnormal extracellular matrix protein crosslinking, decreased angiogenesis and altered cell signaling among others. The brain, heart, and vessels as well as tissues such as lung, skin and hair, and systems including the skeletal, immune, and blood systems, are negatively affected by suboptimal Cu during development. Additionally, persistent structural, biochemical, and functional adverse effects in the offspring are noted even when Cu supplementation is initiated after birth, supporting the concept that adequate Cu nutriture during pregnancy and lactation is critical for normal development. Although Cu-containing IUDs are an effective method for increasing intrauterine Cu concentrations and for reducing the risk of pregnancy, high amounts of dietary Cu are not thought to represent a direct developmental risk.
Collapse
Affiliation(s)
- Janet Y Uriu-Adams
- Department of Nutrition, University of California, Davis, Davis, CA 95616, USA.
| | | | | | | |
Collapse
|
26
|
Wikenheiser J, Wolfram JA, Gargesha M, Yang K, Karunamuni G, Wilson DL, Semenza GL, Agani F, Fisher SA, Ward N, Watanabe M. Altered hypoxia-inducible factor-1 alpha expression levels correlate with coronary vessel anomalies. Dev Dyn 2010; 238:2688-700. [PMID: 19777592 DOI: 10.1002/dvdy.22089] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The outflow tract myocardium and other regions corresponding to the location of the major coronary vessels of the developing chicken heart, display a high level of hypoxia as assessed by the hypoxia indicator EF5. The EF5-positive tissues were also specifically positive for nuclear-localized hypoxia inducible factor-1 alpha (HIF-1alpha), the oxygen-sensitive component of the hypoxia inducible factor-1 (HIF-1) heterodimer. This led to our hypothesis that there is a "template" of hypoxic tissue that determines the stereotyped pattern of the major coronary vessels. In this study, we disturbed this template by altering ambient oxygen levels (hypoxia 15%; hyperoxia 75-40%) during the early phases of avian coronary vessel development, in order to alter tissue hypoxia, HIF-1alpha protein expression, and its downstream target genes without high mortality. We also altered HIF-1alpha gene expression in the embryonic outflow tract cardiomyocytes by injecting an adenovirus containing a constitutively active form of HIF-1alpha (AdCA5). We assayed for coronary anomalies using anti-alpha-smooth muscle actin immunohistology. When incubated under abnormal oxygen levels or injected with a low titer of the AdCA5, coronary arteries displayed deviations from their normal proximal connections to the aorta. These deviations were similar to known clinical anomalies of coronary arteries. These findings indicated that developing coronary vessels may be subject to a level of regulation that is dependent on differential oxygen levels within cardiac tissues and subsequent HIF-1 regulation of gene expression.
Collapse
Affiliation(s)
- Jamie Wikenheiser
- Department of Pediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, Ohio 44106-6011, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
In humans, congenital heart diseases are common. Since the rapid progression of transgenic technologies, the mouse has become the major animal model of defective cardiovascular development. Moreover, genetically modified mice frequently die in utero, commonly due to abnormal cardiovascular development. A variety of publications address specific developmental stages or structures of the mouse heart, but a single reference reviewing and describing the anatomy and histology of cardiac developmental events, stage by stage, has not been available. The aim of this color atlas, which demonstrates embryonic/fetal heart development, is to provide a tool for pathologists and biomedical scientists to use for detailed histological evaluation of hematoxylin and eosin (H&E)-stained sections of the developing mouse heart with emphasis on embryonic days (E) 11.5-18.5. The selected images illustrate the main structures and developmental events at each stage and serve as reference material for the confirmation of the chronological age of the embryo/early fetus and assist in the identification of any abnormalities. An extensive review of the literature covering cardiac development pre-E11.5 is summarized in the introduction. Although the focus of this atlas is on the descriptive anatomic and histological development of the normal mouse heart from E11.5 to E18.5, potential embryonic cardiac lesions are discussed with a list of the most common transgenic pre- and perinatal heart defects. Representative images of hearts at E11.5-15.5 and E18.5 are provided in Figures 2-4, 6, 8, and 9. A complete set of labeled images (Figures E11.5-18.5) is available on the CD enclosed in this issue of Toxicologic Pathology. All digital images can be viewed online at https://niehsimages.epl-inc.com with the username "ToxPath" and the password "embryohearts."
Collapse
Affiliation(s)
- Saija M Savolainen
- NIEHS, Cellular and Molecular Pathology Branch, Research Triangle Park, North Carolina 27709, USA
| | | | | |
Collapse
|
28
|
MacGowan GA. Good news for mice with heart attacks: preventing acute myocardial injury by inhibiting apoptosis. Cardiovasc Res 2008; 81:1-2. [DOI: 10.1093/cvr/cvn300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
29
|
Kumar SD, Yong SK, Dheen ST, Bay BH, Tay SSW. Cardiac malformations are associated with altered expression of vascular endothelial growth factor and endothelial nitric oxide synthase genes in embryos of diabetic mice. Exp Biol Med (Maywood) 2008; 233:1421-32. [PMID: 18824721 DOI: 10.3181/0806-rm-186] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The aim of this study was to investigate the role of nitric oxide (NO), and the expression of endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF) genes in developing hearts at embryonic day 13.5 of embryos from diabetic mice. The protein and mRNA expression levels of eNOS and VEGF were significantly altered in the developing hearts of embryos from diabetic mice. The NO level was significantly decreased, whereas the VEGF concentration was significantly increased in the developing hearts of the embryos from diabetic mice. In vitro study showed a significant reduction in eNOS expression and cell proliferation in cardiac myoblast cells exposed to high glucose concentrations. Further, high glucose induced apoptosis in myoblast cells. Ultrastructural changes characteristics of apoptosis, including cell blebbing, aggregation of ribosomes and vacuoles in the cytoplasm were also evident in myoblast cells exposed to high glucose. It is suggested that hyperglycemia alters the expression of eNOS and VEGF genes that are involved in the regulation of cell growth and vasculogenesis, thereby contributing to the cardiac malformations seen in embryos from diabetic mice.
Collapse
Affiliation(s)
- Srinivasan Dinesh Kumar
- Department of Anatomy, National University Health System, National University of Singapore, Singapore 117597, Singapore
| | | | | | | | | |
Collapse
|
30
|
Liu H, Fisher SA. Hypoxia-inducible transcription factor-1alpha triggers an autocrine survival pathway during embryonic cardiac outflow tract remodeling. Circ Res 2008; 102:1331-9. [PMID: 18467628 PMCID: PMC2737478 DOI: 10.1161/circresaha.107.167858] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The cardiac outflow tract (OFT) of birds and mammals undergoes complex remodeling in the transition to a dual circulation. We have previously suggested a role of myocardial hypoxia and hypoxia inducible factor (HIF)-1 in the apoptosis-dependent remodeling of the OFT. In the present study, we transduced recombinant adenovirus-mediated HIF-1alpha in embryonic chick OFT myocardium to test its role in OFT remodeling. HIF-1alpha reduced the prevalence of apoptosis in OFT cardiomyocytes at stages 25 and 30, as determined by lysosome accumulation and caspase-3 activity. Associated conotruncal defects included malrotation of the aorta and excessive infundibular myocardium. HIF-1 targets induced in these gain-of-function experiments included vascular endothelial growth factor (VEGF), inducible nitric oxide synthase, and stromal cell-derived factor-1. To test the role of VEGF in this context, an adenovirus expressing secreted Flk1 (VEGF receptor 2) that binds and blocks VEGF signaling was targeted to the OFT myocardium. This caused increased cell death in the OFT myocardium at stages 25 and 30. Associated conotruncal heart defects included malrotation of the aorta, defects in the subpulmonic infundibulum associated with a small right ventricle, and increased OFT mesenchyme with failure of semilunar valve formation. We conclude that hypoxia signaling through HIF-1 and VEGF provides an autocrine survival signal in the developing cardiac OFT and that perturbation in this pathway causes OFT defects that model congenital human conotruncal heart defects.
Collapse
Affiliation(s)
- Hongbin Liu
- Department of Medicine (Cardiology), Case Western Reserve University, Cleveland, OH 44106, USA
| | | |
Collapse
|
31
|
Bajolle F, Zaffran S, Meilhac SM, Dandonneau M, Chang T, Kelly RG, Buckingham ME. Myocardium at the base of the aorta and pulmonary trunk is prefigured in the outflow tract of the heart and in subdomains of the second heart field. Dev Biol 2008; 313:25-34. [DOI: 10.1016/j.ydbio.2007.09.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Revised: 09/12/2007] [Accepted: 09/14/2007] [Indexed: 10/22/2022]
|
32
|
Abstract
How multicellular organisms obtain and use oxygen and other substrates has evolved over hundreds of millions of years in parallel with the evolution of oxygen-delivery systems. A steady supply of oxygen is critical to the existence of organisms that depend on oxygen as a primary source of fuel (i.e., those that live by aerobic metabolism). Not surprisingly, a number of mechanisms have evolved to defend against oxygen deprivation. This review highlights evolutionary and developmental aspects of O2 delivery to allow understanding of adaptive responses to O2 deprivation (hypoxia). First, we consider how the drive for more efficient oxygen delivery from the heart to the periphery may have shaped the evolution of the cardiovascular system, with particular attention to the routing of oxygenated and deoxygenated blood in the cardiac outlet. Then we consider the role of O2 in the morphogenesis of the cardiovascular system of animals of increasing size and complexity. We conclude by suggesting areas for future research regarding the role of oxygen deprivation and oxidative stress in the normal development of the heart and vasculature or in the pathogenesis of congenital heart defects.
Collapse
Affiliation(s)
- Steven A Fisher
- Case Western Reserve School of Medicine, Cleveland, Ohio 44106-7290, USA.
| | | |
Collapse
|
33
|
Kern CB, Norris RA, Thompson RP, Argraves WS, Fairey SE, Reyes L, Hoffman S, Markwald RR, Mjaatvedt CH. Versican proteolysis mediates myocardial regression during outflow tract development. Dev Dyn 2007; 236:671-83. [PMID: 17226818 PMCID: PMC1828600 DOI: 10.1002/dvdy.21059] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
An important phase of cardiac outflow tract (OFT) formation is the remodeling of the distal region of the common outlet in which the myocardial sleeve is replaced by with smooth muscle. Here we demonstrate that expression of the proteoglycan versican is reduced before the loss of myocardium from the distal cardiac outlet concomitant with an increase in production of the N-terminal cleavage fragment of versican. To test whether versican proteolysis plays a role in OFT remodeling, we determined the effects of adenoviral-mediated expression of a versican isoform devoid of known matrix metalloproteinase cleavage sites (V3) and an N-terminal fragment of versican (G1). V3 expression promoted an increase in thickness of the proximal OFT myocardial layer independent of proliferation. In contrast, the G1 domain caused thinning and interruptions of the OFT myocardium. These in vivo findings were consistent with findings using cultured primary cardiomyocytes showing that the V3 promoted myocardial cell-cell association while the G1 domain caused a loss of myocardial cell-cell association. Taken together, we conclude that intact versican and G1-containing versican cleavage products have opposing effects on myocardial cells and that versican proteolysis may facilitate the loss of distal myocardium during OFT remodeling.
Collapse
Affiliation(s)
- Christine B Kern
- Department of Cell Biology and Anatomy, Medical University of South Carolina, Charleston, SC 29425-2204, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Chen YH, Ishii M, Sun J, Sucov HM, Maxson RE. Msx1 and Msx2 regulate survival of secondary heart field precursors and post-migratory proliferation of cardiac neural crest in the outflow tract. Dev Biol 2007; 308:421-37. [PMID: 17601530 DOI: 10.1016/j.ydbio.2007.05.037] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Revised: 05/21/2007] [Accepted: 05/29/2007] [Indexed: 11/29/2022]
Abstract
Msx1 and Msx2 are highly conserved, Nk-related homeodomain transcription factors that are essential for a variety of tissue-tissue interactions during vertebrate organogenesis. Here we show that combined deficiencies of Msx1 and Msx2 cause conotruncal anomalies associated with malalignment of the cardiac outflow tract (OFT). Msx1 and Msx2 play dual roles in outflow tract morphogenesis by both protecting secondary heart field (SHF) precursors against apoptosis and inhibiting excessive proliferation of cardiac neural crest, endothelial and myocardial cells in the conotruncal cushions. During incorporation of SHF precursors into the OFT myocardium, ectopic apoptosis in the Msx1-/-; Msx2-/- mutant SHF is associated with reduced expression of Hand1 and Hand2, which from work on Hand1 and Hand2 mutants may be functionally important in the inhibition of apoptosis in Msx1/2 mutants. Later during aorticopulmonary septation, excessive proliferation in the OFT cushion mesenchyme and myocardium of Msx1-/-; Msx2-/- mutants is associated with premature down-regulation of p27(KIP1), an inhibitor of cyclin-dependent kinases. Diminished accretion of SHF precursors to the elongating OFT myocardium and excessive accumulation of mesenchymal cells in the conotruncal cushions may work together to perturb the rotation of the truncus arteriosus, leading to OFT malalignment defects including double-outlet right ventricle, overriding aorta and pulmonary stenosis.
Collapse
Affiliation(s)
- Yi-Hui Chen
- Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center and Hospital, University of Southern California Keck School of Medicine, 1441 Eastlake Avenue, Los Angeles, CA 90033, USA
| | | | | | | | | |
Collapse
|
35
|
Jenkins KJ, Correa A, Feinstein JA, Botto L, Britt AE, Daniels SR, Elixson M, Warnes CA, Webb CL. Noninherited risk factors and congenital cardiovascular defects: current knowledge: a scientific statement from the American Heart Association Council on Cardiovascular Disease in the Young: endorsed by the American Academy of Pediatrics. Circulation 2007; 115:2995-3014. [PMID: 17519397 DOI: 10.1161/circulationaha.106.183216] [Citation(s) in RCA: 561] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prevention of congenital cardiovascular defects has been hampered by a lack of information about modifiable risk factors for abnormalities in cardiac development. Over the past decade, there have been major breakthroughs in the understanding of inherited causes of congenital heart disease, including the identification of specific genetic abnormalities for some types of malformations. Although relatively less information has been available on noninherited modifiable factors that may have an adverse effect on the fetal heart, there is a growing body of epidemiological literature on this topic. This statement summarizes the currently available literature on potential fetal exposures that might alter risk for cardiovascular defects. Information is summarized for periconceptional multivitamin or folic acid intake, which may reduce the risk of cardiac disease in the fetus, and for additional types of potential exposures that may increase the risk, including maternal illnesses, maternal therapeutic and nontherapeutic drug exposures, environmental exposures, and paternal exposures. Information is highlighted regarding definitive risk factors such as maternal rubella; phenylketonuria; pregestational diabetes; exposure to thalidomide, vitamin A cogeners, or retinoids; and indomethacin tocolysis. Caveats regarding interpretation of possible exposure-outcome relationships from case-control studies are given because this type of study has provided most of the available information. Guidelines for prospective parents that could reduce the likelihood that their child will have a major cardiac malformation are given. Issues related to pregnancy monitoring are discussed. Knowledge gaps and future sources of new information on risk factors are described.
Collapse
|
36
|
Rana MS, Horsten NCA, Tesink-Taekema S, Lamers WH, Moorman AFM, van den Hoff MJB. Trabeculated right ventricular free wall in the chicken heart forms by ventricularization of the myocardium initially forming the outflow tract. Circ Res 2007; 100:1000-7. [PMID: 17347476 DOI: 10.1161/01.res.0000262688.14288.b8] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent molecular lineage analyses in mouse have demonstrated that the right ventricle is recruited from anterior mesoderm in later stages of cardiac development. This is in contrast to current views of development in the chicken heart, which suggest that the initial heart tube contains a subset of right ventricular precursors. We investigated the fate of the outflow tract myocardium using immunofluorescent staining of the myocardium, and lineage tracer, as well as cell death experiments. These analyses showed that the outflow tract is initially myocardial in its entirety, increasing in length up to HH24. The outflow tract myocardium, subsequently, shortens as a result of ventricularization, contributing to the trabeculated free wall, as well as the infundibulum, of the right ventricle. During this shortening, the overall length of the outflow tract is maintained because of the formation of a nonmyocardial portion between the distal myocardial border and the pericardial reflections. Cell death and transdifferentiation were found to play a more limited contribution to the initial shortening than is generally appreciated, if they play any part at all. Cell death, nonetheless, plays an important role in the disappearance of the myocardial collar that continues to invest the aorta and pulmonary trunk around HH30, and in the separation of the intrapericardial arterial vessels. Taken together, we show, as opposed to some current beliefs, the development of the arterial pole is similar in mammals and birds.
Collapse
Affiliation(s)
- M Sameer Rana
- Heart Failure Research Center, Academic Medical Center, University of Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
37
|
Gong J, Qian L, Kong X, Yang R, Zhou L, Sheng Y, Sun W, Sun F, Huang Y, Cao K. Cardiomyocyte apoptosis in the right auricle of patients with ostium secundum atrial septal defect diseases. Life Sci 2007; 80:1143-51. [PMID: 17275858 DOI: 10.1016/j.lfs.2006.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2006] [Revised: 11/26/2006] [Accepted: 12/11/2006] [Indexed: 11/15/2022]
Abstract
Ostium secundum atrial septal defect (osASD) is one of the most commonly occurring cardiac malformations. Although some embryological pathways have been elucidated, the molecular etiologies of ASD are not fully understood. Previous microarray analysis in our laboratory identified differentially expressed genes between osASD and normal right auricular myocardium. Of the 1056 differentially expressed genes, 14 genes were related to apoptosis: eight pro-apoptotic genes were up-regulated and six anti-apoptotic genes were down-regulated in ASD patients. In the current study, we utilized semi-quantitative RT-PCR, electron microscopy, TUNEL and flow cytometry to further understand the role of apoptosis in the atrium of osASD patients. RT-PCR results confirmed differential expression data from previous microarray studies. Additionally, while apoptosis was detected in the right auricular myocardium of all osASD patients, it was absent in controls. These data suggested apoptosis may play an important role in the pathogenesis of osASD or possibly occurs as a consequence of volume overload and hemodynamic changes in right atrium of osASD patients.
Collapse
Affiliation(s)
- Jie Gong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Eralp I, Lie-Venema H, Bax NAM, Wijffels MCEF, Van Der Laarse A, Deruiter MC, Bogers AJJC, Van Den Akker NMS, Gourdie RG, Schalij MJ, Poelmann RE, Gittenberger-De Groot AC. Epicardium-derived cells are important for correct development of the Purkinje fibers in the avian heart. THE ANATOMICAL RECORD. PART A, DISCOVERIES IN MOLECULAR, CELLULAR, AND EVOLUTIONARY BIOLOGY 2006; 288:1272-80. [PMID: 17075847 PMCID: PMC2610390 DOI: 10.1002/ar.a.20398] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
During embryonic development, the proepicardial organ (PEO) grows out over the heart surface to form the epicardium. Following epithelial-mesenchymal transformation, epicardium-derived cells (EPDCs) migrate into the heart and contribute to the developing coronary arteries, to the valves, and to the myocardium. The peripheral Purkinje fiber network develops from differentiating cardiomyocytes in the ventricular myocardium. Intrigued by the close spatial relationship between the final destinations of migrating EPDCs and Purkinje fiber differentiation in the avian heart, that is, surrounding the coronary arteries and at subendocardial sites, we investigated whether inhibition of epicardial outgrowth would disturb cardiomyocyte differentiation into Purkinje fibers. To this end, epicardial development was inhibited mechanically with a membrane, or genetically, by suppressing epicardial epithelial-to-mesenchymal transformation with antisense retroviral vectors affecting Ets transcription factor levels (n=4, HH39-41). In both epicardial inhibition models, we evaluated Purkinje fiber development by EAP-300 immunohistochemistry and found that restraints on EPDC development resulted in morphologically aberrant differentiation of Purkinje fibers. Purkinje fiber hypoplasia was observed both periarterially and at subendocardial positions. Furthermore, the cells were morphologically abnormal and not aligned in orderly Purkinje fibers. We conclude that EPDCs are instrumental in Purkinje fiber differentiation, and we hypothesize that they cooperate directly with endothelial and endocardial cells in the development of the peripheral conduction system.
Collapse
Affiliation(s)
- Ismail Eralp
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Barbosky L, Lawrence DK, Karunamuni G, Wikenheiser JC, Doughman YQ, Visconti RP, Burch JBE, Watanabe M. Apoptosis in the developing mouse heart. Dev Dyn 2006; 235:2592-602. [PMID: 16881058 DOI: 10.1002/dvdy.20885] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Apoptosis occurs at high frequency in the myocardium of the developing avian cardiac outflow tract (OFT). Up- or down-regulating apoptosis results in defects resembling human conotruncal heart anomalies. This finding suggested that regulated levels of apoptosis are critical for normal morphogenesis of the four-chambered heart. Recent evidence supports an important role for hypoxia of the OFT myocardium in regulating cell death and vasculogenesis. The purpose of this study was to determine whether apoptosis in the outflow tract myocardium occurs in the mouse heart during developmental stages comparable to the avian heart and to determine whether differential hypoxia is also present at this site in the murine heart. Apoptosis was detected using a fluorescent vital dye, Lysotracker Red (LTR), in the OFT myocardium of the mouse starting at embryonic day (E) 12.5, peaking at E13.5-14.5, and declining thereafter to low or background levels by E18.5. In addition, high levels of apoptosis were detected in other cardiac regions, including the apices of the ventricles and along the interventricular sulcus. Apoptosis in the myocardium was detected by double-labeling with LTR and cardiomyocyte markers. Terminal deoxynucleotidyl transferase-mediated deoxyuridinetriphosphate nick end-labeling (TUNEL) and immunostaining for cleaved Caspase-3 were used to confirm the LTR results. At the peak of OFT apoptosis in the mouse, the OFT myocardium was relatively hypoxic, as indicated by specific and intense EF5 staining and HIF1alpha nuclear localization, and was surrounded by the developing vasculature as in the chicken embryo. These findings suggest that cardiomyocyte apoptosis is an evolutionarily conserved mechanism for normal morphogenesis of the outflow tract myocardium in avian and mammalian species.
Collapse
Affiliation(s)
- Laura Barbosky
- Department of Pediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106-6011, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang WD, Huang CJ, Lu YF, Hsin JP, Prabhakar VR, Cheng CF, Hwang SPL. Heart-targeted overexpression of Nip3a in zebrafish embryos causes abnormal heart development and cardiac dysfunction. Biochem Biophys Res Commun 2006; 347:979-87. [PMID: 16854375 DOI: 10.1016/j.bbrc.2006.06.174] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Accepted: 06/29/2006] [Indexed: 11/30/2022]
Abstract
We transiently expressed a proapoptotic protein, Nip3a, by a heart-specific BMP4 promoter in zebrafish embryos and generated two variants of embryos with abnormal heart phenotypes (A and B). Embryos with phenotype A heart defects showed hypoplastic or elongated ventricles, elongated or enlarged atriums with no normal cardiac looping resulting a significant longer SV-BA distance, and bradycardia at 48 h post-fertilization (hpf). Embryos with phenotype B heart defects showed an enlarged fluid-filled pericardium, severe hypoplasia, non-contracting ventricles, and elongated or enlarged slowly beating atriums with no normal looping. Histological sections further revealed the absence of a proper atrioventricular boundary and no endocardial cells lining this region in both 48- and 72-hpf Nip3a-overexpressing embryos, implicating defective endocardial cushion formation. These phenotypes are reminiscent of atrioventricular canal defects in humans. In addition, induced apoptotic myocardium cells were clustered in the presumptive atrioventricular boundary as well as in the adjacent ventricle and atrium of 48- and 72-hpf Nip3a-overexpressing embryos. Nip3a expression was readily detected in 80% epiboly BMP4-Nip3a-injected embryos, and defects in heart development were observed in both the linear heart tube and subsequent chamber formation stages. These results showed that myocyte apoptosis is a universal pathogenic factor for congenital heart failure using zebrafish as a model organism.
Collapse
Affiliation(s)
- Wen-Der Wang
- Vanderbilt University, LH529, 2215 Garland Avenue, Nashville, TN 37232-0275, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Grimes AC, Stadt HA, Shepherd IT, Kirby ML. Solving an enigma: Arterial pole development in the zebrafish heart. Dev Biol 2006; 290:265-76. [PMID: 16405941 DOI: 10.1016/j.ydbio.2005.11.042] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2005] [Accepted: 11/03/2005] [Indexed: 11/27/2022]
Abstract
It is a widely held belief that the arterial pole of the zebrafish heart is unusual among models of comparative cardiogenesis. This is based, in part, on the report that the bulbus arteriosus undergoes a striated-to-smooth muscle phenotypic transition during development. An implication of this is that the zebrafish, a model almost ubiquitously accepted in other fields of comparative biology, may be poorly suited to the study of conotruncal abnormalities in human disease. However, while the use of atrioventricular-specific molecular markers has allowed extensive characterization of the development of the atrium and ventricle, the lack of any bulbus-specific markers has meant that this region of the zebrafish heart is poorly characterized and quite possibly misunderstood. We have discovered that the fluorescent nitric oxide indicator 4,5-diaminofluorescein diacetate (DAF-2DA) specifically labels the bulbus arteriosus throughout development from approximately 48 h post-fertilization. Therefore, using DAF-2DA and an immunohistochemical approach, we attempted to further characterize the development of the bulbus. We have concluded that no such phenotypic transition occurs, that contrary to current thinking, aspects of zebrafish arterial pole development are evolutionarily conserved, and that the bulbus should not be considered a chamber, being more akin to the arterial trunk(s) of higher vertebrates.
Collapse
Affiliation(s)
- Adrian C Grimes
- Medical University of South Carolina, Molecular and Cellular Biology and Pathobiology, 173 Ashley Avenue, Charleston, SC 29425, USA.
| | | | | | | |
Collapse
|
42
|
Ward C, Stadt H, Hutson M, Kirby ML. Ablation of the secondary heart field leads to tetralogy of Fallot and pulmonary atresia. Dev Biol 2005; 284:72-83. [PMID: 15950213 DOI: 10.1016/j.ydbio.2005.05.003] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2005] [Revised: 04/22/2005] [Accepted: 05/04/2005] [Indexed: 11/23/2022]
Abstract
Recent studies in chick and mouse embryos have identified a previously unrecognized secondary heart field (SHF), located in the ventral midline splanchnic mesenchyme, which provides additional myocardial cells to the outflow tract as the heart tube lengthens during cardiac looping. In order to further delineate the contribution of this secondary myocardium to outflow development, we labeled the right SHF of Hamburger-Hamilton (HH) stage 14 chick embryos via microinjection of DiI/rhodamine and followed the fluorescently labeled cells over a 96-h time period. These experiments confirmed the movement of the SHF into the outflow and its spiraling migration distally, with the right side of the SHF contributing to the left side of the outflow. In contrast, when the right SHF was labeled at HH18, the fluorescence was limited to the caudal wall of the lengthening aortic sac. We then injected a combination of DiI and neutral red dye, and ablated the SHF in HH14 or 18 chick embryos. Embryos were allowed to develop until day 9, and harvested for assessment of outflow alignment. Of the embryos ablated at HH14, 76% demonstrated cardiac defects including overriding aorta and pulmonary atresia, while none of the sham-operated controls were affected. In addition, the more severely affected embryos demonstrated coronary artery anomalies. The embryos ablated at HH18 also manifested coronary artery anomalies but maintained normal outflow alignment. Therefore, the myocardium added to the outflow by the SHF at earlier stages is required for the elongation and appropriate alignment of the outflow tract. However, at later stages, the SHF contributes to the smooth muscle component of the outflow vessels above the pulmonary and aortic valves which is important for the development of the coronary artery stems. This work suggests a role for the SHF in a subset of congenital heart defects that have overriding aorta and coronary artery anomalies, such as tetralogy of Fallot and double outlet right ventricle.
Collapse
Affiliation(s)
- Cary Ward
- Department of Medicine (Cardiology), Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
43
|
Abstract
Cardiac progenitors of the splanchnic mesoderm (primary and secondary heart field), cardiac neural crest, and the proepicardium are the major embryonic contributors to chick heart development. Their contribution to cardiac development occurs with precise timing and regulation during such processes as primary heart tube fusion, cardiac looping and accretion, cardiac septation, and the development of the coronary vasculature. Heart development is even more complex if one follows the development of the cardiac innervation, cardiac pacemaking and conduction system, endocardial cushions, valves, and even the importance of apoptosis for proper cardiac formation. This review is meant to provide a reference guide (Table 1) on the developmental timing according to the staging of Hamburger and Hamilton (1951) (HH) of these important topics in heart development for those individuals new to a chick heart research laboratory. Even individuals outside of the heart field, who are working on a gene that is also expressed in the heart, will gain information on what to look for during chick heart development. This reference guide provides complete and easy reference to the stages involved in heart development, as well as a global perspective of how these cardiac developmental events overlap temporally and spatially, making it a good bench top companion to the many recently written in-depth cardiac reviews of the molecular aspects of cardiac development.
Collapse
Affiliation(s)
- Brad J Martinsen
- Department of Pediatrics, Division of Pediatric Cardiology, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA.
| |
Collapse
|
44
|
Verzi MP, McCulley DJ, De Val S, Dodou E, Black BL. The right ventricle, outflow tract, and ventricular septum comprise a restricted expression domain within the secondary/anterior heart field. Dev Biol 2005; 287:134-45. [PMID: 16188249 DOI: 10.1016/j.ydbio.2005.08.041] [Citation(s) in RCA: 405] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2005] [Revised: 08/13/2005] [Accepted: 08/29/2005] [Indexed: 11/25/2022]
Abstract
The vertebrate heart arises from the fusion of bilateral regions of anterior mesoderm to form a linear heart tube. Recent studies in mouse and chick have demonstrated that a second cardiac progenitor population, known as the anterior or secondary heart field, is progressively added to the heart at the time of cardiac looping. While it is clear that this second field contributes to the myocardium, its precise boundaries, other lineages derived from this population, and its contributions to the postnatal heart remain unclear. In this study, we used regulatory elements from the mouse mef2c gene to direct the expression of Cre recombinase exclusively in the anterior heart field and its derivatives in transgenic mice. By crossing these mice, termed mef2c-AHF-Cre, to Cre-dependent lacZ reporter mice, we generated a fate map of the embryonic, fetal, and postnatal heart. These studies show that the endothelial and myocardial components of the outflow tract, right ventricle, and ventricular septum are derivatives of mef2c-AHF-Cre expressing cells within the anterior heart field and its derivatives. These studies also show that the atria, epicardium, coronary vessels, and the majority of outflow tract smooth muscle are not derived from this anterior heart field population. Furthermore, a transgene marker specific for the anterior heart field is expressed in the common ventricular chamber in mef2c mutant mice, suggesting that the cardiac looping defect in these mice is not due to a failure in anterior heart field addition to the heart. Finally, the Cre transgenic mice described here will be a crucial tool for conditional gene inactivation exclusively in the anterior heart field and its derivatives.
Collapse
Affiliation(s)
- Michael P Verzi
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-2240, USA
| | | | | | | | | |
Collapse
|
45
|
Abstract
The chick embryo is a versatile model system, in which classical embryology can be combined with modern molecular approaches. In the last two decades, several efficient methods have been developed to introduce exogenous genes into the chick embryo. These techniques allow alteration of gene expression levels in a spatially and temporally restricted manner, thereby circumventing embryonic lethality and/or eliminating secondary effects in other tissues. Here, we present the current status of avian somatic transgenic techniques, focusing on electroporation and retrovirus-mediated gene transfer. Electroporation allows quick and efficient gain-of-function studies based on transient misexpression of genes. Retroviral vectors, which are capable of integrating exogenous genes into the host chromosome, permit analysis of long-term effects of gene misexpression. The variety of methods available for somatic transgenesis, along with the recent completion of the chicken genome, are transforming the chick embryo into one of the most attractive model systems to examine function of genes that are important for embryonic development.
Collapse
Affiliation(s)
- Yasuo Ishii
- Department of Cell and Developmental Biology, Cornell University Medical College, New York, New York 10021, USA
| | | |
Collapse
|
46
|
Sugishita Y, Leifer DW, Agani F, Watanabe M, Fisher SA. Hypoxia-responsive signaling regulates the apoptosis-dependent remodeling of the embryonic avian cardiac outflow tract. Dev Biol 2004; 273:285-96. [PMID: 15328013 DOI: 10.1016/j.ydbio.2004.05.036] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2004] [Revised: 05/19/2004] [Accepted: 05/25/2004] [Indexed: 12/01/2022]
Abstract
We proposed a model in which myocardial hypoxia triggers the apoptosis-dependent remodeling of the avian outflow tract (OFT) in the transition of the embryo to a dual circulation. In this study, we examined hypoxia-dependent signaling in cardiomyocyte apoptosis and outflow tract remodeling. The hypoxia-inducible transcription factor HIF-1alpha was specifically present in the nuclei of OFT cardiomyocytes from stages 25-32, the period of hypoxia-dependent OFT remodeling. HIF-1alpha expression was sensitive to changes in ambient oxygen concentrations, while its dimerization partner HIF-1beta was constitutively expressed. There was not a simple relationship between HIF-1alpha expression and apoptosis. Apoptotic cardiomyocytes were detected in HIF-1alpha-positive and -negative regions, and a hypoxic stimulus sufficient to induce nuclear accumulation of HIF-1alpha did not induce cardiomyocyte apoptosis. The hypoxia-dependent expression of the vascular endothelial growth factor receptor (VEGFR2) in the distal OFT myocardium may be protective as cardiomyocyte apoptosis in the early stages (25-30) of OFT remodeling was absent from this region. Furthermore, recombinant adenoviral-mediated expression of dominant negative Akt, an inhibitor of tyrosine kinase receptor signaling, augmented cardiomyocyte apoptosis in the OFT and constitutively active Akt suppressed it. Adenovirus-mediated forced expression of VEGF165 induced conotruncal malformation such as double outlet right ventricle (DORV) and ventricular septal defect (VSD), similar to defects observed when apoptosis-dependent remodeling of the OFT was specifically targeted. We conclude that normal developmental remodeling of the embryonic avian cardiac OFT involves hypoxia/HIF-1-dependent signaling and cardiomyocyte apoptosis. Autocrine signaling through VEGF/VEGFR2 and Akt provides survival signals for the hypoxic OFT cardiomyocytes, and regulated VEGF signaling is required for the normal development of the OFT.
Collapse
Affiliation(s)
- Yasuyuki Sugishita
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | |
Collapse
|
47
|
Meilhac SM, Esner M, Kerszberg M, Moss JE, Buckingham ME. Oriented clonal cell growth in the developing mouse myocardium underlies cardiac morphogenesis. ACTA ACUST UNITED AC 2004; 164:97-109. [PMID: 14709543 PMCID: PMC2171975 DOI: 10.1083/jcb.200309160] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
During heart morphogenesis, cardiac chambers arise by differential expansion of regions of the primitive cardiac tube. This process is under the control of specific transcription factors such as Tbx5 and dHAND. To gain insight into the cellular mechanisms that underlie cardiogenesis, we have used a retrospective clonal approach based on the spontaneous recombination of an nlaacZ reporter gene targeted to the murine alpha-cardiac actin locus. We show that clonal growth of myocardial cells is oriented. At embryonic day (E) 10.5, the shape of clones is characteristic of a given cardiac region and reflects its morphology. This is already detectable in the primitive cardiac tube at E8.5, and is maintained after septation at E14.5 with additional modulations. The clonal analysis reveals new subdivisions of the myocardium, including an interventricular boundary region. Our results show that the myocardium, from the time of its formation, is a polarized and regionalized tissue and point to the role of oriented clonal cell growth in cardiac chamber morphogenesis.
Collapse
Affiliation(s)
- Sigolène M Meilhac
- Département de Biologie du Développement, Institut Pasteur, 28 Rue de Dr. Roux, 75724 Paris Cedex 15, France
| | | | | | | | | |
Collapse
|
48
|
Sugishita Y, Watanabe M, Fisher SA. The Development of the Embryonic Outflow Tract Provides Novel Insights into Cardiac Differentiation and Remodeling. Trends Cardiovasc Med 2004; 14:235-41. [PMID: 15451515 DOI: 10.1016/j.tcm.2004.06.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The embryonic cardiac outflow tract (OFT) connects the developing ventricles with the aortic sac. In birds and mammals, OFT cardiomyocytes are generated from a "secondary (anterior)," heart-forming field well after the formation of the primitive heart tube. The OFT cardiomyocytes have unique properties and developmental fates as compared with the myocytes of the atrial and ventricular chambers. Many of the OFT cardiomyocytes of the avian embryo are eliminated by programmed cell death (PCD) during OFT remodeling in the transition from a single- to a dual-series circulation. Targeted PCD gain and loss-of-function studies indicate that PCD drives the shortening and rotation of the OFT required for the aorta and pulmonary artery to connect with the left and right ventricles, respectively. Defects in this process model aspects of the relatively common and often life-threatening congenital human conotruncal heart defects. Using indicators of tissue hypoxia, we suggest that OFT myocardial hypoxia may be the trigger for the PCD-dependent remodeling of the OFT. This review discusses these aspects of the formation and remodeling of the embryonic OFT in the context of the broader questions of cardiac muscle biology.
Collapse
Affiliation(s)
- Yasuyuki Sugishita
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | |
Collapse
|
49
|
Sugishita Y, Watanabe M, Fisher SA. Role of myocardial hypoxia in the remodeling of the embryonic avian cardiac outflow tract. Dev Biol 2004; 267:294-308. [PMID: 15013795 DOI: 10.1016/j.ydbio.2003.11.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2003] [Revised: 09/23/2003] [Accepted: 11/17/2003] [Indexed: 10/26/2022]
Abstract
The embryonic cardiac outflow myocardium originates from a secondary heart-forming field to connect the developing ventricles with the aortic sac. The outflow tract (OFT) subsequently undergoes complex remodeling in the transition of the embryo to a dual circulation. In avians, elimination of OFT cardiomyocytes by apoptosis (stages 25-32) precedes coronary vasculogenesis and is necessary for the shortening of the OFT and the posterior rotation of the aorta. We hypothesized that regional myocardial hypoxia triggers OFT remodeling. We used immunohistochemical detection of the nitroimidazole EF5, administered by intravascular infusion in ovo, as an indicator of relative tissue oxygen concentrations. EF5 binding was increased in the OFT myocardium relative to other myocardium during these stages (25-32) of OFT remodeling. The intensity of EF5 binding paralleled the prevalence of apoptosis in the OFT myocardium, which are first detected at stage 25, maximal at stage 30, and diminished by stage 32. Evidence of coincident hypoxia-dependent responses included the expression of the vascular endothelial growth factor (VEGF) receptor 2 by the OFT myocardium, the predominant expression of VEGF122 (diffusible) isoform in the OFT, and the recruitment of QH1-positive pro-endothelial cells to the OFT and vasculogenesis. Exposure of embryos to hyperoxia (95% O(2)/5% CO(2)) during this developmental window reduced the prevalence of cardiomyocyte apoptosis and attenuated the shortening and rotation of the OFT, resulting in double-outlet right ventricle morphology, similar to that observed when apoptosis is directly inhibited. These results suggest that regional myocardial hypoxia triggers cardiomyocyte apoptosis and remodeling of the OFT in the transition to a dual circulation, and that VEGF autocrine/paracrine signaling may regulate these processes.
Collapse
Affiliation(s)
- Yasuyuki Sugishita
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
50
|
Sallee D, Qiu Y, Liu J, Watanabe M, Fisher SA. Fas ligand gene transfer to the embryonic heart induces programmed cell death and outflow tract defects. Dev Biol 2004; 267:309-19. [PMID: 15013796 DOI: 10.1016/j.ydbio.2003.11.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2003] [Revised: 11/18/2003] [Accepted: 11/20/2003] [Indexed: 10/26/2022]
Abstract
The remodeling of the embryonic avian cardiac outflow tract (OFT) involves the removal of cardiomyocytes by programmed cell death (PCD). In contrast, the prevalence of PCD is low in the atrial or ventricular myocytes during this period of development. To determine if this selective PCD is due to the unique ability of the OFT cardiomyocytes to execute PCD, we transduced the embryonic chicken heart in ovo with recombinant adenovirus expressing a death (FasL) ligand. This resulted in programmed cell death in atrial, ventricular, and OFT cardiomyocytes as evidenced by chromosomal fragmentation, accumulation of lysosomes, and Caspase enzymatic activity. Consistent with the widespread induction of PCD, transcripts for the Fas receptor were detected in all chambers of the heart throughout development. The precocious and widespread activation of PCD in the OFT myocardium resulted in a marked dimunition of the subpulmonic myocardial infundibulum, and transposition of the aorta side-by-side with the pulmonary artery and connecting to the right ventricle. Defects in other cardiac structures are also described. We conclude that the regulated removal of OFT cardiomyocytes by PCD is required for the great vessels to make their proper connections with the ventricles in the transition to a dual circulation. The malalignment of the great vessels described in this animal model are similar to those described in congenital human conotruncal heart defects, suggesting that PCD-dependent remodeling of the OFT myocardium could be a target of genetic mutations or teratogens that cause human conotruncal heart defects.
Collapse
Affiliation(s)
- Denver Sallee
- Department of Pediatrics (Cardiology), Case Western Reserve School of Medicine, Cleveland, OH 44106, USA
| | | | | | | | | |
Collapse
|