1
|
Wolton M, Davey MG, Dietrich S. At early stages of heart development, the first and second heart fields are a continuum of lateral head mesoderm-derived, cardiogenic cells. Dev Biol 2025; 520:200-223. [PMID: 39848483 DOI: 10.1016/j.ydbio.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/25/2025]
Abstract
Pioneering work in the chicken established that the initial development of the heart consists of two stages: the quick assembly of a beating heart, followed by the recruitment of cells from adjacent tissues to deliver the mature in-and outflow tract. Cells to build the primitive heart were dubbed the first heart field (FHF) cells, cells to be recruited later the second heart field (SHF) cells. The current view is that these cells represent distinct, maybe even pre-determined lineages. However, it is still unclear where exactly FHF and SHF are located at different stages of development, and whether there is a sharp boundary or rather an overlap between the two. It is also unclear whether both FHF cells and SHF cells originate from the lateral head mesoderm (LHM), whether the paraxial head mesoderm (PHM) contributes to the SHF, and where the LHM-PHM boundary might be. To investigate this problem, we exploited the size, ease of access and exquisite anatomy of the chicken embryo and used traditional strategies as well as newly developed transgenic lines to trace the location of cardiogenic fields and boundaries from the time the first heart-markers are expressed to the time SHF cell recruitment ceases. Our work shows that both FHF and SHF stem from the LHM. We also found that FHF and SHF lack a distinct anatomical boundary. Rather, FHF and SHF are a continuum, and the recruitment of cells into the heart is a chance event depending on morphogenetic movements, the position of cells within the moving tissues, the separation of the somatic and splanchnic LHM, and the separation of the heart from the splanchnic subpharyngeal mesoderm during heart-looping. Reconciling our and previous studies we propose that first and second heart field precursors are specified but not determined, thus relying on morphogenetic processes and local environments to realise their cardiogenic potential.
Collapse
Affiliation(s)
- Matthew Wolton
- Institute of Life Sciences and Health (ILSH), School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DT, UK
| | - Megan G Davey
- Functional Genetics, The Roslin Institute, The Royal (Dick) School of Veterinary Studies, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Susanne Dietrich
- Institute of Life Sciences and Health (ILSH), School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DT, UK.
| |
Collapse
|
2
|
Phillips M, Nimmo M, Rugonyi S. Developmental and Evolutionary Heart Adaptations Through Structure-Function Relationships. J Cardiovasc Dev Dis 2025; 12:83. [PMID: 40137081 PMCID: PMC11942974 DOI: 10.3390/jcdd12030083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/27/2025] Open
Abstract
While the heart works as an efficient pump, it also has a high level of adaptivity by changing its structure to maintain function during healthy and diseased states. In this Review, we present examples of structure-function relationships across species and throughout embryonic development in mammals and birds. We also summarize current research on avian models aiming at understanding how biophysical and biological mechanisms closely interact during heart formation. We conclude by underscoring similarities between cardiac adaptations and structural changes over developmental and evolutionary time scales and how understanding the mechanisms behind these adaptations can help prevent or alleviate the effects of cardiac malformations and contribute to cardiac regeneration efforts.
Collapse
Affiliation(s)
| | | | - Sandra Rugonyi
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97239, USA; (M.P.); (M.N.)
| |
Collapse
|
3
|
Ren P, Jiang B, Hassab A, Li G, Li W, Assi R, Tellides G. Heterogeneous Cardiac-Derived and Neural Crest-Derived Aortic Smooth Muscle Cells Exhibit Similar Transcriptional Changes After TGFβ Signaling Disruption. Arterioscler Thromb Vasc Biol 2025; 45:260-276. [PMID: 39697172 PMCID: PMC12053597 DOI: 10.1161/atvbaha.124.321706] [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: 08/16/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Smooth muscle cells (SMCs) of cardiac and neural crest origin contribute to the developing proximal aorta and are linked to disease propensity in adults. METHODS We analyzed single-cell transcriptomes of aortic SMCs from adult mice to determine basal states and changes after disrupting TGFβ (transforming growth factor-β) signaling necessary for aortic homeostasis. RESULTS A minority of Myh11 lineage-marked SMCs differentially expressed genes suggestive of embryological origin. Additional analyses in Nkx2-5 and Wnt1 lineage-marked SMCs derived from cardiac and neural crest progenitors, respectively, showed both lineages contributed to a major common cluster and each lineage to a minor distinct cluster. Common cluster SMCs extended from root to arch, cardiac subset cluster SMCs from root to ascending, and neural crest subset cluster SMCs were restricted to the arch. The neural crest subset cluster had greater expression of a subgroup of TGFβ-dependent genes. Nonetheless, conditional deletion of TGFβ receptors resulted in similar transcriptional changes among all SMC clusters. Several disease-associated transcriptional responses were comparable among SMC clusters in a mouse model of Marfan syndrome aortopathy, while many embryological markers of murine aortic SMCs were not detected in adult human aortas. CONCLUSIONS There are multiple subtypes of cardiac-derived and neural crest-derived SMCs with shared or distinctive transcriptional profiles; neural crest subset cluster SMCs with increased expression of certain TGFβ-inducible genes are not spatially linked to the aortic root predisposed to aneurysms from aberrant TGFβ signaling; and loss of TGFβ responses after receptor deletion is uniform among SMC clusters.
Collapse
Affiliation(s)
- Pengwei Ren
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT, USA
| | - Bo Jiang
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT, USA
- Current affiliation: Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Abdulrahman Hassab
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT, USA
| | - Guangxin Li
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT, USA
- Current affiliation: Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Wei Li
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT, USA
- Current affiliation: Department of Vascular Surgery, Peking University People’s Hospital, Beijing, China
| | - Roland Assi
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, CT, USA
| | - George Tellides
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
4
|
Arriagada C, Lin E, Schonning M, Astrof S. Mesodermal fibronectin controls cell shape, polarity, and mechanotransduction in the second heart field during cardiac outflow tract development. Dev Cell 2025; 60:62-84.e7. [PMID: 39413783 PMCID: PMC11706711 DOI: 10.1016/j.devcel.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/06/2024] [Accepted: 09/13/2024] [Indexed: 10/18/2024]
Abstract
Failure in the elongation of the cardiac outflow tract (OFT) results in congenital heart disease due to the misalignment of the great arteries with the left and right ventricles. The OFT lengthens via the accretion of progenitors from the second heart field (SHF). SHF cells are exquisitely regionalized and organized into an epithelial-like layer, forming the dorsal pericardial wall (DPW). Tissue tension, cell polarity, and proliferation within the DPW are important for the addition of SHF-derived cells to the heart and OFT elongation. However, the genes controlling these processes are not completely characterized. Using conditional mutagenesis in the mouse, we show that fibronectin (FN1) synthesized by the mesoderm coordinates multiple cellular behaviors in the anterior DPW. FN1 is enriched in the anterior DPW and plays a role in OFT elongation by maintaining a balance between pro- and anti-adhesive cell-extracellular matrix (ECM) interactions and controlling DPW cell shape, polarity, cohesion, proliferation, and mechanotransduction.
Collapse
Affiliation(s)
- Cecilia Arriagada
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Evan Lin
- Princeton Day School, Princeton, NJ, USA
| | - Michael Schonning
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA.
| |
Collapse
|
5
|
Prill K, Windsor Reid P, Pilgrim D. Heart Morphogenesis Requires Smyd1b for Proper Incorporation of the Second Heart Field in Zebrafish. Genes (Basel) 2025; 16:52. [PMID: 39858599 PMCID: PMC11764850 DOI: 10.3390/genes16010052] [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: 12/02/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Abnormal development of the second heart field significantly contributes to congenital heart defects, often caused by disruptions in tightly regulated molecular pathways. Smyd1, a gene encoding a protein with SET and MYND domains, is essential for heart and skeletal muscle development. Mutations in SMYD1 result in severe cardiac malformations and misregulation of Hand2 expression in mammals. This study examines the role of Smyd1b in zebrafish cardiac morphogenesis to elucidate its function and the mechanisms underlying congenital heart defects. Methods: Smyd1b (still heart) mutant embryos were analyzed for cardiac defects, and changes in gene expression related to heart development using live imaging, in situ hybridization, quantitative PCR and immunofluorescent comparisons and analysis. Results: Smyd1b mutants displayed severe cardiac defects, including failure to loop, severe edema, and an expansion of cardiac jelly linked to increased has2 expression. Additionally, the expression of key cardiac transcription factors, such as gata4, gata5, and nkx2.5, was notably reduced, indicating disrupted transcriptional regulation. The migration of cardiac progenitors was impaired and the absence of Islet-1-positive cells in the mutant hearts suggests a failed contribution of SHF progenitor cells. Conclusions: These findings underscore the essential role of Smyd1b in regulating cardiac morphogenesis and the development of the second heart field. This study highlights the potential of Smyd1b as a key factor in understanding the genetic and molecular mechanisms underlying congenital heart defects and cardiac development.
Collapse
Affiliation(s)
- Kendal Prill
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada; (K.P.); (P.W.R.)
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 1Y2, Canada
| | - Pamela Windsor Reid
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada; (K.P.); (P.W.R.)
- Department of Biological Science, MacEwan University, Edmonton, AB T5J 4S2, Canada
| | - Dave Pilgrim
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada; (K.P.); (P.W.R.)
| |
Collapse
|
6
|
Piñeiro-Sabarís R, MacGrogan D, de la Pompa JL. Deficient GATA6-CXCR7 signaling leads to bicuspid aortic valve. Dis Model Mech 2024; 17:dmm050934. [PMID: 39253784 PMCID: PMC11413932 DOI: 10.1242/dmm.050934] [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: 06/04/2024] [Accepted: 07/31/2024] [Indexed: 09/11/2024] Open
Abstract
The cardiac outflow tract (OFT) transiently links the ventricles to the aortic sac and forms the arterial valves. Abnormalities in these valves, such as bicuspid aortic valve (BAV), are common congenital anomalies. GATA6-inactivating variants cause cardiac OFT defects and BAV, but their mechanisms are unclear. We generated Gata6STOP/+ mice using CRISPR-Cas9, which show highly penetrant BAV (70%) and membranous ventricular septal defects (43%). These mice exhibited decreased proliferation and increased ISL1-positive progenitor cells in the OFT, indicating abnormal cardiovascular differentiation. Gata6 deletion with the Mef2cCre driver line recapitulated Gata6STOP/+ phenotypes, indicating a cell-autonomous role for Gata6 in the second heart field. Gata6STOP/+ mice showed reduced OFT length and caliber, associated with deficient cardiac neural crest cell contribution, which may cause valvulo-septal defects. RNA-sequencing analysis showed depletion in pathways related to cell proliferation and migration, highlighting Cxcr7 (also known as Ackr3) as a candidate gene. Reduced mesenchymal cell migration and invasion were observed in Gata6STOP/+ OFT tissue. CXCR7 agonists reduced mesenchymal cell migration and increased invasion in wild-type but not in Gata6STOP/+ explants, indicating the GATA6-dependent role of CXCR7 in OFT development and its potential link to BAV.
Collapse
Affiliation(s)
- Rebeca Piñeiro-Sabarís
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Donal MacGrogan
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| |
Collapse
|
7
|
Yang D, Jian Z, Tang C, Chen Z, Zhou Z, Zheng L, Peng X. Zebrafish Congenital Heart Disease Models: Opportunities and Challenges. Int J Mol Sci 2024; 25:5943. [PMID: 38892128 PMCID: PMC11172925 DOI: 10.3390/ijms25115943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Congenital heart defects (CHDs) are common human birth defects. Genetic mutations potentially cause the exhibition of various pathological phenotypes associated with CHDs, occurring alone or as part of certain syndromes. Zebrafish, a model organism with a strong molecular conservation similar to humans, is commonly used in studies on cardiovascular diseases owing to its advantageous features, such as a similarity to human electrophysiology, transparent embryos and larvae for observation, and suitability for forward and reverse genetics technology, to create various economical and easily controlled zebrafish CHD models. In this review, we outline the pros and cons of zebrafish CHD models created by genetic mutations associated with single defects and syndromes and the underlying pathogenic mechanism of CHDs discovered in these models. The challenges of zebrafish CHD models generated through gene editing are also discussed, since the cardiac phenotypes resulting from a single-candidate pathological gene mutation in zebrafish might not mirror the corresponding human phenotypes. The comprehensive review of these zebrafish CHD models will facilitate the understanding of the pathogenic mechanisms of CHDs and offer new opportunities for their treatments and intervention strategies.
Collapse
|
8
|
Ramirez A, Vyzas CA, Zhao H, Eng K, Degenhardt K, Astrof S. Buffering Mechanism in Aortic Arch Artery Formation and Congenital Heart Disease. Circ Res 2024; 134:e112-e132. [PMID: 38618720 PMCID: PMC11081845 DOI: 10.1161/circresaha.123.322767] [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: 03/08/2023] [Accepted: 03/27/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND The resiliency of embryonic development to genetic and environmental perturbations has been long appreciated; however, little is known about the mechanisms underlying the robustness of developmental processes. Aberrations resulting in neonatal lethality are exemplified by congenital heart disease arising from defective morphogenesis of pharyngeal arch arteries (PAAs) and their derivatives. METHODS Mouse genetics, lineage tracing, confocal microscopy, and quantitative image analyses were used to investigate mechanisms of PAA formation and repair. RESULTS The second heart field (SHF) gives rise to the PAA endothelium. Here, we show that the number of SHF-derived endothelial cells (ECs) is regulated by VEGFR2 (vascular endothelial growth factor receptor 2) and Tbx1. Remarkably, when the SHF-derived EC number is decreased, PAA development can be rescued by the compensatory endothelium. Blocking such compensatory response leads to embryonic demise. To determine the source of compensating ECs and mechanisms regulating their recruitment, we investigated 3-dimensional EC connectivity, EC fate, and gene expression. Our studies demonstrate that the expression of VEGFR2 by the SHF is required for the differentiation of SHF-derived cells into PAA ECs. The deletion of 1 VEGFR2 allele (VEGFR2SHF-HET) reduces SHF contribution to the PAA endothelium, while the deletion of both alleles (VEGFR2SHF-KO) abolishes it. The decrease in SHF-derived ECs in VEGFR2SHF-HET and VEGFR2SHF-KO embryos is complemented by the recruitment of ECs from the nearby veins. Compensatory ECs contribute to PAA derivatives, giving rise to the endothelium of the aortic arch and the ductus in VEGFR2SHF-KO mutants. Blocking the compensatory response in VEGFR2SHF-KO mutants results in embryonic lethality shortly after mid-gestation. The compensatory ECs are absent in Tbx1+/- embryos, a model for 22q11 deletion syndrome, leading to unpredictable arch artery morphogenesis and congenital heart disease. Tbx1 regulates the recruitment of the compensatory endothelium in an SHF-non-cell-autonomous manner. CONCLUSIONS Our studies uncover a novel buffering mechanism underlying the resiliency of PAA development and remodeling.
Collapse
Affiliation(s)
- AnnJosette Ramirez
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Christina A. Vyzas
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Huaning Zhao
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Kevin Eng
- Department of Statistics, Rutgers University, School of Arts and Sciences, Piscataway, NJ 08854
| | - Karl Degenhardt
- Children's Hospital of Pennsylvania, University of Pennsylvania, Philadelphia, PA 19107
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| |
Collapse
|
9
|
Ren P, Jiang B, Hassab A, Li G, Li W, Assi R, Tellides G. Heterogeneous Cardiac- and Neural Crest-Derived Aortic Smooth Muscle Cells have Similar Transcriptional Changes after TGFβ Signaling Disruption. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.28.591539. [PMID: 38746256 PMCID: PMC11092432 DOI: 10.1101/2024.04.28.591539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Smooth muscle cells (SMCs) of cardiac and neural crest origin contribute to the developing proximal aorta and are linked to disease propensity in adults. We analyzed single-cell transcriptomes of SMCs from mature thoracic aortas in mice to determine basal states and changes after disrupting transforming growth factor-β (TGFβ) signaling necessary for aortic homeostasis. A minority of Myh11 lineage-marked SMCs differentially expressed genes suggestive of embryological origin. Additional analyses in Nkx2-5 and Wnt1 lineage-marked SMCs derived from cardiac and neural crest progenitors, respectively, showed both lineages contributed to a major common cluster and each lineage to a minor distinct cluster. Common cluster SMCs extended from root to arch, cardiac subset cluster SMCs from root to mid-ascending, while neural crest subset cluster SMCs were restricted to the arch. The neural crest subset cluster had greater expression of a subgroup of TGFβ-dependent genes suggesting specific responsiveness or skewed extracellular matrix synthesis. Nonetheless, deletion of TGFβ receptors in SMCs resulted in similar transcriptional changes among all clusters, primarily decreased extracellular matrix molecules and modulators of TGFβ signaling. Many embryological markers of murine aortic SMCs were not confirmed in adult human aortas. We conclude: (i) there are multiple subtypes of cardiac- and neural crest-derived SMCs with shared or distinctive transcriptional profiles, (ii) neural crest subset SMCs with increased expression of certain TGFβ-inducible genes are not spatially linked to the aortic root predisposed to aneurysms from aberrant TGFβ signaling, and (iii) loss of TGFβ responses after receptor deletion is uniform among SMCs of different embryological origins.
Collapse
|
10
|
Guijarro C, Kelly RG. On the involvement of the second heart field in congenital heart defects. C R Biol 2024; 347:9-18. [PMID: 38488639 DOI: 10.5802/crbiol.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 03/19/2024]
Abstract
Congenital heart defects (CHD) affect 1 in 100 live births and result from defects in cardiac development. Growth of the early heart tube occurs by the progressive addition of second heart field (SHF) progenitor cells to the cardiac poles. The SHF gives rise to ventricular septal, right ventricular and outflow tract myocardium at the arterial pole, and atrial, including atrial septal myocardium, at the venous pole. SHF deployment creates the template for subsequent cardiac septation and has been implicated in cardiac looping and in orchestrating outflow tract development with neural crest cells. Genetic or environmental perturbation of SHF deployment thus underlies a spectrum of common forms of CHD affecting conotruncal and septal morphogenesis. Here we review the major properties of SHF cells as well as recent insights into the developmental programs that drive normal cardiac progenitor cell addition and the origins of CHD.
Collapse
|
11
|
Lin A, Ramaswamy Y, Misra A. Developmental heterogeneity of vascular cells: Insights into cellular plasticity in atherosclerosis? Semin Cell Dev Biol 2024; 155:3-15. [PMID: 37316416 DOI: 10.1016/j.semcdb.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023]
Abstract
Smooth muscle cells, endothelial cells and macrophages display remarkable heterogeneity within the healthy vasculature and under pathological conditions. During development, these cells arise from numerous embryological origins, which confound with different microenvironments to generate postnatal vascular cell diversity. In the atherosclerotic plaque milieu, all these cell types exhibit astonishing plasticity, generating a variety of plaque burdening or plaque stabilizing phenotypes. And yet how developmental origin influences intraplaque cell plasticity remains largely unexplored despite evidence suggesting this may be the case. Uncovering the diversity and plasticity of vascular cells is being revolutionized by unbiased single cell whole transcriptome analysis techniques that will likely continue to pave the way for therapeutic research. Cellular plasticity is only just emerging as a target for future therapeutics, and uncovering how intraplaque plasticity differs across vascular beds may provide key insights into why different plaques behave differently and may confer different risks of subsequent cardiovascular events.
Collapse
Affiliation(s)
- Alexander Lin
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, NSW, Australia; School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Yogambha Ramaswamy
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Ashish Misra
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, NSW, Australia; Heart Research Institute, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
12
|
Ramirez A, Vyzas CA, Zhao H, Eng K, Degenhardt K, Astrof S. Identification of novel buffering mechanisms in aortic arch artery development and congenital heart disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.02.530833. [PMID: 38370627 PMCID: PMC10871175 DOI: 10.1101/2023.03.02.530833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Rationale The resiliency of embryonic development to genetic and environmental perturbations has been long appreciated; however, little is known about the mechanisms underlying the robustness of developmental processes. Aberrations resulting in neonatal lethality are exemplified by congenital heart disease (CHD) arising from defective morphogenesis of pharyngeal arch arteries (PAA) and their derivatives. Objective To uncover mechanisms underlying the robustness of PAA morphogenesis. Methods and Results The second heart field (SHF) gives rise to the PAA endothelium. Here, we show that the number of SHF-derived ECs is regulated by VEGFR2 and Tbx1 . Remarkably, when SHF-derived EC number is decreased, PAA development can be rescued by the compensatory endothelium. Blocking such compensatory response leads to embryonic demise. To determine the source of compensating ECs and mechanisms regulating their recruitment, we investigated three-dimensional EC connectivity, EC fate, and gene expression. Our studies demonstrate that the expression of VEGFR2 by the SHF is required for the differentiation of SHF-derived cells into PAA ECs. The deletion of one VEGFR2 allele (VEGFR2 SHF-HET ) reduces SHF contribution to the PAA endothelium, while the deletion of both alleles (VEGFR2 SHF-KO ) abolishes it. The decrease in SHF-derived ECs in VEGFR2 SHF-HET and VEGFR2 SHF-KO embryos is complemented by the recruitment of ECs from the nearby veins. Compensatory ECs contribute to PAA derivatives, giving rise to the endothelium of the aortic arch and the ductus in VEGFR2 SHF-KO mutants. Blocking the compensatory response in VEGFR2 SHF-KO mutants results in embryonic lethality shortly after mid-gestation. The compensatory ECs are absent in Tbx1 +/- embryos, a model for 22q11 deletion syndrome, leading to unpredictable arch artery morphogenesis and CHD. Tbx1 regulates the recruitment of the compensatory endothelium in an SHF-non-cell-autonomous manner. Conclusions Our studies uncover a novel buffering mechanism underlying the resiliency of PAA development and remodeling. Nonstandard Abbreviations and Acronyms in Alphabetical Order CHD - congenital heart disease; ECs - endothelial cells; IAA-B - interrupted aortic arch type B; PAA - pharyngeal arch arteries; RERSA - retro-esophageal right subclavian artery; SHF - second heart field; VEGFR2 - Vascular endothelial growth factor receptor 2.
Collapse
|
13
|
Kelly RG. Molecular Pathways and Animal Models of Tetralogy of Fallot and Double Outlet Right Ventricle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:645-659. [PMID: 38884739 DOI: 10.1007/978-3-031-44087-8_37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Tetralogy of Fallot and double-outlet right ventricle are outflow tract (OFT) alignment defects situated on a continuous disease spectrum. A myriad of upstream causes can impact on ventriculoarterial alignment that can be summarized as defects in either i) OFT elongation during looping morphogenesis or ii) OFT remodeling during cardiac septation. Embryological processes underlying these two developmental steps include deployment of second heart field cardiac progenitor cells, establishment and transmission of embryonic left/right information driving OFT rotation and OFT cushion and valve morphogenesis. The formation and remodeling of pulmonary trunk infundibular myocardium is a critical component of both steps. Defects in myocardial, endocardial, or neural crest cell lineages can result in alignment defects, reflecting the complex intercellular signaling events that coordinate arterial pole development. Importantly, however, OFT alignment is mechanistically distinct from neural crest-driven OFT septation, although neural crest cells impact indirectly on alignment through their role in modulating signaling during SHF development. As yet poorly understood nongenetic causes of alignment defects that impact the above processes include hemodynamic changes, maternal exposure to environmental teratogens, and stochastic events. The heterogeneity of causes converging on alignment defects characterizes the OFT as a hotspot of congenital heart defects.
Collapse
Affiliation(s)
- Robert G Kelly
- Aix Marseille Université, Institut de Biologie du Dévelopment de Marseille, Marseille, France.
| |
Collapse
|
14
|
Houyel L. Ventricular Septal Defects: Molecular Pathways and Animal Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:535-549. [PMID: 38884730 DOI: 10.1007/978-3-031-44087-8_28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Ventricular septation is a complex process which involves the major genes of cardiac development, acting on myocardial cells from first and second heart fields, and on mesenchymal cells from endocardial cushions. These genes, coding for transcription factors, interact with each other, and their differential expression conditions the severity of the phenotype. In this chapter, we will describe the formation of the ventricular septum in the normal heart, as well as the molecular mechanisms leading to the four main anatomic types of ventricular septal defects: outlet, inlet, muscular, and central perimembranous, resulting from failure of development of the different parts of the ventricular septum. Experiments on animal models, particularly transgenic mouse lines, have helped us to decipher the molecular determinants of ventricular septation. However, a precise description of the anatomic phenotypes found in these models is mandatory to achieve a better comprehension of the complex mechanisms responsible for the various types of VSDs.
Collapse
Affiliation(s)
- Lucile Houyel
- Pediatric and Congenital Cardiology Unit, Necker-Enfants Malades Hospital - M3C, University of Paris, Paris, France.
| |
Collapse
|
15
|
Xing J, Wang H, Xie Y, Fan T, Cui C, Li Y, Wang S, Gu W, Wang C, Tang H, Liu L. Novel rare genetic variants of familial and sporadic pulmonary atresia identified by whole-exome sequencing. Open Life Sci 2023; 18:20220593. [PMID: 37215497 PMCID: PMC10199322 DOI: 10.1515/biol-2022-0593] [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: 11/09/2022] [Revised: 02/14/2023] [Accepted: 03/12/2023] [Indexed: 05/24/2023] Open
Abstract
Pulmonary atresia (PA) is a severe cyanotic congenital heart disease. Although some genetic mutations have been described to be associated with PA, the knowledge of pathogenesis is insufficient. The aim of this research was to use whole-exome sequencing (WES) to determine novel rare genetic variants in PA patients. We performed WES in 33 patients (27 patient-parent trios and 6 single probands) and 300 healthy control individuals. By applying an enhanced analytical framework to incorporate de novo and case-control rare variation, we identified 176 risk genes (100 de novo variants and 87 rare variants). Protein‒protein interaction (PPI) analysis and Genotype-Tissue Expression analysis revealed that 35 putative candidate genes had PPIs with known PA genes with high expression in the human heart. Expression quantitative trait loci analysis revealed that 27 genes that were identified as novel PA genes that could be affected by the surrounding single nucleotide polymorphism were screened. Furthermore, we screened rare damaging variants with a threshold of minor allele frequency at 0.5% in the ExAC_EAS and GnomAD_exome_EAS databases, and the deleteriousness was predicted by bioinformatics tools. For the first time, 18 rare variants in 11 new candidate genes have been identified that may play a role in the pathogenesis of PA. Our research provides new insights into the pathogenesis of PA and helps to identify the critical genes for PA.
Collapse
Affiliation(s)
- Junyue Xing
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou, Henan, 451464, China
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Heart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou, Henan, 451464, China
| | - Hongdan Wang
- Medical Genetics Institute of Henan Province, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, China
- National Health Commission Key Laboratory of Birth Defects Prevention, Henan Key Laboratory of Population Defects Prevention, Zhengzhou 450002, China
| | - Yuanyuan Xie
- Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, China
| | - Taibing Fan
- Department of Children’s Heart Center, Henan Provincial People’s Hospital, Department of Children’s Heart Center of Central China Fuwai Hospital, Henan Key Medical Laboratory of Tertiary Prevention and Treatment for Congenital Heart Disease, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Cunying Cui
- Department of Ultrasound, Fuwai Central China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 451464, China
| | - Yanan Li
- Department of Ultrasound, Fuwai Central China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 451464, China
| | - Shuai Wang
- Department of Translational Medicine Center, Chigene (Beijing) Translational Medical Research Center Co., Beijing, 100176, China
| | - Weiyue Gu
- Department of Translational Medicine Center, Chigene (Beijing) Translational Medical Research Center Co., Beijing, 100176, China
| | - Chengzeng Wang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Hao Tang
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou, Henan, 451464, China
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Heart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou, Henan, 451464, China
| | - Lin Liu
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| |
Collapse
|
16
|
Baral K, D'amato G, Kuschel B, Bogan F, Jones BW, Large CL, Whatley JD, Red-Horse K, Sharma B. APJ+ cells in the SHF contribute to the cells of aorta and pulmonary trunk through APJ signaling. Dev Biol 2023; 498:77-86. [PMID: 37037405 DOI: 10.1016/j.ydbio.2023.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 03/26/2023] [Accepted: 04/07/2023] [Indexed: 04/12/2023]
Abstract
Outflow tract develops from cardiac progenitor cells in the second heart field (SHF) domain. APJ, a G-Protein Coupled Receptor, is expressed by cardiac progenitor cells in the SHF. By lineage tracing APJ + SHF cells, we show that these cardiac progenitor cell contribute to the cells of outflow tract (OFT), which eventually give rise to aorta and pulmonary trunk/artery upon its morphogenesis. Furthermore, we show that early APJ + cells give rise to both aorta and pulmonary cells but late APJ + cells predominantly give rise to pulmonary cells. APJ is expressed by the outflow tract progenitors but its role in the SHF is unclear. We performed knockout studies to determine the role of APJ in SHF cell proliferation and survival. Our data suggested that APJ knockout in the SHF reduced the proliferation of SHF progenitors, while there was no significant impact on survival of the SHF progenitors. In addition, we show that ectopic overexpression of WNT in these cells disrupted aorta and pulmonary morphogenesis from outflow tract. Overall, our study have identified APJ + progenitor population within the SHF that give rise to aorta and pulmonary trunk/artery cells. Furthermore, we show that APJ signaling stimulate proliferation of these cells in the SHF.
Collapse
Affiliation(s)
- Kamal Baral
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Gaetano D'amato
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Bryce Kuschel
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Frank Bogan
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Brendan W Jones
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Colton L Large
- Department of Biology, Ball State University, Muncie, IN, USA
| | | | | | - Bikram Sharma
- Department of Biology, Ball State University, Muncie, IN, USA.
| |
Collapse
|
17
|
Lesieur E, Zaffran S, Chaoui R, Quarello E. Prenatal screening and diagnosis of pulmonary artery anomalies. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2023; 61:445-457. [PMID: 36178851 DOI: 10.1002/uog.26078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/08/2022] [Accepted: 09/11/2022] [Indexed: 06/16/2023]
Abstract
Congenital pulmonary artery anomalies are rare. Their antenatal diagnosis requires good knowledge of fetal cardiac anatomy because their clinical presentation varies depending on the type and severity of the underlying lesion. Screening of these vascular anomalies can be straightforward in some cases because of significant associated consequences that are detected easily on ultrasound, while other anomalies have considerably less obvious features. There may be an associated genetic syndrome. The aim of this review was to define anomalies of the main pulmonary artery and its branches and to propose, through the identification of suspicious findings during routine antenatal heart examination, an optimal screening method for the pulmonary artery pathway. We propose that pulmonary artery anomalies can be classified antenatally into four types of disorder. Herein we describe 14 cases subgrouped accordingly as: anomalies of the pulmonary valvular region, with stenosis or atresia of the valve (n = 4); conotruncal abnormalities (n = 4); anomalies associated with abnormal origin or course of the pulmonary artery (n = 4); and anomalies associated with abnormal growth of the pulmonary artery and its branches (n = 2). We highlight the need to differentiate the three-vessel view from the three-vessel-and-trachea view when assessing a fetus with a congenital pulmonary artery anomaly. © 2022 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- E Lesieur
- Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, Timone Hospital, Aix-Marseille University, Marseille, France
- UMR 7268 ADES, Aix-Marseille University, CNRS, EFS, Marseille, France
| | - S Zaffran
- Aix-Marseille University, INSERM, MMG, U1251, Marseille, France
| | - R Chaoui
- Center for Prenatal Diagnosis and Human Genetics, Berlin, Germany
| | - E Quarello
- Screening and Diagnosis Unit, Department of Obstetrics and Gynecology, Saint Joseph Hospital, Marseille, France
- IMAGE2 Center, Marseille, France
| |
Collapse
|
18
|
Should Prenatal Chromosomal Microarray Analysis Be Offered for Pulmonary Atresia? A Single-Center Retrospective Study in China. Genes (Basel) 2023; 14:genes14030722. [PMID: 36980994 PMCID: PMC10047995 DOI: 10.3390/genes14030722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
(1) Objective: To evaluate the application of chromosomal microarray analysis (CMA) in fetuses with pulmonary atresia (PA) and to explore the risk factors for predicting chromosomal imbalances and adverse perinatal outcomes. (2) Methods: This study investigated 428 cases of PA singleton pregnancies that were tested using CMA and quantitative fluorescent polymerase chain reaction (QF-PCR) as first-line genetic testing. The PA cases were divided into two groups: an isolated group and a non-isolated group. (3) Results: CMA revealed clinically relevant copy number variations (CNVs) in 9/139 (6.47%) PA fetuses, i.e., pathogenic copy number variations (pCNVs) in 8/139 (5.76%) fetuses and likely pathogenic CNVs in 1/139 (0.72%) fetuses. Stratified analysis showed that the incidence of clinically significant variants was higher in non-isolated PA fetuses than in isolated PA fetuses (12.50%, 6/48 vs. 3.30%, 3/91, p = 0.036). Regression analysis showed that a combination of other structural abnormalities at diagnosis of PA represented the principal risk factor for chromosomal imbalances (OR = 2.672). A combination of other structural abnormalities and a high maternal age increased the risk of adverse pregnancy outcomes in PA cases, including intrauterine fetal death (IUFD), termination of pregnancy (TOP), and preterm delivery. (4) Conclusions: The value of CMA for locating imbalanced genetic variations in fetuses with PA was highlighted by this study, particularly when combined with additional structural abnormalities.
Collapse
|
19
|
Omer SO, Alhabshan FM, Jijeh AMZ, Caimbon NC, Enriquez CC, Männer J, Yelbuz TM. Is Transposition of the Great Arteries Associated With Shortening of the Intrapericardial Portions of the Great Arterial Trunks? An Echocardiographic Analysis on Newborn Infants With Simple Transposition of the Great Arteries to Explore an Animal Model-Based Hypothesis on Human Beings. J Am Heart Assoc 2021; 10:e019334. [PMID: 34278802 PMCID: PMC8475693 DOI: 10.1161/jaha.120.019334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 05/17/2021] [Indexed: 11/30/2022]
Abstract
Background The pathogenesis of transposition of the great arteries (TGA) as a congenital heart defect of the outflow tract with discordant ventriculoarterial connections remains an enigma. TGA usually have parallel great arteries suggesting that deficient torsion of the embryonic arterial heart pole might cause discordant ventriculoarterial connections. It has been speculated that deficient elongation of the embryonic outflow tract might prevent its normal torsion resulting in TGA. The aim of our study was to clarify whether the intrapericardial portions of the great arteries in human patients with TGA might be indeed shorter than in normal hearts. Methods and Results Thirty-four newborns with simple TGA and 35 newborns with normal hearts were analyzed by using images of the outflow tract in their echocardiograms and the following defined lengths of the great arteries were measured: aortic length 1, (AoL-1) and aortic length 2 (AoL-2) = distance between left and right aortic valve level and origin of the brachiocephalic artery, respectively. Pulmonary trunk length 1 (PTL-1) and pulmonary trunk length 2 (PTL 2) = distance between left and right pulmonary valve level and origin of left and right pulmonary artery, respectively. All measurements of the AoL were significantly shorter in TGA compared to normal hearts (AoL-1: 1.6±0.2 versus 2.05±0.1; P<0.0001; AoL-2: 1.55±0.2 versus 2.13±0.1; P<0.0001). With regard to the pulmonary trunk (PT), PTL-1 and PTL-2 were found to be shorter and longer, respectively, in TGA compared with normal hearts, reflecting the differences in the spatial arrangement of the PT between the 2 groups as in TGA the PT is showing a mirror image of the normal anatomy. However, the overall length of the PT between the 2 groups did not differ. Conclusions Our data demonstrate that, compared with normal newborns, the ascending aorta is significantly shorter in newborns with TGA whereas the overall length of the PT does not differ between the 2 groups. This finding is in accord with the animal model-based hypothesis that TGA may result from a growth deficit at the arterial pole of the embryonic heart.
Collapse
Affiliation(s)
- Seham O. Omer
- Department of Cardiac SciencesKing Abdulaziz Cardiac CenterMinistry of National Guard Health AffairsRiyadhSaudi Arabia
- King Abdullah International Medical Research Center (KAIMRC)RiyadhSaudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences (KSAU‐HS)RiyadhSaudi Arabia
| | - Fahad M. Alhabshan
- Department of Cardiac SciencesKing Abdulaziz Cardiac CenterMinistry of National Guard Health AffairsRiyadhSaudi Arabia
- King Abdullah International Medical Research Center (KAIMRC)RiyadhSaudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences (KSAU‐HS)RiyadhSaudi Arabia
| | - Abdulraouf M. Z. Jijeh
- Department of Cardiac SciencesKing Abdulaziz Cardiac CenterMinistry of National Guard Health AffairsRiyadhSaudi Arabia
- King Abdullah International Medical Research Center (KAIMRC)RiyadhSaudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences (KSAU‐HS)RiyadhSaudi Arabia
| | - Natalia C. Caimbon
- Department of Cardiac SciencesKing Abdulaziz Cardiac CenterMinistry of National Guard Health AffairsRiyadhSaudi Arabia
- King Abdullah International Medical Research Center (KAIMRC)RiyadhSaudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences (KSAU‐HS)RiyadhSaudi Arabia
| | - Carmelita C. Enriquez
- Department of Cardiac SciencesKing Abdulaziz Cardiac CenterMinistry of National Guard Health AffairsRiyadhSaudi Arabia
- King Abdullah International Medical Research Center (KAIMRC)RiyadhSaudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences (KSAU‐HS)RiyadhSaudi Arabia
| | - Jörg Männer
- Institute for Anatomy and EmbryologyUMGGeorg‐August‐University of GöttingenGöttingenGermany
| | - Talat Mesud Yelbuz
- Department of Cardiac SciencesKing Abdulaziz Cardiac CenterMinistry of National Guard Health AffairsRiyadhSaudi Arabia
- King Abdullah International Medical Research Center (KAIMRC)RiyadhSaudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences (KSAU‐HS)RiyadhSaudi Arabia
| |
Collapse
|
20
|
Dyer LA, Rugonyi S. Fetal Blood Flow and Genetic Mutations in Conotruncal Congenital Heart Disease. J Cardiovasc Dev Dis 2021; 8:90. [PMID: 34436232 PMCID: PMC8397097 DOI: 10.3390/jcdd8080090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/19/2022] Open
Abstract
In congenital heart disease, the presence of structural defects affects blood flow in the heart and circulation. However, because the fetal circulation bypasses the lungs, fetuses with cyanotic heart defects can survive in utero but need prompt intervention to survive after birth. Tetralogy of Fallot and persistent truncus arteriosus are two of the most significant conotruncal heart defects. In both defects, blood access to the lungs is restricted or non-existent, and babies with these critical conditions need intervention right after birth. While there are known genetic mutations that lead to these critical heart defects, early perturbations in blood flow can independently lead to critical heart defects. In this paper, we start by comparing the fetal circulation with the neonatal and adult circulation, and reviewing how altered fetal blood flow can be used as a diagnostic tool to plan interventions. We then look at known factors that lead to tetralogy of Fallot and persistent truncus arteriosus: namely early perturbations in blood flow and mutations within VEGF-related pathways. The interplay between physical and genetic factors means that any one alteration can cause significant disruptions during development and underscore our need to better understand the effects of both blood flow and flow-responsive genes.
Collapse
Affiliation(s)
- Laura A. Dyer
- Department of Biology, University of Portland, Portland, OR 97203, USA;
| | - Sandra Rugonyi
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
21
|
Schussler O, Gharibeh L, Mootoosamy P, Murith N, Tien V, Rougemont AL, Sologashvili T, Suuronen E, Lecarpentier Y, Ruel M. Cardiac Neural Crest Cells: Their Rhombomeric Specification, Migration, and Association with Heart and Great Vessel Anomalies. Cell Mol Neurobiol 2021; 41:403-429. [PMID: 32405705 PMCID: PMC11448677 DOI: 10.1007/s10571-020-00863-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
Abstract
Outflow tract abnormalities are the most frequent congenital heart defects. These are due to the absence or dysfunction of the two main cell types, i.e., neural crest cells and secondary heart field cells that migrate in opposite directions at the same stage of development. These cells directly govern aortic arch patterning and development, ascending aorta dilatation, semi-valvular and coronary artery development, aortopulmonary septation abnormalities, persistence of the ductus arteriosus, trunk and proximal pulmonary arteries, sub-valvular conal ventricular septal/rotational defects, and non-compaction of the left ventricle. In some cases, depending on the functional defects of these cells, additional malformations are found in the expected spatial migratory area of the cells, namely in the pharyngeal arch derivatives and cervico-facial structures. Associated non-cardiovascular anomalies are often underestimated, since the multipotency and functional alteration of these cells can result in the modification of multiple neural, epidermal, and cervical structures at different levels. In most cases, patients do not display the full phenotype of abnormalities, but congenital cardiac defects involving the ventricular outflow tract, ascending aorta, aortic arch and supra-aortic trunks should be considered as markers for possible impaired function of these cells. Neural crest cells should not be considered as a unique cell population but on the basis of their cervical rhombomere origins R3-R5 or R6-R7-R8 and specific migration patterns: R3-R4 towards arch II, R5-R6 arch III and R7-R8 arch IV and VI. A better understanding of their development may lead to the discovery of unknown associated abnormalities, thereby enabling potential improvements to be made to the therapeutic approach.
Collapse
Affiliation(s)
- Olivier Schussler
- Department of Cardiovascular Surgery Adult and Pediatric, Geneva University Hospital, Geneva, Switzerland.
- Cardiovascular Research Laboratory, Faculty of Medicine of the University of Geneva, Rue Michel Servet 1, 1211, Geneva 4, Switzerland.
| | - Lara Gharibeh
- Molecular Genetics and Cardiac Regeneration Laboratory, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Parmeseeven Mootoosamy
- Department of Cardiovascular Surgery Adult and Pediatric, Geneva University Hospital, Geneva, Switzerland
| | - Nicolas Murith
- Department of Cardiovascular Surgery Adult and Pediatric, Geneva University Hospital, Geneva, Switzerland
| | - Vannary Tien
- Department of Pathology and Immunology, Faculty of Medicine of the University of Geneva, Geneva, Switzerland
| | | | - Tornike Sologashvili
- Department of Cardiovascular Surgery Adult and Pediatric, Geneva University Hospital, Geneva, Switzerland
| | - Erik Suuronen
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Cardiovascular Surgery, University of Ottawa Heart Institute and School of Epidemiology, Ottawa, ON, Canada
| | | | - Marc Ruel
- Department of Cardiovascular Surgery, University of Ottawa Heart Institute and School of Epidemiology, Ottawa, ON, Canada
| |
Collapse
|
22
|
Abstract
Cardiac neural crest (CNC) cells are pluripotent cells derived from the dorsal neural tube that migrate and contribute to the remodeling of pharyngeal arch arteries and septation of the cardiac outflow tract (OFT). Numerous molecular cascades regulate the induction, specification, delamination, and migration of the CNC. Extensive analyses of the CNC ranging from chick ablation models to molecular biology studies have explored the mechanisms of heart development and disease, particularly involving the OFT and aortic arch (AA) system. Recent studies focus more on reciprocal signaling between the CNC and cells originated from the second heart field (SHF), which are essential for the development of the OFT myocardium, providing new insights into the molecular mechanisms underlying congenital heart diseases (CHDs) and some human syndromes.
Collapse
Affiliation(s)
- Hiroyuki Yamagishi
- Division of Pediatric Cardiology, Department of Pediatrics, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
23
|
Al Nasef M, Alghamdi MH, Bello Valls ML, Zahrani AM, AlAkfash A, Ardah HI, Diranneya OM, Alhabshan F. Commissural Malalignment as a predictor of coronary artery abnormalities in patients with transposition of great arteries. JOURNAL OF CONGENITAL CARDIOLOGY 2020. [DOI: 10.1186/s40949-020-00039-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
In patients with transposition of the great arteries (TGA), commissural malalignment (CM) between semilunar valves may be associated with abnormal coronary (CA) pattern. We intend to assess the degree of CM with incidence of unusual CA anatomy.
Methods
We proposed a ratio to measure the distance of both ends of the anterior facing sinuses of the pulmonary valve from the facing commissure of the aortic valve. We labeled it as D1 and D2 distance. A ratio (C ratio) of the smaller distance (either D1 or D2 whichever is shorter) over the sum of both D1 and D2 was taken (D1 or D2 whichever is shorter / D1 + D2). We related this ratio with the incidence of the unusual CA anatomy in D-TGA patients.
Results
We had a total of 158 patients. We defined the point beyond which the C-Ratio becomes significantly associated with abnormal coronary artery pattern, this represents the median effective level (EL50). The EL50 of the C-Ratio was found to be equal to 31% (0.31). The prediction revealed that the CA pattern would most probably be usual when there is a minor commissural malalignment (C-Ratio less than the EL50) and most probably be unusual when there is a major malalignment (C-Ratio is greater than the EL50). The sensitivity was 71% and the specificity 88% (p-value < 0.0001).
Conclusions
The C-Ratio helps to categorize the degree of CM as minor (less than 0.31) or major (more than 0.31). A higher C-Ratio predicts a higher incidence of unusual CA pattern.
Collapse
|
24
|
BVES downregulation in non-syndromic tetralogy of fallot is associated with ventricular outflow tract stenosis. Sci Rep 2020; 10:14167. [PMID: 32843646 PMCID: PMC7447802 DOI: 10.1038/s41598-020-70806-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 08/04/2020] [Indexed: 11/14/2022] Open
Abstract
BVES is a transmembrane protein, our previous work demonstrated that single nucleotide mutations of BVES in tetralogy of fallot (TOF) patients cause a downregulation of BVES transcription. However, the relationship between BVES and the pathogenesis of TOF has not been determined. Here we reported our research results about the relationship between BVES and the right ventricular outflow tract (RVOT) stenosis. BVES expression was significantly downregulated in most TOF samples compared with controls. The expression of the second heart field (SHF) regulatory network genes, including NKX2.5, GATA4 and HAND2, was also decreased in the TOF samples. In zebrafish, bves knockdown resulted in looping defects and ventricular outflow tract (VOT) stenosis, which was mostly rescued by injecting bves mRNA. bves knockdown in zebrafish also decreased the expression of SHF genes, such as nkx2.5, gata4 and hand2, consistent with the TOF samples` results. The dual-fluorescence reporter system analysis showed that BVES positively regulated the transcriptional activity of GATA4, NKX2.5 and HAND2 promoters. In zebrafish, nkx2.5 mRNA partially rescued VOT stenosis caused by bves knockdown. These results indicate that BVES downregulation may be associated with RVOT stenosis of non-syndromic TOF, and bves is probably involved in the development of VOT in zebrafish.
Collapse
|
25
|
Mengmeng X, Yuejuan X, Sun C, Yanan L, Fen L, Kun S. Novel mutations of the SRF gene in Chinese sporadic conotruncal heart defect patients. BMC MEDICAL GENETICS 2020; 21:95. [PMID: 32380971 PMCID: PMC7203814 DOI: 10.1186/s12881-020-01032-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 04/22/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND Conotruncal heart defects (CTDs) are a group of congenital heart malformations that cause anomalies of cardiac outflow tracts. In the past few decades, many genes related to CTDs have been reported. Serum response factor (SRF) is a ubiquitous nuclear protein that acts as transcription factor, and SRF was found to be a critical factor in heart development and to be strongly expressed in the myocardium of the developing mouse and chicken hearts. The targeted inactivation of SRF during heart development leads to embryonic lethality and myocardial defects in mice. METHODS To illustrate the relationship between SRF and human heart defects, we screened SRF mutations in 527 CTD patients, a cross sectional study. DNA was extracted from peripheral leukocyte cells for target sequencing. The mutations of SRF were detected and validated by Sanger sequencing. The affection of the mutations on wild-type protein was analyzed by in silico softwares. Western blot and real time PCR were used to analyze the changes of the expression of the mutant mRNA and protein. In addition, we carried out dual luciferase reporter assay to explore the transcriptional activity of the mutant SRF. RESULTS Among the target sequencing results of 527 patients, two novel mutations (Mut1: c.821A > G p.G274D, the adenine(A) was mutated to guanine(G) at position 821 of the SRF gene coding sequences (CDS), lead to the Glycine(G) mutated to Asparticacid(D) at position 274 of the SRF protein amino acid sequences; Mut2: c.880G > T p.G294C, the guanine(G) was mutated to thymine (T) at position 880 of the SRF CDS, lead to the Glycine(G) mutated to Cysteine (C) at position 294 of the SRF protein amino acid sequences.) of SRF (NM_003131.4) were identified. Western blotting and real-time PCR showed that there were no obvious differences between the protein expression and mRNA transcription of mutants and wild-type SRF. A dual luciferase reporter assay showed that both SRF mutants (G274D and G294C) impaired SRF transcriptional activity at the SRF promoter and atrial natriuretic factor (ANF) promoter (p < 0.05), additionally, the mutants displayed reduced synergism with GATA4. CONCLUSION These results suggest that SRF-p.G274D and SRF-p.G294C may have potential pathogenic effects.
Collapse
Affiliation(s)
- Xu Mengmeng
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang road, Shanghai, 200092, China
| | - Xu Yuejuan
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang road, Shanghai, 200092, China.
| | - Chen Sun
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang road, Shanghai, 200092, China
| | - Lu Yanan
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang road, Shanghai, 200092, China
| | - Li Fen
- Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, No. 1678, Dongfang Road, Shanghai, 200127, China
| | - Sun Kun
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang road, Shanghai, 200092, China.
| |
Collapse
|
26
|
Follow Me! A Tale of Avian Heart Development with Comparisons to Mammal Heart Development. J Cardiovasc Dev Dis 2020; 7:jcdd7010008. [PMID: 32156044 PMCID: PMC7151090 DOI: 10.3390/jcdd7010008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/16/2020] [Accepted: 02/21/2020] [Indexed: 12/19/2022] Open
Abstract
Avian embryos have been used for centuries to study development due to the ease of access. Because the embryos are sheltered inside the eggshell, a small window in the shell is ideal for visualizing the embryos and performing different interventions. The window can then be covered, and the embryo returned to the incubator for the desired amount of time, and observed during further development. Up to about 4 days of chicken development (out of 21 days of incubation), when the egg is opened the embryo is on top of the yolk, and its heart is on top of its body. This allows easy imaging of heart formation and heart development using non-invasive techniques, including regular optical microscopy. After day 4, the embryo starts sinking into the yolk, but still imaging technologies, such as ultrasound, can tomographically image the embryo and its heart in vivo. Importantly, because like the human heart the avian heart develops into a four-chambered heart with valves, heart malformations and pathologies that human babies suffer can be replicated in avian embryos, allowing a unique developmental window into human congenital heart disease. Here, we review avian heart formation and provide comparisons to the mammalian heart.
Collapse
|
27
|
Yang Q, Wu F, Mi Y, Wang F, Cai K, Yang X, Zhang R, Liu L, Zhang Y, Wang Y, Wang X, Xu M, Gui Y, Li Q. Aberrant expression of miR-29b-3p influences heart development and cardiomyocyte proliferation by targeting NOTCH2. Cell Prolif 2020; 53:e12764. [PMID: 32077168 PMCID: PMC7106969 DOI: 10.1111/cpr.12764] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 12/24/2019] [Accepted: 12/30/2019] [Indexed: 12/13/2022] Open
Abstract
Objectives microRNA‐29 (miR‐29) family have shown different expression patterns in cardiovascular diseases. Our study aims to explore the effect and mechanism of miR‐29 family on cardiac development. Materials and methods A total of 13 patients with congenital heart disease (CHD) and 7 controls were included in our study. Tissues were obtained from the right ventricular outflow tract (RVOT) after surgical resection or autopsy. The next‐generation sequencing was applied to screen the microRNA expression profiles of CHD. Quantitative RT‐PCR and Western blot were employed to measure genes expression. Tg Cmlc2: GFP reporter zebrafish embryos were injected with microRNA (miRNA) to explore its role in cardiac development in vivo. Dual‐luciferase reporter assay was designed to validate the target gene of miRNAs. CCK‐8 and EdU incorporation assays were performed to evaluate cardiomyocyte proliferation. Results Our study showed miR‐29b‐3p expression was significantly increased in the RVOT of the CHD patients. Injection of miR‐29b‐3p into zebrafish embryos induced higher mortality and malformation rates, developmental delay, cardiac malformation and dysfunction. miR‐29b‐3p inhibited cardiomyocyte proliferation, and its inhibitor promoted cardiomyocyte proliferation in vitro and in vivo. Furthermore, we identified that miR‐29b‐3p influenced cardiomyocyte proliferation by targeting NOTCH2, which was down‐regulated in the RVOT of the CHD patients. Conclusion This study reveals that miR‐29b‐3p functions as a novel regulator of cardiac development and inhibits cardiomyocyte proliferation via NOTCH2, which provides novel insights into the aetiology and potential treatment of CHD.
Collapse
Affiliation(s)
- Qian Yang
- Translational Medical Center for Development and Disease, Institute of Pediatrics, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Fang Wu
- Translational Medical Center for Development and Disease, Institute of Pediatrics, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Yaping Mi
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Feng Wang
- Translational Medical Center for Development and Disease, Institute of Pediatrics, Shanghai Key Laboratory of Birth Defect, 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
| | - Xiaoshan Yang
- Department of Bioscience, Bengbu Medical College, Bengbu, China
| | - Ranran Zhang
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Lian Liu
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Yawen Zhang
- Translational Medical Center for Development and Disease, Institute of Pediatrics, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Youhua Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu Wang
- Cancer Metabolism Laboratory, Cancer Institute, Fudan University, Shanghai, China
| | - Mingqing Xu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Yonghao Gui
- Translational Medical Center for Development and Disease, Institute of Pediatrics, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, Shanghai, China.,Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
| | - Qiang Li
- Translational Medical Center for Development and Disease, Institute of Pediatrics, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
28
|
Burggren W, Filogonio R, Wang T. Cardiovascular shunting in vertebrates: a practical integration of competing hypotheses. Biol Rev Camb Philos Soc 2019; 95:449-471. [PMID: 31859458 DOI: 10.1111/brv.12572] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 10/30/2019] [Accepted: 11/07/2019] [Indexed: 12/20/2022]
Abstract
This review explores the long-standing question: 'Why do cardiovascular shunts occur?' An historical perspective is provided on previous research into cardiac shunts in vertebrates that continues to shape current views. Cardiac shunts and when they occur is then described for vertebrates. Nearly 20 different functional reasons have been proposed as specific causes of shunts, ranging from energy conservation to improved gas exchange, and including a plethora of functions related to thermoregulation, digestion and haemodynamics. It has even been suggested that shunts are merely an evolutionary or developmental relic. Having considered the various hypotheses involving cardiovascular shunting in vertebrates, this review then takes a non-traditional approach. Rather than attempting to identify the single 'correct' reason for the occurrence of shunts, we advance a more holistic, integrative approach that embraces multiple, non-exclusive suites of proposed causes for shunts, and indicates how these varied functions might at least co-exist, if not actually support each other as shunts serve multiple, concurrent physiological functions. It is argued that deposing the 'monolithic' view of shunting leads to a more nuanced view of vertebrate cardiovascular systems. This review concludes by suggesting new paradigms for testing the function(s) of shunts, including experimentally placing organ systems into conflict in terms of their perfusion needs, reducing sources of variation in physiological experiments, measuring possible compensatory responses to shunt ablation, moving experiments from the laboratory to the field, and using cladistics-related approaches in the choice of experimental animals.
Collapse
Affiliation(s)
- Warren Burggren
- Department of Biological Sciences, Developmental Integrative Biology Cluster, University of North Texas, Denton, TX, 76203-5220, U.S.A
| | - Renato Filogonio
- Department of Physiological Sciences, Federal University of São Carlos, São Carlos, SP, 13565-905, Brazil
| | - Tobias Wang
- Zoophysiology, Department of Bioscience, Aarhus University, Aarhus C, 8000, Denmark.,Aarhus Institute of Advanced Sciences (AIAS), Aarhus University, Aarhus C, 8000, Denmark
| |
Collapse
|
29
|
Shi Y, Li Y, Wang Y, Zhuang J, Wang H, Hu M, Mo X, Yue S, Chen Y, Fan X, Chen J, Cai W, Zhu X, Wan Y, Zhong Y, Ye X, Li F, Zhou Z, Dai G, Luo R, Ocorr K, Jiang Z, Li X, Zhu P, Wu X, Yuan W. The Functional Polymorphism R129W in the BVES Gene Is Associated with Sporadic Tetralogy of Fallot in the Han Chinese Population. Genet Test Mol Biomarkers 2019; 23:601-609. [PMID: 31386585 DOI: 10.1089/gtmb.2019.0085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background: Tetralogy of Fallot (TOF) accounts for ∼10% of congenital heart disease cases. The blood vessel epicardial substance (BVES) gene has been reported to play a role in the function of adult hearts. However, whether allelic variants in BVES contribute to the risk of TOF and its possible mechanism remains unknown. Methods: The open reading frame of the BVES gene was sequenced using samples from 146 TOF patients and 100 unrelated healthy controls. qRT-PCR and western blot assays were used to confirm the expression of mutated BVES variants in the TOF samples. The online software Polyphen2 and SIFT were used to predict the deleterious effects of the observed allelic variants. The effects of these allelic variants on the transcriptional activities of genes were examined using dual-fluorescence reporter assays. Results: We genotyped four single nucleotide polymorphisms (SNPs) in the BVES gene from each of the 146 TOF patients. Among them, the minor allelic frequencies of c.385C>T (p.R129W) were 0.035% in TOF, but ∼0.025% in 100 controls and the Chinese Millionome Database. This allelic variant was predicted to be a potentially harmful alteration by the Polyphen2 and SIFT softwares. qRT-PCR and western blot analyses indicated that the expression of BVES in the six right ventricular outflow tract samples with the c.385C>T allelic variant was significantly downregulated. A dual-fluorescence reporter system showed that the c.385C>T allelic variant significantly decreased the transcriptional activity of the BVES gene and also decreased transcription from the GATA4 and NKX2.5 promoters. Conclusions: c.385C>T (p.R129W) is a functional SNP of the BVES gene that reduces the transcriptional activity of BVES in vitro and in vivo in TOF tissues. This subsequently affects the transcriptional activities of GATA4 and NKX2.5 related to TOF. These findings suggest that c.385C>T may be associated with the risk of TOF in the Han Chinese population.
Collapse
Affiliation(s)
- Yan Shi
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yongqing Li
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yuequn Wang
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jian Zhuang
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Heng Wang
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Min Hu
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiaoyang Mo
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Shusheng Yue
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yu Chen
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiongwei Fan
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jimei Chen
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wanwan Cai
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiaolan Zhu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yongqi Wan
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ying Zhong
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiangli Ye
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Fang Li
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Zuoqiong Zhou
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China.,Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guo Dai
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Rong Luo
- Department of Cardiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Karen Ocorr
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California
| | - Zhigang Jiang
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiaoping Li
- Department of Cardiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Ping Zhu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiushan Wu
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Wuzhou Yuan
- State Key Lab of Development Biology of Freshwater Fish, Key Lab of MOE for Development Biology and Protein Chemistry, The Center for Heart Development, College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
30
|
Nakajima Y. Retinoic acid signaling in heart development. Genesis 2019; 57:e23300. [PMID: 31021052 DOI: 10.1002/dvg.23300] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/01/2019] [Accepted: 04/04/2019] [Indexed: 12/30/2022]
Abstract
Retinoic acid (RA) is a vitamin A metabolite that acts as a morphogen and teratogen. Excess or defective RA signaling causes developmental defects including in the heart. The heart develops from the anterior lateral plate mesoderm. Cardiogenesis involves successive steps, including formation of the primitive heart tube, cardiac looping, septation, chamber development, coronary vascularization, and completion of the four-chambered heart. RA is dispensable for primitive heart tube formation. Before looping, RA is required to define the anterior/posterior boundaries of the heart-forming mesoderm as well as to form the atrium and sinus venosus. In outflow tract elongation and septation, RA signaling is required to maintain/differentiate cardiogenic progenitors in the second heart field at the posterior pharyngeal arches level. Epicardium-secreted insulin-like growth factor, the expression of which is regulated by hepatic mesoderm-derived erythropoietin under the control of RA, promotes myocardial proliferation of the ventricular wall. Epicardium-derived RA induces the expression of angiogenic factors in the myocardium to form the coronary vasculature. In cardiogenic events at different stages, properly controlled RA signaling is required to establish the functional heart.
Collapse
Affiliation(s)
- Yuji Nakajima
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| |
Collapse
|
31
|
Gibb N, Lazic S, Yuan X, Deshwar AR, Leslie M, Wilson MD, Scott IC. Hey2 regulates the size of the cardiac progenitor pool during vertebrate heart development. Development 2018; 145:dev.167510. [PMID: 30355727 DOI: 10.1242/dev.167510] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 10/13/2018] [Indexed: 01/04/2023]
Abstract
A key event in heart development is the timely addition of cardiac progenitor cells, defects in which can lead to congenital heart defects. However, how the balance and proportion of progenitor proliferation versus addition to the heart is regulated remains poorly understood. Here, we demonstrate that Hey2 functions to regulate the dynamics of cardiac progenitor addition to the zebrafish heart. We found that the previously noted increase in myocardial cell number found in the absence of Hey2 function was due to a pronounced expansion in the size of the cardiac progenitor pool. Expression analysis and lineage tracing of hey2-expressing cells showed that hey2 is active in cardiac progenitors. Hey2 acted to limit proliferation of cardiac progenitors, prior to heart tube formation. Use of a transplantation approach demonstrated a likely cell-autonomous (in cardiac progenitors) function for Hey2. Taken together, our data suggest a previously unappreciated role for Hey2 in controlling the proliferative capacity of cardiac progenitors, affecting the subsequent contribution of late-differentiating cardiac progenitors to the developing vertebrate heart.
Collapse
Affiliation(s)
- Natalie Gibb
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Savo Lazic
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Ontario M5S 1A8, Canada
| | - Xuefei Yuan
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Ontario M5S 1A8, Canada
| | - Ashish R Deshwar
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Ontario M5S 1A8, Canada
| | - Meaghan Leslie
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Ontario M5S 1A8, Canada
| | - Michael D Wilson
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Ontario M5S 1A8, Canada
| | - Ian C Scott
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada .,Department of Molecular Genetics, University of Toronto, Ontario M5S 1A8, Canada.,Ted Rogers Centre for Heart Research, Toronto, Ontario M5G 1M1, Canada.,Heart and Stroke Richard Lewar Centres of Excellence in Cardiovascular Research, Toronto, Ontario M5S 3H2, Canada
| |
Collapse
|
32
|
Unolt M, Versacci P, Anaclerio S, Lambiase C, Calcagni G, Trezzi M, Carotti A, Crowley TB, Zackai EH, Goldmuntz E, Gaynor JW, Digilio MC, McDonald-McGinn DM, Marino B. Congenital heart diseases and cardiovascular abnormalities in 22q11.2 deletion syndrome: From well-established knowledge to new frontiers. Am J Med Genet A 2018; 176:2087-2098. [PMID: 29663641 PMCID: PMC6497171 DOI: 10.1002/ajmg.a.38662] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 12/11/2022]
Abstract
Congenital heart diseases (CHDs) and cardiovascular abnormalities are one of the pillars of clinical diagnosis of 22q11.2 deletion syndrome (22q11.2DS) and still represent the main cause of mortality in the affected children. In the past 30 years, much progress has been made in describing the anatomical patterns of CHD, in improving their diagnosis, medical treatment, and surgical procedures for these conditions, as well as in understanding the underlying genetic and developmental mechanisms. However, further studies are still needed to better determine the true prevalence of CHDs in 22q11.2DS, including data from prenatal studies and on the adult population, to further clarify the genetic mechanisms behind the high variability of phenotypic expression of 22q11.2DS, and to fully understand the mechanism responsible for the increased postoperative morbidity and for the premature death of these patients. Moreover, the increased life expectancy of persons with 22q11.2DS allowed the expansion of the adult population that poses new challenges for clinicians such as acquired cardiovascular problems and complexity related to multisystemic comorbidity. In this review, we provide a comprehensive review of the existing literature about 22q11.2DS in order to summarize the knowledge gained in the past years of clinical experience and research, as well as to identify the remaining gaps in comprehension of this syndrome and the possible future research directions.
Collapse
Affiliation(s)
- Marta Unolt
- Department of Pediatrics and Pediatric Neuropsychiatry, “Sapienza” University of Rome, Rome, Italy
| | - Paolo Versacci
- Department of Pediatrics and Pediatric Neuropsychiatry, “Sapienza” University of Rome, Rome, Italy
| | - Silvia Anaclerio
- Department of Pediatrics and Pediatric Neuropsychiatry, “Sapienza” University of Rome, Rome, Italy
| | - Caterina Lambiase
- Department of Pediatrics and Pediatric Neuropsychiatry, “Sapienza” University of Rome, Rome, Italy
| | - Giulio Calcagni
- Department of Pediatric Cardiology and Cardiac Surgery, Bambino Gesù Pediatric Hospital, Rome, Italy
| | - Matteo Trezzi
- Department of Pediatric Cardiology and Cardiac Surgery, Bambino Gesù Pediatric Hospital, Rome, Italy
| | - Adriano Carotti
- Department of Pediatric Cardiology and Cardiac Surgery, Bambino Gesù Pediatric Hospital, Rome, Italy
| | - Terrence Blaine Crowley
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Elaine H. Zackai
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Elizabeth Goldmuntz
- The Cardiac Center, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - James William Gaynor
- The Cardiac Center, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | | | - Bruno Marino
- Department of Pediatrics and Pediatric Neuropsychiatry, “Sapienza” University of Rome, Rome, Italy
| |
Collapse
|
33
|
Xie H, Zhang E, Hong N, Fu Q, Li F, Chen S, Yu Y, Sun K. Identification of TBX2 and TBX3 variants in patients with conotruncal heart defects by target sequencing. Hum Genomics 2018; 12:44. [PMID: 30223900 PMCID: PMC6142335 DOI: 10.1186/s40246-018-0176-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Background Conotruncal heart defects (CTDs) are heterogeneous congenital heart malformations that result from outflow tract dysplasia; however, the genetic determinants underlying CTDs remain unclear. Increasing evidence demonstrates that dysfunctional TBX2 and TBX3 result in outflow tract malformations, implying that both of them are involved in CTD pathogenesis. We screened for TBX2 and TBX3 variants in a large cohort of CTD patients (n = 588) and population-matched healthy controls (n = 300) by target sequencing and genetically analyzed the expression and function of these variants. Results The probably damaging variants p.R608W, p.T249I, and p.R616Q of TBX2 and p.A192T, p.M65L, and p.A562V of TBX3 were identified in CTD patients, but none in controls. All altered amino acids were highly conserved evolutionarily. Moreover, our data suggested that mRNA and protein expressions of TBX2 and TBX3 variants were altered compared with those of the wild-type. We screened PEA3 and MEF2C as novel downstream genes of TBX2 and TBX3, respectively. Functional analysis revealed that TBX2R608W and TBX2R616Q variant proteins further activated HAS2 promoter but failed to activate PEA3 promoter and that TBX3A192T and TBX3A562V variant proteins showed a reduced transcriptional activity over MEF2C promoter. Conclusions Our results indicate that the R608W and R616Q variants of TBX2 as well as the A192T and A562V variants of TBX3 contribute to CTD etiology; this was the first association of variants of TBX2 and TBX3 to CTDs based on a large population. Electronic supplementary material The online version of this article (10.1186/s40246-018-0176-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Huilin Xie
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Erge Zhang
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Nanchao Hong
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Qihua Fu
- Medical Laboratory, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Fen Li
- Department of Pediatric Cardiology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Sun Chen
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Yu Yu
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China.
| | - Kun Sun
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China.
| |
Collapse
|
34
|
Al-Zahrani RS, Alharbi SH, Tuwaijri RMA, Alzomaili BT, Althubaiti A, Yelbuz TM. Transposition of the great arteries: A laterality defect in the group of heterotaxy syndromes or an outflow tract malformation? Ann Pediatr Cardiol 2018; 11:237-249. [PMID: 30271012 PMCID: PMC6146851 DOI: 10.4103/apc.apc_24_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND/AIM Transposition of the great arteries (TGA) is traditionally classified as a "conotruncal heart defect", implying that TGA evolves from abnormal development of the outflow tract (OFT) of the embryonic heart. However, recently published genetic data suggest that TGA may be linked to laterality gene defects rather than OFT gene defects. The aim of our study was to clarify whether there is any statistically significant link between TGA and clinically diagnosed laterality defects (heterotaxy). METHODS Retrospective cross-sectional analysis of 533 patients diagnosed with TGA at our cardiac center over a period of 13 years (2002-2015). Hospital informatics and digital data recording systems were used for collecting patients' data and all patients were reviewed to check the echocardiograms for verification of the diagnosis, type (TGA, congenitally corrected TGA (ccTGA), and levo-position of the great arteries (LGA)), complexity of TGA, and all other variables (e.g., abdominal organ arrangement, cardiac position, presence or absence of other cardiac defects). RESULTS Of 533 TGA patients, 495 (92.9%) had the usual arrangement of the internal organs, 21 (3.9%) had mirror-imagery, 7 (1.3%) had left and 10 (1.8%) had right isomerism. 444 (83.3%) patients had TGA. The number of patients who had usual visceral arrangement in each TGA type was: 418 (94.1%) in TGA, 49 (92.4%) in ccTGA, and 28 (77.7%) in LGA. 6 (1.4%) TGA patients, 4 (11.1%) patients with LGA were found to have right isomerism, while no ccTGA patient presented with this asymmetry. 4 (0.9%) TGA patients, 1 (1.9%) ccTGA patient and 2 (5.6%) patients with LGA had left isomerism. Heterotaxy (mirror-imagery, left and right isomerism) was more associated with LGA than TGA or ccTGA with a statistically significant difference (P value of 0.001). CONCLUSION In contrast to recently published genetic data, our morphological data do not disclose a significant link between TGA and heterotaxy.
Collapse
Affiliation(s)
- Rana S Al-Zahrani
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Samaher H Alharbi
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Rawan M A Tuwaijri
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Bayan T Alzomaili
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Alaa Althubaiti
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Talat Mesud Yelbuz
- Department of Cardiac Sciences, King Abdulaziz Cardiac Center, Section of Pediatric Cardiology, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
35
|
Pinard A, Eudes N, Mitchell J, Bajolle F, Grelet M, Okoronkwo J, Bonnet D, Collod-Béroud G, Zaffran S. Analysis of HOXB1 gene in a cohort of patients with sporadic ventricular septal defect. Mol Biol Rep 2018; 45:1507-1513. [PMID: 29923154 DOI: 10.1007/s11033-018-4212-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/13/2018] [Indexed: 11/30/2022]
Abstract
Ventricular septal defect (VSD) including outlet VSD of double outlet right ventricle (DORV) and perimembranous VSD are among the most common congenital heart diseases found at birth. HOXB1 encodes a homeodomain transcription factor essential for normal cardiac outflow tract development. The aim of the present study was to investigate the possible genetic effect of sequence variations in HOXB1 on VSD. The coding regions and splice junctions of the HOXB1 gene were sequenced in 57 unrelated VSD patients. As a result, a homozygous c.74_82dup (p.Pro28delinsHisSerAlaPro) variant was identified in one individual with DORV. We also identified five previously reported polymorphisms (rs35114525, rs12946855, rs14534040, rs12939811, and rs7207109) in 18 patients (12 DORV and 6 perimembranous VSD). Our study did not show any pathogenic alterations in the coding region of HOXB1 among patients with VSD. To our knowledge this is the first study investigating the role of HOXB1 in nonsyndromic VSD, which provide more insight on the etiology of this disease.
Collapse
Affiliation(s)
- Amélie Pinard
- Aix Marseille Université, INSERM U1251, MMG, Marseille, France.,Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Nathalie Eudes
- Aix Marseille Université, INSERM U1251, MMG, Marseille, France
| | - Julia Mitchell
- Aix Marseille Université, INSERM U1251, MMG, Marseille, France.,Service de Chirurgie des Cardiopathies Congénitales, Hôpital Cardiologique Louis Pradel, Avenue du Doyen Lépine, 69394, Lyon, France
| | - Fanny Bajolle
- Centre de Référence Malformations Cardiaques Congénitales Complexes (M3C), Unité Médico-Chirurgicale de Cardiologie Congénitale et Pédiatrique, AP-HP, Hôpital Necker-Enfants-Malades, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maude Grelet
- Aix Marseille Université, INSERM U1251, MMG, Marseille, France
| | - Joséphine Okoronkwo
- Centre de Référence Malformations Cardiaques Congénitales Complexes (M3C), Unité Médico-Chirurgicale de Cardiologie Congénitale et Pédiatrique, AP-HP, Hôpital Necker-Enfants-Malades, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Damien Bonnet
- Centre de Référence Malformations Cardiaques Congénitales Complexes (M3C), Unité Médico-Chirurgicale de Cardiologie Congénitale et Pédiatrique, AP-HP, Hôpital Necker-Enfants-Malades, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Stéphane Zaffran
- Aix Marseille Université, INSERM U1251, MMG, Marseille, France. .,Faculté de Médecine, Aix Marseille Université, INSERM U1251, Marseille Medical Genetics, 27 Bd Jean Moulin, 13005, Marseille, France.
| |
Collapse
|
36
|
Versacci P, Pugnaloni F, Digilio MC, Putotto C, Unolt M, Calcagni G, Baban A, Marino B. Some Isolated Cardiac Malformations Can Be Related to Laterality Defects. J Cardiovasc Dev Dis 2018; 5:jcdd5020024. [PMID: 29724030 PMCID: PMC6023464 DOI: 10.3390/jcdd5020024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/21/2018] [Accepted: 04/25/2018] [Indexed: 12/22/2022] Open
Abstract
Human beings are characterized by a left–right asymmetric arrangement of their internal organs, and the heart is the first organ to break symmetry in the developing embryo. Aberrations in normal left–right axis determination during embryogenesis lead to a wide spectrum of abnormal internal laterality phenotypes, including situs inversus and heterotaxy. In more than 90% of instances, the latter condition is accompanied by complex and severe cardiovascular malformations. Atrioventricular canal defect and transposition of the great arteries—which are particularly frequent in the setting of heterotaxy—are commonly found in situs solitus with or without genetic syndromes. Here, we review current data on morphogenesis of the heart in human beings and animal models, familial recurrence, and upstream genetic pathways of left–right determination in order to highlight how some isolated congenital heart diseases, very common in heterotaxy, even in the setting of situs solitus, may actually be considered in the pathogenetic field of laterality defects.
Collapse
Affiliation(s)
- Paolo Versacci
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| | - Flaminia Pugnaloni
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| | - Maria Cristina Digilio
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital and Research Institute, 00165 Rome, Italy.
| | - Carolina Putotto
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| | - Marta Unolt
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| | - Giulio Calcagni
- Department of Pediatric Cardiology and Cardiac Surgery, Bambino Gesù Children's Hospital and Research Institute, 00165 Rome, Italy.
| | - Anwar Baban
- Department of Pediatric Cardiology and Cardiac Surgery, Bambino Gesù Children's Hospital and Research Institute, 00165 Rome, Italy.
| | - Bruno Marino
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| |
Collapse
|
37
|
The role of histone modification and a regulatory single-nucleotide polymorphism (rs2071166) in the Cx43 promoter in patients with TOF. Sci Rep 2017; 7:10435. [PMID: 28874875 PMCID: PMC5585261 DOI: 10.1038/s41598-017-10756-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/14/2017] [Indexed: 12/21/2022] Open
Abstract
Abnormal level of Cx43 expression could result in CHD. Epigenetic modification and disease-associated, non-coding SNPs might influence gene transcription and expression. Our study aimed to determine the role of histone modification and an rSNP (rs2071166) in the Cx43 promoter in patients with TOF. Our results indicate that H3K18ac bind to Cx43 promoter and that their levels are reduced in TOF patients relative to controls. The relationship between the non-coding SNP in the Cx43 gene and TOF patients was evaluated in 158 patients and 300 controls. The C allele of rs2071166 was confirmed to result in an increased risk of TOF (OR = 1.586, 95%CI 1.149–2.189). Individuals with the CC genotype at rs2071166 also showed a significant susceptibility to TOF (OR = 2.961, 95%CI 1.452–6.038). The mRNA level in TOF who were CC genotype was lower than that in patients with the AA/AC genotype. Functional analysis in cells and transgenic zebrafish models showed that rs2071166 decreased the activity of the promoter and could block the interaction between RXRα and RARE. This is the first study to illustrate that epigenetic modification and an rSNP in the Cx43 promoter region play a critical role in TOF by impacting the transcriptional activity and expression level of Cx43.
Collapse
|
38
|
Calcagni G, Unolt M, Digilio MC, Baban A, Versacci P, Tartaglia M, Baldini A, Marino B. Congenital heart disease and genetic syndromes: new insights into molecular mechanisms. Expert Rev Mol Diagn 2017; 17:861-870. [PMID: 28745539 DOI: 10.1080/14737159.2017.1360766] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Advances in genetics allowed a better definition of the role of specific genetic background in the etiology of syndromic congenital heart defects (CHDs). The identification of a number of disease genes responsible for different syndromes have led to the identification of several transcriptional regulators and signaling transducers and modulators that are critical for heart morphogenesis. Understanding the genetic background of syndromic CHDs allowed a better characterization of the genetic basis of non-syndromic CHDs. In this sense, the well-known association of typical CHDs in Down syndrome, 22q11.2 microdeletion and Noonan syndrome represent paradigms as chromosomal aneuploidy, chromosomal microdeletion and intragenic mutation, respectively. Area covered: For each syndrome the anatomical features, distinctive cardiac phenotype and molecular mechanisms are discussed. Moreover, the authors include recent genetic findings that may shed light on some aspects of still unclear molecular mechanisms of these syndromes. Expert commentary: Further investigations are needed to enhance the translational approach in the field of genetics of CHDs. When there is a well-established definition of genotype-phenotype (reverse medicine) and genotype-prognosis (predictive and personalized medicine) correlations, hopefully preventive medicine will make its way in this field. Subsequently a reduction will be achieved in the morbidity and mortality of children with CHDs.
Collapse
Affiliation(s)
- Giulio Calcagni
- a Department of Pediatric Cardiology and Cardiac Surgery , Bambino Gesù Children's Hospital and Research Institute , Rome , Italy
| | - Marta Unolt
- b Department of Pediatrics , Sapienza University , Rome , Italy
| | - Maria Cristina Digilio
- c Genetics and Rare Diseases Research Division , Bambino Gesù Children's Hospital and Research Institute , Rome , Italy
| | - Anwar Baban
- a Department of Pediatric Cardiology and Cardiac Surgery , Bambino Gesù Children's Hospital and Research Institute , Rome , Italy
| | - Paolo Versacci
- b Department of Pediatrics , Sapienza University , Rome , Italy
| | - Marco Tartaglia
- c Genetics and Rare Diseases Research Division , Bambino Gesù Children's Hospital and Research Institute , Rome , Italy
| | - Antonio Baldini
- d CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso; Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II , Naples , Italy
| | - Bruno Marino
- b Department of Pediatrics , Sapienza University , Rome , Italy
| |
Collapse
|
39
|
Scott CA, Marsden AN, Rebagliati MR, Zhang Q, Chamling X, Searby CC, Baye LM, Sheffield VC, Slusarski DC. Nuclear/cytoplasmic transport defects in BBS6 underlie congenital heart disease through perturbation of a chromatin remodeling protein. PLoS Genet 2017; 13:e1006936. [PMID: 28753627 PMCID: PMC5550010 DOI: 10.1371/journal.pgen.1006936] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 08/09/2017] [Accepted: 07/20/2017] [Indexed: 01/31/2023] Open
Abstract
Mutations in BBS6 cause two clinically distinct syndromes, Bardet-Biedl syndrome (BBS), a syndrome caused by defects in cilia transport and function, as well as McKusick-Kaufman syndrome, a genetic disorder characterized by congenital heart defects. Congenital heart defects are rare in BBS, and McKusick-Kaufman syndrome patients do not develop retinitis pigmentosa. Therefore, the McKusick-Kaufman syndrome allele may highlight cellular functions of BBS6 distinct from the presently understood functions in the cilia. In support, we find that the McKusick-Kaufman syndrome disease-associated allele, BBS6H84Y; A242S, maintains cilia function. We demonstrate that BBS6 is actively transported between the cytoplasm and nucleus, and that BBS6H84Y; A242S, is defective in this transport. We developed a transgenic zebrafish with inducible bbs6 to identify novel binding partners of BBS6, and we find interaction with the SWI/SNF chromatin remodeling protein Smarcc1a (SMARCC1 in humans). We demonstrate that through this interaction, BBS6 modulates the sub-cellular localization of SMARCC1 and find, by transcriptional profiling, similar transcriptional changes following smarcc1a and bbs6 manipulation. Our work identifies a new function for BBS6 in nuclear-cytoplasmic transport, and provides insight into the disease mechanism underlying the congenital heart defects in McKusick-Kaufman syndrome patients. To understand how mutations in one gene can cause two distinct human syndromes (McKusick-Kaufman syndrome and Bardet-Bield syndrome), we investigated the cellular functions of the implicated gene BBS6. We found that BBS6 is actively transported between the cytoplasm and nucleus, and this interaction is disrupted in McKusick-Kaufman syndrome, but not Bardet-Biedl syndrome. We find that by manipulating BBS6, we can affect another protein, SMARCC1, which has a direct role in regulating gene expression. When we profiled these changes in gene expression, we find that many genes, which can be directly linked to the symptoms of McKusick-Kaufman syndrome, are affected. Therefore, our data support that the nuclear-cytoplasmic transport defect of BBS6, through disruption of proteins controlling gene expression, cause the symptoms observed in McKusick-Kaufman syndrome patients.
Collapse
Affiliation(s)
- Charles Anthony Scott
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Autumn N. Marsden
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, United States of America
| | - Michael R. Rebagliati
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Qihong Zhang
- Department of Pediatrics and Ophthalmology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Xitiz Chamling
- Department of Pediatrics and Ophthalmology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Charles C. Searby
- Department of Pediatrics and Ophthalmology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Lisa M. Baye
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Val C. Sheffield
- Department of Pediatrics and Ophthalmology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Wynn Institute for Vision Research University of Iowa, Iowa City, Iowa, United States of America
| | - Diane C. Slusarski
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
- Wynn Institute for Vision Research University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
40
|
Aghajanian H, Cho YK, Rizer NW, Wang Q, Li L, Degenhardt K, Jain R. Pdgfrα functions in endothelial-derived cells to regulate neural crest cells and the development of the great arteries. Dis Model Mech 2017; 10:1101-1108. [PMID: 28714851 PMCID: PMC5611965 DOI: 10.1242/dmm.029710] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 07/05/2017] [Indexed: 12/16/2022] Open
Abstract
Originating as a single vessel emerging from the embryonic heart, the truncus arteriosus must septate and remodel into the aorta and pulmonary artery to support postnatal life. Defective remodeling or septation leads to abnormalities collectively known as conotruncal defects, which are associated with significant mortality and morbidity. Multiple populations of cells must interact to coordinate outflow tract remodeling, and the cardiac neural crest has emerged as particularly important during this process. Abnormalities in the cardiac neural crest have been implicated in the pathogenesis of multiple conotruncal defects, including persistent truncus arteriosus, double outlet right ventricle and tetralogy of Fallot. However, the role of the neural crest in the pathogenesis of another conotruncal abnormality, transposition of the great arteries, is less well understood. In this report, we demonstrate an unexpected role of Pdgfra in endothelial cells and their derivatives during outflow tract development. Loss of Pdgfra in endothelium and endothelial-derived cells results in double outlet right ventricle and transposition of the great arteries. Our data suggest that loss of Pdgfra in endothelial-derived mesenchyme in the outflow tract endocardial cushions leads to a secondary defect in neural crest migration during development. Summary: Loss of Pdgfrα in endothelial-derived mesenchyme results in defective neural crest behavior and is associated with conotruncal defects including, surprisingly, transposition of the great arteries.
Collapse
Affiliation(s)
- Haig Aghajanian
- Departments of Medicine and Cell and Developmental Biology, Penn Cardiovascular Institute, Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Young Kuk Cho
- Department of Pediatrics, Chonnam National University Medical School, Gwangju, 61186, South Korea
| | - Nicholas W Rizer
- Departments of Medicine and Cell and Developmental Biology, Penn Cardiovascular Institute, Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Qiaohong Wang
- Departments of Medicine and Cell and Developmental Biology, Penn Cardiovascular Institute, Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Li Li
- Departments of Medicine and Cell and Developmental Biology, Penn Cardiovascular Institute, Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karl Degenhardt
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rajan Jain
- Departments of Medicine and Cell and Developmental Biology, Penn Cardiovascular Institute, Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
41
|
Agarwal A, Harris IS, Mahadevan VS, Foster E. Coexistence of abnormal systolic motion of mitral valve in a consecutive group of 324 adult Tetralogy of Fallot patients assessed with echocardiography. Open Heart 2017; 3:e000518. [PMID: 28123759 PMCID: PMC5237749 DOI: 10.1136/openhrt-2016-000518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/02/2016] [Accepted: 11/08/2016] [Indexed: 12/18/2022] Open
Abstract
Background The presence of mitral valve prolapse (MVP) in congenital heart disease (CHD) patients is not well described. Tetralogy of Fallot (TOF) is the most common cyanotic CHD associated with overall good long-term survival after palliation. Since MVP is more often identified in adults and TOF patients are now surviving longer, we thus sought to perform this cohort study with a case–control design to (1) determine the prevalence of MVP and systolic displacement of mitral leaflets (SDML) in adult TOF patients, and (2) describe their clinical and imaging characteristics. Methods Retrospective interrogation of our echocardiography database identified 328 consecutive TOF patients ≥18 years from 1 January 2000 to 31 December 2014. All images were reviewed to identify patients with concomitant MVP (prolapse >2 mm beyond the long-axis annular plane) or SDML (<2 mm beyond the annular plane). Results 26 (8%) TOF patients fulfilled criteria for systolic mitral valve abnormality (SMVA) (15 MVP; 11 SDML). 2 had moderate to severe mitral regurgitation requiring repair. When compared with 52 TOF patients without SMVA, those with SMVA were more likely to be females (60.7% vs 33.9%, p=0.03), less likely to have transannular patch (52% vs 97.4%, p<0.0001), had lower right ventricular ejection fraction (36.5% vs 43.8%, p=0.03) and a trend towards increased risk of atrial (44% vs 30.4%, p=0.5) and ventricular arrhythmias (32% vs 25.5%, p=0.6). On multivariate logistic regression, SMVA was independently associated with the absence of transannular patch (p=0.002) and atrial arrhythmias (p=0.04). Conclusions In this series of adult TOF patients, we describe a novel finding of a high prevalence of systolic mitral valve abnormalities.
Collapse
Affiliation(s)
- Anushree Agarwal
- University of California San Francisco (UCSF) , San Francisco, California , USA
| | - Ian S Harris
- University of California San Francisco (UCSF) , San Francisco, California , USA
| | - Vaikom S Mahadevan
- University of California San Francisco (UCSF) , San Francisco, California , USA
| | - Elyse Foster
- University of California San Francisco (UCSF) , San Francisco, California , USA
| |
Collapse
|
42
|
Gittenberger-de Groot AC, Hoppenbrouwers T, Miquerol L, Kosaka Y, Poelmann RE, Wisse LJ, Yost HJ, Jongbloed MRM, Deruiter MC, Brunelli L. 14-3-3epsilon controls multiple developmental processes in the mouse heart. Dev Dyn 2016; 245:1107-1123. [PMID: 27580238 PMCID: PMC5065397 DOI: 10.1002/dvdy.24440] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 08/10/2016] [Accepted: 08/16/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND 14-3-3ε plays an important role in the maturation of the compact ventricular myocardium by modulating the cardiomyocyte cell cycle via p27kip1 . However, additional cardiac defects are possible given the ubiquitous expression pattern of this protein. RESULTS Germ line deletion of 14-3-3ε led to malalignment of both the outflow tract (OFT) and atrioventricular (AV) cushions, with resulting tricuspid stenosis and atresia, mitral valve abnormalities, and perimembranous ventricular septal defects (VSDs). We confirmed myocardial non-compaction and detected a spongy septum with muscular VSDs and blebbing of the epicardium. These defects were associated with abnormal patterning of p27kip1 expression in the subendocardial and possibly the epicardial cell populations. In addition to abnormal pharyngeal arch artery patterning, we found deep endocardial recesses and paucity of intramyocardial coronary vasculature as a result of defective coronary plexus remodeling. CONCLUSIONS The malalignment of both endocardial cushions provides a new explanation for tricuspid and mitral valve defects, while myocardial non-compaction provides the basis for the abnormal coronary vasculature patterning. These abnormalities might arise from p27kip1 dysregulation and a resulting defect in epithelial-to-mesenchymal transformation. These data suggest that 14-3-3ε, in addition to left ventricular non-compaction (LVNC), might be linked to different forms of congenital heart disease (CHD). Developmental Dynamics 245:1107-1123, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Adriana C Gittenberger-de Groot
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Tamara Hoppenbrouwers
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Yasuhiro Kosaka
- Department of Pediatrics (Neonatology), University of Utah School of Medicine, Salt Lake City, Utah
| | | | - Lambertus J Wisse
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - H Joseph Yost
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah
| | - Monique R M Jongbloed
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marco C Deruiter
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Luca Brunelli
- Department of Pediatrics (Neonatology), University of Utah School of Medicine, Salt Lake City, Utah.
| |
Collapse
|
43
|
Shaikh Qureshi WM, Miao L, Shieh D, Li J, Lu Y, Hu S, Barroso M, Mazurkiewicz J, Wu M. Imaging Cleared Embryonic and Postnatal Hearts at Single-cell Resolution. J Vis Exp 2016. [PMID: 27768060 DOI: 10.3791/54303] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Single clonal tracing and analysis at the whole-heart level can determine cardiac progenitor cell behavior and differentiation during cardiac development, and allow for the study of the cellular and molecular basis of normal and abnormal cardiac morphogenesis. Recent emerging technologies of retrospective single clonal analyses make the study of cardiac morphogenesis at single cell resolution feasible. However, tissue opacity and light scattering of the heart as imaging depth is increased hinder whole-heart imaging at single cell resolution. To overcome these obstacles, a whole-embryo clearing system that can render the heart highly transparent for both illumination and detection must be developed. Fortunately, in the last several years, many methodologies for whole-organism clearing systems such as CLARITY, Scale, SeeDB, ClearT, 3DISCO, CUBIC, DBE, BABB and PACT have been reported. This lab is interested in the cellular and molecular mechanisms of cardiac morphogenesis. Recently, we established single cell lineage tracing via the ROSA26-CreERT2; ROSA26-Confetti system to sparsely label cells during cardiac development. We adapted several whole embryo-clearing methodologies including Scale and CUBIC (clear, unobstructed brain imaging cocktails and computational analysis) to clear the embryo in combination with whole mount staining to image single clones inside the heart. The heart was successfully imaged at single cell resolution. We found that Scale can clear the embryonic heart, but cannot effectively clear the postnatal heart, while CUBIC can clear the postnatal heart, but damages the embryonic heart by dissolving the tissue. The methods described here will permit the study of gene function at a single clone resolution during cardiac morphogenesis, which, in turn, can reveal the cellular and molecular basis of congenital heart defects.
Collapse
Affiliation(s)
| | - Lianjie Miao
- Department of Molecular and Cellular Physiology, Albany Medical College
| | - David Shieh
- Department of Molecular and Cellular Physiology, Albany Medical College
| | - Jingjing Li
- Department of Molecular and Cellular Physiology, Albany Medical College
| | - Yangyang Lu
- Department of Molecular and Cellular Physiology, Albany Medical College
| | - Saiyang Hu
- Department of Molecular and Cellular Physiology, Albany Medical College
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College
| | - Joseph Mazurkiewicz
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| | - Mingfu Wu
- Department of Molecular and Cellular Physiology, Albany Medical College;
| |
Collapse
|
44
|
Nakajima Y. Mechanism responsible for D-transposition of the great arteries: Is this part of the spectrum of right isomerism? Congenit Anom (Kyoto) 2016; 56:196-202. [PMID: 27329052 DOI: 10.1111/cga.12176] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/26/2016] [Accepted: 06/15/2016] [Indexed: 12/25/2022]
Abstract
D-transposition of the great arteries (TGA) is one of the most common conotruncal heart defects at birth and is characterized by a discordant ventriculoarterial connection with a concordant atrioventricular connection. The morphological etiology of TGA is an inverted or arrested rotation of the heart outflow tract (OFT, conotruncus), by which the aorta is transposed in the right ventral direction to the pulmonary trunk. The rotational defect of the OFT is thought to be attributed to hypoplasia of the subpulmonic conus, which originates from the left anterior heart field (AHF) residing in the mesodermal core of the first and second pharyngeal arches. AHF, especially on the left, at the early looped heart stage (corresponding to Carnegie stage 10-11 in the human embryo) is one of the regions responsible for the impediment that causes TGA morphology. In human or experimentally produced right isomerism, malposition of the great arteries including D-TGA is frequently associated. Mutations in genes involving left-right (L-R) asymmetry, such as NODAL, ACTRIIB and downstream target FOXH1, have been found in patients with right isomerism as well as in isolated TGA. The downstream pathways of Nodal-Foxh1 play a critical role not only in L-R determination in the lateral plate mesoderm but also in myocardial specification and differentiation in the AHF, suggesting that TGA is a phenotype in heterotaxia as well as the primary developmental defect of the AHF.
Collapse
Affiliation(s)
- Yuji Nakajima
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| |
Collapse
|
45
|
Síndrome de deleción 22q11: bases embriológicas y algoritmo diagnóstico. REVISTA COLOMBIANA DE CARDIOLOGÍA 2016. [DOI: 10.1016/j.rccar.2016.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
46
|
Jahangiri L, Sharpe M, Novikov N, González-Rosa JM, Borikova A, Nevis K, Paffett-Lugassy N, Zhao L, Adams M, Guner-Ataman B, Burns CE, Burns CG. The AP-1 transcription factor component Fosl2 potentiates the rate of myocardial differentiation from the zebrafish second heart field. Development 2016; 143:113-22. [PMID: 26732840 DOI: 10.1242/dev.126136] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The vertebrate heart forms through successive phases of cardiomyocyte differentiation. Initially, cardiomyocytes derived from first heart field (FHF) progenitors assemble the linear heart tube. Thereafter, second heart field (SHF) progenitors differentiate into cardiomyocytes that are accreted to the poles of the heart tube over a well-defined developmental window. Although heart tube elongation deficiencies lead to life-threatening congenital heart defects, the variables controlling the initiation, rate and duration of myocardial accretion remain obscure. Here, we demonstrate that the AP-1 transcription factor, Fos-like antigen 2 (Fosl2), potentiates the rate of myocardial accretion from the zebrafish SHF. fosl2 mutants initiate accretion appropriately, but cardiomyocyte production is sluggish, resulting in a ventricular deficit coupled with an accumulation of SHF progenitors. Surprisingly, mutant embryos eventually correct the myocardial deficit by extending the accretion window. Overexpression of Fosl2 also compromises production of SHF-derived ventricular cardiomyocytes, a phenotype that is consistent with precocious depletion of the progenitor pool. Our data implicate Fosl2 in promoting the progenitor to cardiomyocyte transition and uncover the existence of regulatory mechanisms to ensure appropriate SHF-mediated cardiomyocyte contribution irrespective of embryonic stage.
Collapse
Affiliation(s)
- Leila Jahangiri
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Michka Sharpe
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Natasha Novikov
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Juan Manuel González-Rosa
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Asya Borikova
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Kathleen Nevis
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Noelle Paffett-Lugassy
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Long Zhao
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Meghan Adams
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Burcu Guner-Ataman
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Caroline E Burns
- Cardiovascular Research Center, Department of Medicine, 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, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
47
|
Hox Genes in Cardiovascular Development and Diseases. J Dev Biol 2016; 4:jdb4020014. [PMID: 29615581 PMCID: PMC5831787 DOI: 10.3390/jdb4020014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 03/16/2016] [Accepted: 03/23/2016] [Indexed: 11/23/2022] Open
Abstract
Congenital heart defects (CHD) are the leading cause of death in the first year of life. Over the past 20 years, much effort has been focused on unraveling the genetic bases of CHD. In particular, studies in human genetics coupled with those of model organisms have provided valuable insights into the gene regulatory networks underlying CHD pathogenesis. Hox genes encode transcription factors that are required for the patterning of the anterior–posterior axis in the embryo. In this review, we focus on the emerging role of anteriorly expressed Hox genes (Hoxa1, Hoxb1, and Hoxa3) in cardiac development, specifically their contribution to patterning of cardiac progenitor cells and formation of the great arteries. Recent evidence regarding the cooperative regulation of heart development by Hox proteins with members of the TALE-class of homeodomain proteins such as Pbx and Meis transcription factors is also discussed. These findings are highly relevant to human pathologies as they pinpoint new genes that increase susceptibility to cardiac anomalies and provide novel mechanistic insights into CHD.
Collapse
|
48
|
Li FF, Han Y, Shi S, Li X, Zhu XD, Zhou J, Shao QL, Li XQ, Liu SL. Characterization of Transcriptional Repressor Gene MSX1 Variations for Possible Associations with Congenital Heart Diseases. PLoS One 2015; 10:e0142666. [PMID: 26556783 PMCID: PMC4640503 DOI: 10.1371/journal.pone.0142666] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/26/2015] [Indexed: 01/26/2023] Open
Abstract
Background The human heart consists of several cell types with distinct lineage origins. Interactions between these cardiac progenitors are very important for heart formation. The muscle segment homeobox gene family plays a key role in the cell morphogenesis and growth, controlled cellular proliferation, differentiation, and apoptosis, but the relationships between the genetic abnormalities and CHD phenotypes still remain largely unknown. The aim of this work was to evaluate variations in MSX1 and MSX2 for their possible associations with CHD. Methods We sequenced the MSX1 and MSX2 genes for 300 Chinese Han CHD patients and 400 normal controls and identified the variations. The statistical analyses were conducted using Chi-Square Tests as implemented in SPSS (version 19.0). The Hardy-Weinberg equilibrium test of the population was carried out using the online software OEGE. Results Six variations rs4647952, rs2048152, rs4242182, rs61739543, rs111542301 and rs3087539 were identified in the MSX2 gene, but the genetic heterozygosity of those SNPs was very low. In contrast, the genetic heterozygosity of two variations rs3821949 near the 5’UTR and rs12532 within 3’UTR of the MSX1 gene was considerably high. Statistical analyses showed that rs3821949 and rs12532 were associated with the risk of CHD (specifically VSD). Conclusions The SNPs rs3821949 and rs12532 in the MSX1 gene were associated with CHD in Chinese Han populations.
Collapse
Affiliation(s)
- Fei-Feng Li
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Ying Han
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuai Shi
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xia Li
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
| | - Xi-Dong Zhu
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jing Zhou
- Intensive care unit, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qing-Liang Shao
- Department of Neonatalogy, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xue-Qi Li
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
- * E-mail: (S-LL); (X-QL)
| | - Shu-Lin Liu
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Canada
- * E-mail: (S-LL); (X-QL)
| |
Collapse
|
49
|
Martin PS, Kloesel B, Norris RA, Lindsay M, Milan D, Body SC. Embryonic Development of the Bicuspid Aortic Valve. J Cardiovasc Dev Dis 2015; 2:248-272. [PMID: 28529942 PMCID: PMC5438177 DOI: 10.3390/jcdd2040248] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bicuspid aortic valve (BAV) is the most common congenital valvular heart defect with an overall frequency of 0.5%–1.2%. BAVs result from abnormal aortic cusp formation during valvulogenesis, whereby adjacent cusps fuse into a single large cusp resulting in two, instead of the normal three, aortic cusps. Individuals with BAV are at increased risk for ascending aortic disease, aortic stenosis and coarctation of the aorta. The frequent occurrence of BAV and its anatomically discrete but frequent co-existing diseases leads us to suspect a common cellular origin. Although autosomal-dominant transmission of BAV has been observed in a few pedigrees, notably involving the gene NOTCH1, no single-gene model clearly explains BAV inheritance, implying a complex genetic model involving interacting genes. Several sequencing studies in patients with BAV have identified rare and uncommon mutations in genes of cardiac embryogenesis. But the extensive cell-cell signaling and multiple cellular origins involved in cardiac embryogenesis preclude simplistic explanations of this disease. In this review, we examine the series of events from cellular and transcriptional embryogenesis of the heart, to development of the aortic valve.
Collapse
Affiliation(s)
- Peter S. Martin
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St., Th724, Boston, MA 02115, USA; E-Mails: (P.S.M.); (B.K.)
| | - Benjamin Kloesel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St., Th724, Boston, MA 02115, USA; E-Mails: (P.S.M.); (B.K.)
| | - Russell A. Norris
- Department of Regenerative Medicine and Cell Biology, Children’s Research Institute, Medical University of South Carolina, 173 Ashley St, Charleston, SC 29403, USA; E-Mail:
| | - Mark Lindsay
- Cardiovascular Research Center, Richard B. Simches Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; E-Mails: (M.L.); (D.M.)
| | - David Milan
- Cardiovascular Research Center, Richard B. Simches Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; E-Mails: (M.L.); (D.M.)
| | - Simon C. Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St., Th724, Boston, MA 02115, USA; E-Mails: (P.S.M.); (B.K.)
- Author to whom correspondence should be addressed: E-Mail: ; Tel.: +1-617-732-7330; Fax: +1-617-730-2813
| |
Collapse
|
50
|
Lewandowski SL, Janardhan HP, Trivedi CM. Histone Deacetylase 3 Coordinates Deacetylase-independent Epigenetic Silencing of Transforming Growth Factor-β1 (TGF-β1) to Orchestrate Second Heart Field Development. J Biol Chem 2015; 290:27067-27089. [PMID: 26420484 DOI: 10.1074/jbc.m115.684753] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Indexed: 11/06/2022] Open
Abstract
About two-thirds of human congenital heart disease involves second heart field-derived structures. Histone-modifying enzymes, histone deacetylases (HDACs), regulate the epigenome; however, their functions within the second heart field remain elusive. Here we demonstrate that histone deacetylase 3 (HDAC3) orchestrates epigenetic silencing of Tgf-β1, a causative factor in congenital heart disease pathogenesis, in a deacetylase-independent manner to regulate development of second heart field-derived structures. In murine embryos lacking HDAC3 in the second heart field, increased TGF-β1 bioavailability is associated with ascending aortic dilatation, outflow tract malrotation, overriding aorta, double outlet right ventricle, aberrant semilunar valve development, bicuspid aortic valve, ventricular septal defects, and embryonic lethality. Activation of TGF-β signaling causes aberrant endothelial-to-mesenchymal transition and altered extracellular matrix homeostasis in HDAC3-null outflow tracts and semilunar valves, and pharmacological inhibition of TGF-β rescues these defects. HDAC3 recruits components of the PRC2 complex, methyltransferase EZH2, EED, and SUZ12, to the NCOR complex to enrich trimethylation of Lys-27 on histone H3 at the Tgf-β1 regulatory region and thereby maintains epigenetic silencing of Tgf-β1 specifically within the second heart field-derived mesenchyme. Wild-type HDAC3 or catalytically inactive HDAC3 expression rescues aberrant endothelial-to-mesenchymal transition and epigenetic silencing of Tgf-β1 in HDAC3-null outflow tracts and semilunar valves. These findings reveal that epigenetic dysregulation within the second heart field is a predisposing factor for congenital heart disease.
Collapse
Affiliation(s)
- Sara L Lewandowski
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Harish P Janardhan
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Chinmay M Trivedi
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605.
| |
Collapse
|