Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 304
Filtrar
1.
Environ Toxicol Pharmacol ; 89: 103776, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34798236

RESUMO

Triphenyl phosphate (TPHP) is an organophosphate ester-based plasticizer and flame retardant. The objective of this study was to identify the potential role of epidermal ionocytes in mediating TPHP-induced pericardial edema within zebrafish embryos. Exposure to TPHP from 24 to 72 h post fertilization (hpf) resulted in a significant increase in pericardial edema and the number of ionocytes at 72 hpf relative to time-matched embryos treated with vehicle. In addition, co-exposure of embryos to mannitol (an osmotic diuretic) blocked TPHP-induced pericardial edema and effects on ionocyte abundance. However, knockdown of ATPase1a1.4 - an abundant Na+/K+-ATPase localized to epidermal ionocytes - mitigated TPHP-induced effects on ionocyte abundance but not pericardial edema, whereas co-exposure of embryos to ouabain - a Na+/K+-ATPase inhibitor - enhanced TPHP-induced pericardial edema but not ionocyte abundance. Overall, our findings suggest that TPHP may have multiple mechanisms of toxicity leading to an increase in ionocyte abundance and pericardial edema within developing zebrafish embryos.


Assuntos
Células Epidérmicas/efeitos dos fármacos , Organofosfatos/toxicidade , Pericárdio/efeitos dos fármacos , Animais , Edema/induzido quimicamente , Embrião não Mamífero/efeitos dos fármacos , Retardadores de Chama/toxicidade , Pericárdio/embriologia , Peixe-Zebra/embriologia
2.
Int J Mol Sci ; 22(14)2021 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-34299295

RESUMO

Nearly three decades ago, the Wilms' tumor suppressor Wt1 was identified as a crucial regulator of heart development. Wt1 is a zinc finger transcription factor with multiple biological functions, implicated in the development of several organ systems, among them cardiovascular structures. This review summarizes the results from many research groups which allowed to establish a relevant function for Wt1 in cardiac development and disease. During development, Wt1 is involved in fundamental processes as the formation of the epicardium, epicardial epithelial-mesenchymal transition, coronary vessel development, valve formation, organization of the cardiac autonomous nervous system, and formation of the cardiac ventricles. Wt1 is further implicated in cardiac disease and repair in adult life. We summarize here the current knowledge about expression and function of Wt1 in heart development and disease and point out controversies to further stimulate additional research in the areas of cardiac development and pathophysiology. As re-activation of developmental programs is considered as paradigm for regeneration in response to injury, understanding of these processes and the molecules involved therein is essential for the development of therapeutic strategies, which we discuss on the example of WT1.


Assuntos
Coração/crescimento & desenvolvimento , Miocárdio/metabolismo , Proteínas WT1/metabolismo , Animais , Transição Epitelial-Mesenquimal/fisiologia , Expressão Gênica/genética , Coração/embriologia , Cardiopatias/genética , Cardiopatias/metabolismo , Cardiopatias/fisiopatologia , Humanos , Pericárdio/embriologia , Pericárdio/patologia , Fatores de Transcrição/metabolismo , Proteínas WT1/genética , Proteínas WT1/fisiologia
3.
Nat Commun ; 12(1): 4155, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34230480

RESUMO

The organization of an integrated coronary vasculature requires the specification of immature endothelial cells (ECs) into arterial and venous fates based on their localization within the heart. It remains unclear how spatial information controls EC identity and behavior. Here we use single-cell RNA sequencing at key developmental timepoints to interrogate cellular contributions to coronary vessel patterning and maturation. We perform transcriptional profiling to define a heterogenous population of epicardium-derived cells (EPDCs) that express unique chemokine signatures. We identify a population of Slit2+ EPDCs that emerge following epithelial-to-mesenchymal transition (EMT), which we term vascular guidepost cells. We show that the expression of guidepost-derived chemokines such as Slit2 are induced in epicardial cells undergoing EMT, while mesothelium-derived chemokines are silenced. We demonstrate that epicardium-specific deletion of myocardin-related transcription factors in mouse embryos disrupts the expression of key guidance cues and alters EPDC-EC signaling, leading to the persistence of an immature angiogenic EC identity and inappropriate accumulation of ECs on the epicardial surface. Our study suggests that EC pathfinding and fate specification is controlled by a common mechanism and guided by paracrine signaling from EPDCs linking epicardial EMT to EC localization and fate specification in the developing heart.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/metabolismo , Pericárdio/citologia , Pericárdio/metabolismo , Animais , Quimiocinas , Vasos Coronários/metabolismo , Embrião de Mamíferos , Transição Epitelial-Mesenquimal , Expressão Gênica , Coração , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso , Proteínas Nucleares , Pericárdio/embriologia , Fator de Resposta Sérica , Transdução de Sinais , Transativadores , Fatores de Transcrição/metabolismo , Transcriptoma
4.
Science ; 371(6533)2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33414188

RESUMO

The mammalian heart is derived from multiple cell lineages; however, our understanding of when and how the diverse cardiac cell types arise is limited. We mapped the origin of the embryonic mouse heart at single-cell resolution using a combination of transcriptomic, imaging, and genetic lineage labeling approaches. This mapping provided a transcriptional and anatomic definition of cardiac progenitor types. Furthermore, it revealed a cardiac progenitor pool that is anatomically and transcriptionally distinct from currently known cardiac progenitors. Besides contributing to cardiomyocytes, these cells also represent the earliest progenitor of the epicardium, a source of trophic factors and cells during cardiac development and injury. This study provides detailed insights into the formation of early cardiac cell types, with particular relevance to the development of cell-based cardiac regenerative therapies.


Assuntos
Coração/embriologia , Mioblastos Cardíacos/metabolismo , Miocárdio/citologia , Pericárdio/citologia , Pericárdio/embriologia , Animais , Diferenciação Celular/genética , Perfilação da Expressão Gênica , Camundongos , Mioblastos Cardíacos/classificação , Mioblastos Cardíacos/citologia , Miócitos Cardíacos/citologia , Análise de Célula Única , Transcriptoma
5.
Dev Dyn ; 249(12): 1455-1469, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33103836

RESUMO

BACKGROUND: The epicardium is the outer mesothelial layer of the heart. It encloses the myocardium and plays key roles in heart development and regeneration. It derives from the proepicardium (PE), cell clusters that appear in the dorsal pericardium (DP) close to the atrioventricular canal and the venous pole of the heart, and are released into the pericardial cavity. PE cells are advected around the beating heart until they attach to the myocardium. Bmp and Notch signaling influence PE formation, but it is unclear how both signaling pathways interact during this process in the zebrafish. RESULTS: Here, we show that the developing PE is influenced by Notch signaling derived from the endothelium. Overexpression of the intracellular receptor of notch in the endothelium enhances bmp expression, increases the number of pSmad1/5 positive cells in the DP and PE, and enhances PE formation. On the contrary, pharmacological inhibition of Notch1 impairs PE formation. bmp2b overexpression can rescue loss of PE formation in the presence of a Notch1 inhibitor, but Notch gain-of-function could not recover PE formation in the absence of Bmp signaling. CONCLUSIONS: Endothelial Notch signaling activates bmp expression in the heart tube, which in turn induces PE cluster formation from the DP layer.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Coração/embriologia , Organogênese/fisiologia , Pericárdio/embriologia , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular/fisiologia , Pericárdio/metabolismo , Peixe-Zebra
6.
Histol Histopathol ; 35(9): 1035-1046, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32633330

RESUMO

The epicardium is the outer mesothelial layer of the heart. It covers the myocardium and plays important roles in both heart development and regeneration. It is derived from the proepicardium (PE), groups of cells that emerges at early developmental stages from the dorsal pericardial layer (DP) close to the atrio-ventricular canal and the venous pole of the heart-tube. In zebrafish, PE cells extrude apically into the pericardial cavity as a consequence of DP tissue constriction, a process that is dependent on Bmp pathway signaling. Expression of the transcription factor Wilms tumor-1, Wt1, which is a leader of important morphogenetic events such as apoptosis regulation or epithelial-mesenchymal cell transition, is also necessary during PE formation. In this study, we used the zebrafish model to compare intensity level of the wt1a reporter line epi:GFP in PE and its original tissue, the DP. We found that GFP is present at higher intensity level in the PE tissue, and differentially wt1 expression at pericardial tissues could be involved in the PE formation process. Our results reveal that bmp2b overexpression leads to enhanced GFP level both in DP and in PE tissues.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Organogênese/genética , Pericárdio/embriologia , Proteínas WT1/genética , Proteínas de Peixe-Zebra/genética , Animais , Pericárdio/metabolismo , Proteínas WT1/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
7.
Sci Rep ; 10(1): 9334, 2020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32518241

RESUMO

Cardiovascular development is a complex developmental process in which multiple cell lineages are involved, namely the deployment of first and second heart fields. Beside the contribution of these cardiogenic fields, extracardiac inputs to the developing heart are provided by the migrating cardiac neural crest cells and the proepicardial derived cells. The proepicardium (PE) is a transitory cauliflower-like structure located between the cardiac and hepatic primordia. The PE is constituted by an internal mesenchymal component surrounded by an external epithelial lining. With development, cells derived from the proepicardium migrate to the neighboring embryonic heart and progressive cover the most external surface, leading to the formation of the embryonic epicardium. Experimental evidence in chicken have nicely demonstrated that epicardial derived cells can distinctly contribute to fibroblasts, endothelial and smooth muscle cells. Surprisingly, isolation of the developing PE anlage and ex vivo culturing spontaneously lead to differentiation into beating cardiomyocytes, a process that is enhanced by Bmp but halted by Fgf administration. In this study we provide a comprehensive characterization of the developmental expression profile of multiple microRNAs during epicardial development in chicken. Subsequently, we identified that miR-125, miR-146, miR-195 and miR-223 selectively enhance cardiomyogenesis both in the PE/ST explants as well as in the embryonic epicardium, a Smurf1- and Foxp1-driven process. In addition we identified three novel long non-coding RNAs with enhanced expression in the PE/ST, that are complementary regulated by Bmp and Fgf administration and well as by microRNAs that selectively promote cardiomyogenesis, supporting a pivotal role of these long non coding RNAs in microRNA-mediated cardiomyogenesis of the PE/ST cells.


Assuntos
Diferenciação Celular , Fatores de Transcrição Forkhead/metabolismo , MicroRNAs/genética , Miócitos Cardíacos/citologia , Pericárdio/embriologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem da Célula , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Pericárdio/citologia
8.
Cell Rep ; 31(10): 107739, 2020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32521264

RESUMO

Epicardial cells are cardiac progenitors that give rise to the majority of cardiac fibroblasts, coronary smooth muscle cells, and pericytes during development. An integral phase of epicardial fate transition is epithelial-to-mesenchymal transition (EMT) that confers motility. We uncover an essential role for the protein arginine methyltransferase 1 (PRMT1) in epicardial invasion and differentiation. Using scRNA-seq, we show that epicardial-specific deletion of Prmt1 reduced matrix and ribosomal gene expression in epicardial-derived cell lineages. PRMT1 regulates splicing of Mdm4, which is a key controller of p53 stability. Loss of PRMT1 leads to accumulation of p53 that enhances Slug degradation and blocks EMT. During heart development, the PRMT1-p53 pathway is required for epicardial invasion and formation of epicardial-derived lineages: cardiac fibroblasts, coronary smooth muscle cells, and pericytes. Consequently, this pathway modulates ventricular morphogenesis and coronary vessel formation. Altogether, our study reveals molecular mechanisms involving the PRMT1-p53 pathway and establish its roles in heart development.


Assuntos
Pericárdio/embriologia , Pericárdio/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Diferenciação Celular/fisiologia , Transição Epitelial-Mesenquimal , Feminino , Coração/embriologia , Camundongos , Miocárdio/citologia , Miocárdio/metabolismo , Pericárdio/citologia , Gravidez , Transdução de Sinais
9.
Stem Cell Reports ; 14(5): 770-787, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32359445

RESUMO

The embryonic epicardium, originating from the proepicardial organ (PEO), provides a source of multipotent progenitors for cardiac lineages, including pericytes, fibroblasts, and vascular smooth muscle cells. Maximizing the regenerative capacity of the adult epicardium depends on recapitulating embryonic cell fates. The potential of the epicardium to contribute coronary endothelium is unclear, due to conflicting Cre-based lineage trace data. Controversy also surrounds when epicardial cell fate becomes restricted. Here, we systematically investigate expression of five widely used epicardial markers, Wt1, Tcf21, Tbx18, Sema3d, and Scx, over the course of development. We show overlap of markers in all PEO and epicardial cells until E13.5, and find no evidence for discrete proepicardial sub-compartments that might contribute coronary endothelium via the epicardial layer. Our findings clarify a number of prevailing discrepancies and support the notion that epicardium-derived cell fate, to form fibroblasts or mural cells, is specified after epithelial-mesenchymal transition, not pre-determined within the PEO.


Assuntos
Linhagem da Célula , Células-Tronco Embrionárias Murinas/metabolismo , Pericárdio/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas/citologia , Células Musculares/citologia , Células Musculares/metabolismo , Pericárdio/embriologia , Pericárdio/metabolismo , Pericitos/citologia , Pericitos/metabolismo , Semaforinas/genética , Semaforinas/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Proteínas WT1/genética , Proteínas WT1/metabolismo
10.
Dev Cell ; 52(5): 574-590.e6, 2020 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-32084358

RESUMO

The epicardium is essential during cardiac development, homeostasis, and repair, and yet fundamental insights into its underlying cell biology, notably epicardium formation, lineage heterogeneity, and functional cross-talk with other cell types in the heart, are currently lacking. In this study, we investigated epicardial heterogeneity and the functional diversity of discrete epicardial subpopulations in the developing zebrafish heart. Single-cell RNA sequencing uncovered three epicardial subpopulations with specific genetic programs and distinctive spatial distribution. Perturbation of unique gene signatures uncovered specific functions associated with each subpopulation and established epicardial roles in cell adhesion, migration, and chemotaxis as a mechanism for recruitment of leukocytes into the heart. Understanding which mechanisms epicardial cells employ to establish a functional epicardium and how they communicate with other cardiovascular cell types during development will bring us closer to repairing cellular relationships that are disrupted during cardiovascular disease.


Assuntos
Linhagem da Célula , Pericárdio/citologia , Transcriptoma , Animais , Regulação da Expressão Gênica no Desenvolvimento , Pericárdio/embriologia , Pericárdio/metabolismo , RNA-Seq , Análise de Célula Única , Peixe-Zebra
11.
FASEB J ; 34(4): 5223-5239, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32068311

RESUMO

The embryonic epicardium generates a population of epicardial-derived mesenchymal cells (EPDC) whose contribution to the coronary endothelium is minor or, according to some reports, negligible. We have compared four murine cell-tracing models related to the EPDC in order to elucidate this contribution. Cre recombinase was expressed under control of the promoters of the Wilms' tumor suppressor (Wt1), the cardiac troponin (cTnT), and the GATA5 genes, activating expression of the R26REYFP reporter. We have also used the G2 enhancer of the GATA4 gene as a driver due to its activation in the proepicardium. Recombination was found in most of the epicardium/EPDC in all cases. The contribution of these lineages to the cardiac endothelium was analyzed using confocal microscopy and flow cytometry. G2-GATA4 lineage cells are the most frequent in the endothelium, probably due to the recruitment of circulating endothelial progenitors. The contribution of the WT1 cell lineage increases along gestation due to further endothelial expression of WT1. GATA5 and cTnT lineages represent 4% of the cardiac endothelial cells throughout the gestation, probably standing for the actual EPDC contribution to the coronary endothelium. These results suggest caution when using a sole cell-tracing model to study the fate of the EPDC.


Assuntos
Linhagem da Célula , Vasos Coronários/citologia , Endotélio Vascular/citologia , Pericárdio/citologia , Animais , Vasos Coronários/embriologia , Vasos Coronários/metabolismo , Endotélio Vascular/embriologia , Endotélio Vascular/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Integrases , Camundongos , Pericárdio/embriologia , Pericárdio/metabolismo
12.
Artigo em Inglês | MEDLINE | ID: mdl-31451510

RESUMO

The epicardium, the outermost tissue layer that envelops all vertebrate hearts, plays a crucial role in cardiac development and regeneration and has been implicated in potential strategies for cardiac repair. The heterogenous cell population that composes the epicardium originates primarily from a transient embryonic cell cluster known as the proepicardial organ (PE). Characterized by its high cellular plasticity, the epicardium contributes to both heart development and regeneration in two critical ways: as a source of progenitor cells and as a critical signaling hub. Despite this knowledge, there are many unanswered questions in the field of epicardial biology, the resolution of which will advance the understanding of cardiac development and repair. We review current knowledge in cross-species epicardial involvement, specifically in relation to lineage specification and differentiation during cardiac development.


Assuntos
Pericárdio/embriologia , Animais , Diferenciação Celular , Pericárdio/citologia , Pericárdio/metabolismo , Regeneração , Células-Tronco/fisiologia
13.
Artigo em Inglês | MEDLINE | ID: mdl-31767650

RESUMO

Heart development is a complex process and begins with the long-range migration of cardiac progenitor cells during gastrulation. This culminates in the formation of a simple contractile tube with multiple layers, which undergoes remodeling into a four-chambered heart. During this morphogenesis, additional cell populations become incorporated. It is important to unravel the underlying genetic and cellular mechanisms to be able to identify the embryonic origin of diseases, including congenital malformations, which impair cardiac function and may affect life expectancy or quality. Owing to the evolutionary conservation of development, observations made in nonamniote and amniote vertebrate species allow us to extrapolate to human. This review will focus on the contributions made to a better understanding of heart development through studying avian embryos-mainly the chicken but also quail embryos. We will illustrate the classic and recent approaches used in the avian system, give an overview of the important discoveries made, and summarize the early stages of cardiac development up to the establishment of the four-chambered heart.


Assuntos
Embrião de Galinha , Galinhas/fisiologia , Coração/embriologia , Modelos Animais , Codorniz/embriologia , Codorniz/fisiologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem da Célula , Movimento Celular , Células Cultivadas , Corantes Fluorescentes , Cardiopatias Congênitas/embriologia , Ventrículos do Coração/embriologia , Humanos , Morfogênese , Crista Neural/embriologia , Organogênese , Pericárdio/embriologia , Transgenes
14.
Development ; 146(13)2019 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-31175121

RESUMO

The epicardium, the outer mesothelial layer enclosing the myocardium, plays key roles in heart development and regeneration. During embryogenesis, the epicardium arises from the proepicardium (PE), a cell cluster that appears in the dorsal pericardium (DP) close to the venous pole of the heart. Little is known about how the PE emerges from the pericardial mesothelium. Using a zebrafish model and a combination of genetic tools, pharmacological agents and quantitative in vivo imaging, we reveal that a coordinated collective movement of DP cells drives PE formation. We found that Bmp signaling and the actomyosin cytoskeleton promote constriction of the DP, which enables PE cells to extrude apically. We provide evidence that cell extrusion, which has been described in the elimination of unfit cells from epithelia and the emergence of hematopoietic stem cells, is also a mechanism for PE cells to exit an organized mesothelium and fulfil their developmental fate to form a new tissue layer, the epicardium.


Assuntos
Actinas/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Movimento Celular , Coração/embriologia , Pericárdio/citologia , Pericárdio/embriologia , Células-Tronco/fisiologia , Animais , Animais Geneticamente Modificados , Padronização Corporal/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Movimento Celular/genética , Embrião não Mamífero , Miocárdio/citologia , Organogênese/genética , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
15.
Comp Med ; 69(3): 184-203, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31182184

RESUMO

Cardiovascular disease remains one of the top contributors to morbidity and mortality in the United States. Increasing evidence suggests that many processes, pathways, and programs observed during development and organogenesis are recapitulated in adults in the face of disease. Therefore, a heightened understanding of cardiac development and organogenesis will help increase our understanding of developmental defects and cardiovascular diseases in adults. Chicks have long served as a model system in which to study developmental problems. Detailed descriptions of morphogenesis, low cost, accessibility, ease of manipulation, and the optimization of genetic engineering techniques have made chicks a robust model for studying development and make it a powerful platform for cardiovascular research. This review summarizes the cardiac developmental milestones of embryonic chickens, practical considerations when working with chicken embryos, and techniques available for use in chicks (including tissue chimeras, genetic manipulations, and live imaging). In addition, this article highlights examples that accentuate the utility of the embryonic chicken as model system in which to study cardiac development, particularly epicardial development, and that underscore the importance of how studying development informs our understanding of disease.


Assuntos
Embrião de Galinha/embriologia , Embrião de Galinha/fisiologia , Coração/embriologia , Coração/fisiologia , Criação de Animais Domésticos , Animais , Doenças Cardiovasculares/etiologia , Galinhas/genética , Galinhas/fisiologia , Técnicas Genéticas , Humanos , Modelos Animais , Modelos Cardiovasculares , Organogênese , Pericárdio/embriologia , Pesquisa Translacional Biomédica
16.
J Cell Mol Med ; 23(8): 5553-5565, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31211496

RESUMO

Women with pre-gestational diabetes have a higher risk of producing children with congenital heart defects (CHDs), caused predominantly by hyperglycemia-induced oxidative stress. In this study, we evaluated if exercise during pregnancy could mitigate oxidative stress and reduce the incidence of CHDs in the offspring of diabetic mice. Female mice were treated with streptozotocin to induce pre-gestational diabetes, then mated with healthy males to produce offspring. They were also given access to running wheels 1 week before mating and allowed to exercise voluntarily until E18.5. Heart morphology, gene expression, and oxidative stress were assessed in foetal hearts. Maternal voluntary exercise results in a significantly lower incidence of CHDs from 59.5% to 25%. Additionally, diabetes-induced defects in coronary artery and capillary morphogenesis were also lower with exercise. Myocardial cell proliferation and epithelial-mesenchymal transition at E12.5 was significantly lower with pre-gestational diabetes which was mitigated with maternal exercise. Cardiac gene expression of Notch1, Snail1, Gata4 and Cyclin D1 was significantly higher in the embryos of diabetic mice that exercised compared to the non-exercised group. Furthermore, maternal exercise produced lower reactive oxygen species (ROS) and oxidative stress in the foetal heart. In conclusion, maternal exercise mitigates ROS and oxidative damage in the foetal heart, and results in a lower incidence of CHDs in the offspring of pre-gestational diabetes. Exercise may be an effective intervention to compliment clinical management and further minimize CHD risk in mothers with diabetes.


Assuntos
Diabetes Mellitus Experimental/complicações , Diabetes Gestacional/patologia , Cardiopatias Congênitas/etiologia , Estresse Oxidativo , Condicionamento Físico Animal , Animais , Glicemia/metabolismo , Capilares/anormalidades , Proliferação de Células , Anomalias dos Vasos Coronários/patologia , Embrião de Mamíferos/patologia , Transição Epitelial-Mesenquimal , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Tamanho da Ninhada de Vivíparos , Masculino , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo III/metabolismo , Pericárdio/embriologia , Pericárdio/patologia , Fosforilação , Gravidez
17.
Genesis ; 57(7-8): e23303, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31066193

RESUMO

The vitamin A metabolite, retinoic acid, carries out essential and conserved roles in vertebrate heart development. Retinoic acid signals via retinoic acid receptors (RAR)/retinoid X receptors (RXRs) heterodimers to induce the expression of genes that control cell fate specification, proliferation, and differentiation. Alterations in retinoic acid levels are often associated with congenital heart defects. Therefore, embryonic levels of retinoic acid need to be carefully regulated through the activity of enzymes, binding proteins and transporters involved in vitamin A metabolism. Here, we review evidence of the complex mechanisms that control the fetal uptake and synthesis of retinoic acid from vitamin A precursors. Next, we highlight recent evidence of the role of retinoic acid in orchestrating myocardial compact zone growth and coronary vascular development.


Assuntos
Pericárdio/embriologia , Transdução de Sinais , Tretinoína/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Pericárdio/metabolismo , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo
18.
J Anat ; 234(6): 800-814, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30882904

RESUMO

During embryo development, the heart is the first functioning organ. Although quiescent in the adult, the epicardium is essential during development to form a normal four-chambered heart. Epicardial-derived cells contribute to the heart as it develops with fibroblasts and vascular smooth muscle cells. Previous studies have shown that a heartbeat is required for epicardium formation, but no study to our knowledge has shown the effects of haemodynamic changes on the epicardium. Since the aetiologies of many congenital heart defects are unknown, we suggest that an alteration in the heart's haemodynamics might provide an explanatory basis for some of them. To change the heart's haemodynamics, outflow tract (OFT) banding using a double overhang knot was performed on HH21 chick embryos, with harvesting at different developmental stages. The epicardium of the heart was phenotypically and functionally characterised using a range of techniques. Upon alteration of haemodynamics, the epicardium exhibited abnormal morphology at HH29, even though migration of epicardial cells along the surface of the heart was found to be normal between HH24 and HH28. The abnormal epicardial phenotype was exacerbated at HH35 with severe changes in the structure of the extracellular matrix (ECM). A number of genes tied to ECM production were also differentially expressed in HH29 OFT-banded hearts, including DDR2 and collagen XII. At HH35, the differential expression of these genes was even greater with additional downregulation of collagen I and TCF21. In this study, the epicardium was found to be severely impacted by altered haemodynamics upon OFT banding. The increased volume of the epicardium at HH29, upon OFT-banding, and the expression changes of ECM markers were the first indicative signs of aberrations in epicardial architecture; by HH35, the phenotype had progressed. The decrease in epicardial thickness at HH35 suggests an increase in tension, with a force acting perpendicular to the surface of the epicardium. Although the developing epicardium and the blood flowing through the heart are separated by the endocardium and myocardium, the data presented here demonstrate that altering the blood flow affects the structure and molecular expression of the epicardial layer. Due to the intrinsic role the epicardium in cardiogenesis, defects in epicardial formation could have a role in the formation of a wide range of congenital heart defects.


Assuntos
Hemodinâmica/fisiologia , Pericárdio/embriologia , Animais , Embrião de Galinha , Organogênese/fisiologia
19.
Acta Biochim Biophys Sin (Shanghai) ; 51(4): 402-410, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30877755

RESUMO

Epicardial progenitor cells (EpiCs) which are derived from the proepicardium have the potential to differentiate into coronary vascular smooth muscle cells during development. Whether sphingosine 1-phosphate (S1P), a highly hydrophobic zwitterionic lysophospholipid in signal transduction, induces the differentiation of EpiCs is unknown. In the present study, we demonstrated that S1P significantly induced the expression of smooth muscle cell specific markers α-smooth muscle actin and myosin heavy chain 11 in the EpiCs. And the smooth muscle cells differentiated from the EpiCs stimulated by S1P were further evaluated by gel contraction assay. To further confirm the major subtype of sphingosine 1-phosphate receptors (S1PRs) involved in the differentiation of EpiCs, we used the agonists and antagonists of different S1PRs. The results showed that the S1P1/S1P3 antagonist VPC23019 and the S1P2 antagonist JTE013 significantly attenuated EpiCs differentiation, while the S1P1 agonist SEW2871 and antagonist W146 did not affect EpiCs differentiation. These results collectively suggested that S1P, principally through its receptor S1P3, increases EpiCs differentiation into VSMCs and thus indicated the importance of S1P signaling in the embryonic coronary vasculature, while S1P2 plays a secondary role.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Células-Tronco Embrionárias Murinas/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Pericárdio/citologia , Esfingosina/análogos & derivados , Actinas/genética , Actinas/metabolismo , Animais , Diferenciação Celular/genética , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Pericárdio/embriologia , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Pirazóis/farmacologia , Piridinas/farmacologia , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Esfingosina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...