Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Cell Cardiol ; 195: 1-13, 2024 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-39038734

RESUMO

Revascularization of ischemic myocardium following cardiac damage is an important step in cardiac regeneration. However, the mechanism of arteriogenesis has not been well described during cardiac regeneration. Here we investigated coronary artery remodeling and collateral growth during cardiac regeneration. Neonatal MI was induced by ligature of the left descending artery (LAD) in postnatal day (P) 1 or P7 pups from the Cx40-GFP mouse line and the arterial tree was reconstructed in 3D from images of cleared hearts collected at 1, 2, 4, 7 and 14 days after infarction. We show a rapid remodeling of the left coronary arterial tree induced by neonatal MI and the formation of numerous collateral arteries, which are transient in regenerating hearts after MI at P1 and persistent in non-regenerating hearts after MI at P7. This difference is accompanied by restoration of a perfused or a non-perfused LAD following MI at P1 or P7 respectively. Interestingly, collaterals ameliorate cardiac perfusion and drive LAD repair, and lineage tracing analysis demonstrates that the restoration of the LAD occurs by remodeling of pre-existing arterial cells independently of whether they originate in large arteries or arterioles. These results demonstrate that the restoration of the LAD artery during cardiac regeneration occurs by pruning as the rapidly forming collaterals that support perfusion of the disconnected lower LAD subsequently disappear on restoration of a unique LAD. These results highlight a rapid phase of arterial remodeling that plays an important role in vascular repair during cardiac regeneration.

2.
Adv Exp Med Biol ; 1441: 103-124, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38884707

RESUMO

The heart forms from the first and second heart fields, which contribute to distinct regions of the myocardium. This is supported by clonal analyses, which identify corresponding first and second cardiac cell lineages in the heart. Progenitor cells of the second heart field and its sub-domains are controlled by a gene regulatory network and signaling pathways, which determine their behavior. Multipotent cells in this field can also contribute cardiac endothelial and smooth muscle cells. Furthermore, the skeletal muscles of the head and neck are clonally related to myocardial cells that form the arterial and venous poles of the heart. These lineage relationships, together with the genes that regulate the heart fields, have major implications for congenital heart disease.


Assuntos
Linhagem da Célula , Animais , Humanos , Diferenciação Celular/genética , Linhagem da Célula/genética , Coração/fisiologia , Miocárdio/citologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Transdução de Sinais , Células-Tronco/metabolismo , Células-Tronco/citologia , Células-Tronco/fisiologia
3.
Adv Exp Med Biol ; 1441: 77-85, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38884705

RESUMO

The major events of cardiac development, including early heart formation, chamber morphogenesis and septation, and conduction system and coronary artery development, are briefly reviewed together with a short introduction to the animal species commonly used to study heart development and model congenital heart defects (CHDs).


Assuntos
Modelos Animais de Doenças , Cardiopatias Congênitas , Coração , Animais , Cardiopatias Congênitas/fisiopatologia , Cardiopatias Congênitas/patologia , Coração/embriologia , Coração/fisiopatologia , Coração/crescimento & desenvolvimento , Humanos , Camundongos , Morfogênese
4.
Adv Exp Med Biol ; 1441: 645-659, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38884739

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Dupla Via de Saída do Ventrículo Direito , Transdução de Sinais , Tetralogia de Fallot , Tetralogia de Fallot/genética , Tetralogia de Fallot/patologia , Tetralogia de Fallot/fisiopatologia , Tetralogia de Fallot/embriologia , Animais , Dupla Via de Saída do Ventrículo Direito/genética , Dupla Via de Saída do Ventrículo Direito/patologia , Dupla Via de Saída do Ventrículo Direito/fisiopatologia , Humanos , Crista Neural/metabolismo , Crista Neural/patologia , Crista Neural/embriologia , Morfogênese/genética
5.
C R Biol ; 347: 9-18, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38488639

RESUMO

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.


Les malformations cardiaques congénitales touchent 1 naissance sur 100 et résultent d'anomalies du développement cardiaque. La croissance du tube cardiaque précoce se produit par l'ajout progressif de cellules progénitrices du second champ cardiaque (SHF) aux pôles cardiaques. Le SHF contribue au myocarde septal ventriculaire, au myocarde ventriculaire droit et au myocarde de la voie de sortie au pôle artériel, et au myocarde auriculaire, y compris le myocarde septal auriculaire, au pôle veineux. Le déploiement du SHF est essentiel pour la septation cardiaque et a été impliqué dans la formation du boucle cardiaque et, avec les cellules de la crête neurale, dans l'orchestration du développement de la voie efférente. Perturbation génétique ou environnementale du déploiement du SHF est donc à l'origine d'un spectre de formes communes de maladies cardiaques congénitales affectant la morphogenèse conotroncale et septale. Ici, nous passons en revue les principales propriétés des cellules du SHF ainsi que les découvertes récentes sur les programmes de développement qui contrôlent l'ajout de cellules progénitrices cardiaques ainsi que les origines des malformations cardiaques congénitales.


Assuntos
Cardiopatias Congênitas , Coração , Humanos , Cardiopatias Congênitas/genética , Miocárdio , Células-Tronco , Morfogênese
6.
Differentiation ; : 100741, 2023 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-38040515

RESUMO

Fibroblast growth factor 10 (FGF10) is a major morphoregulatory factor that plays essential signaling roles during vertebrate multiorgan development and homeostasis. FGF10 is predominantly expressed in mesenchymal cells and signals though FGFR2b in adjacent epithelia to regulate branching morphogenesis, stem cell fate, tissue differentiation and proliferation, in addition to autocrine roles. Genetic loss of function analyses have revealed critical requirements for FGF10 signaling during limb, lung, digestive system, ectodermal, nervous system, craniofacial and cardiac development. Heterozygous FGF10 mutations have been identified in human genetic syndromes associated with craniofacial anomalies, including lacrimal and salivary gland aplasia. Elevated Fgf10 expression is associated with poor prognosis in a range of cancers. In addition to developmental and disease roles, FGF10 regulates homeostasis and repair of diverse adult tissues and has been identified as a target for regenerative medicine.

8.
J Cardiovasc Dev Dis ; 10(5)2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-37233161

RESUMO

The ventricular conduction or His-Purkinje system (VCS) mediates the rapid propagation and precise delivery of electrical activity essential for the synchronization of heartbeats. Mutations in the transcription factor Nkx2-5 have been implicated in a high prevalence of developing ventricular conduction defects or arrhythmias with age. Nkx2-5 heterozygous mutant mice reproduce human phenotypes associated with a hypoplastic His-Purkinje system resulting from defective patterning of the Purkinje fiber network during development. Here, we investigated the role of Nkx2-5 in the mature VCS and the consequences of its loss on cardiac function. Neonatal deletion of Nkx2-5 in the VCS using a Cx40-CreERT2 mouse line provoked apical hypoplasia and maturation defects of the Purkinje fiber network. Genetic tracing analysis demonstrated that neonatal Cx40-positive cells fail to maintain a conductive phenotype after Nkx2-5 deletion. Moreover, we observed a progressive loss of expression of fast-conduction markers in persistent Purkinje fibers. Consequently, Nkx2-5-deleted mice developed conduction defects with progressively reduced QRS amplitude and RSR' complex associated with higher duration. Cardiac function recorded by MRI revealed a reduction in the ejection fraction in the absence of morphological changes. With age, these mice develop a ventricular diastolic dysfunction associated with dyssynchrony and wall-motion abnormalities without indication of fibrosis. These results highlight the requirement of postnatal expression of Nkx2-5 in the maturation and maintenance of a functional Purkinje fiber network to preserve contraction synchrony and cardiac function.

9.
Cardiovasc Res ; 119(11): 2089-2105, 2023 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-37052590

RESUMO

AIMS: Haploinsufficiency of the chromo-domain protein CHD7 underlies most cases of CHARGE syndrome, a multisystem birth defect including congenital heart malformation. Context specific roles for CHD7 in various stem, progenitor, and differentiated cell lineages have been reported. Previously, we showed severe defects when Chd7 is absent from cardiopharyngeal mesoderm (CPM). Here, we investigate altered gene expression in the CPM and identify specific CHD7-bound target genes with known roles in the morphogenesis of affected structures. METHODS AND RESULTS: We generated conditional KO of Chd7 in CPM and analysed cardiac progenitor cells using transcriptomic and epigenomic analyses, in vivo expression analysis, and bioinformatic comparisons with existing datasets. We show CHD7 is required for correct expression of several genes established as major players in cardiac development, especially within the second heart field (SHF). We identified CHD7 binding sites in cardiac progenitor cells and found strong association with histone marks suggestive of dynamically regulated enhancers during the mesodermal to cardiac progenitor transition of mESC differentiation. Moreover, CHD7 shares a subset of its target sites with ISL1, a pioneer transcription factor in the cardiogenic gene regulatory network, including one enhancer modulating Fgf10 expression in SHF progenitor cells vs. differentiating cardiomyocytes. CONCLUSION: We show that CHD7 interacts with ISL1, binds ISL1-regulated cardiac enhancers, and modulates gene expression across the mesodermal heart fields during cardiac morphogenesis.


Assuntos
Síndrome CHARGE , Proteínas de Ligação a DNA , Humanos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Síndrome CHARGE/genética , Síndrome CHARGE/metabolismo , Elementos Facilitadores Genéticos , Coração , Miócitos Cardíacos/metabolismo , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , DNA Helicases/genética , DNA Helicases/metabolismo
10.
Dev Biol ; 499: 10-21, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37060937

RESUMO

Development of the outflow tract of the heart requires specification, proliferation and deployment of a progenitor cell population from the second heart field to generate the myocardium at the arterial pole of the heart. Disruption of these processes leads to lethal defects in rotation and septation of the outflow tract. We previously showed that Fibroblast Growth Factor 8 (FGF8) directs a signaling cascade in the second heart field that regulates critical aspects of OFT morphogenesis. Here we show that in addition to the survival and proliferation cues previously described, FGF8 provides instructive and patterning information to OFT myocardial cells and their progenitors that prevents their aberrant differentiation along a working myocardial program.


Assuntos
Coração , Miocárdio , Diferenciação Celular/fisiologia , Fator 8 de Crescimento de Fibroblasto/genética , Fator 8 de Crescimento de Fibroblasto/metabolismo , Mesoderma/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos , Animais , Camundongos
11.
Dev Cell ; 58(4): 257-266, 2023 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-36809764

RESUMO

Organogenesis requires the orchestrated development of multiple cell lineages that converge, interact, and specialize to generate coherent functional structures, exemplified by transformation of the cardiac crescent into a four-chambered heart. Cardiomyocytes originate from the first and second heart fields, which make different regional contributions to the definitive heart. In this review, a series of recent single-cell transcriptomic analyses, together with genetic tracing experiments, are discussed, providing a detailed panorama of the cardiac progenitor cell landscape. These studies reveal that first heart field cells originate in a juxtacardiac field adjacent to extraembryonic mesoderm and contribute to the ventrolateral side of the cardiac primordium. In contrast, second heart field cells are deployed dorsomedially from a multilineage-primed progenitor population via arterial and venous pole pathways. Refining our knowledge of the origin and developmental trajectories of cells that build the heart is essential to address outstanding challenges in cardiac biology and disease.


Assuntos
Coração , Miócitos Cardíacos , Miócitos Cardíacos/metabolismo , Linhagem da Célula/genética , Mesoderma/metabolismo , Diferenciação Celular/genética
12.
Circ Res ; 131(10): 842-858, 2022 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-36205127

RESUMO

BACKGROUND: The arterial pole of the heart is a hotspot for life-threatening forms of congenital heart defects (CHDs). Development of this cardiac region occurs by addition of Second Heart Field (SHF) progenitor cells to the embryonic outflow tract (OFT) and subsequently the base of the ascending aorta and pulmonary trunk. Understanding the cellular and genetic mechanisms driving arterial pole morphogenesis is essential to provide further insights into the cause of CHDs. METHODS: A synergistic combination of bioinformatic analysis and mouse genetics as well as embryo and explant culture experiments were used to dissect the cross-regulatory transcriptional circuitry operating in future subaortic and subpulmonary OFT myocardium. RESULTS: Here, we show that the lipid sensor PPARγ (peroxisome proliferator-activated receptor gamma) is expressed in future subpulmonary myocardium in the inferior wall of the OFT and that PPARγ signaling-related genes display regionalized OFT expression regulated by the transcription factor TBX1 (T-box transcription factor 1). Modulating PPARγ activity in ex vivo cultured embryos treated with a PPARγ agonist or antagonist or deleting Pparγ in cardiac progenitor cells using Mesp1-Cre reveals that Pparγ is required for addition of future subpulmonary myocardium and normal arterial pole development. Additionally, the non-canonical DLK1 (delta-like noncanonical Notch ligand 1)/NOTCH (Notch receptor 1)/HES1 (Hes family bHLH transcription factor 1) pathway negatively regulates Pparγ in future subaortic myocardium in the superior OFT wall. CONCLUSIONS: Together these results identify Pparγ as a regulator of regional transcriptional identity in the developing heart, providing new insights into gene interactions involved in congenital heart defects.


Assuntos
Cardiopatias Congênitas , PPAR gama , Animais , Camundongos , Coração , Cardiopatias Congênitas/genética , Miocárdio/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Fatores de Transcrição/metabolismo , Receptores Notch/metabolismo
13.
Dis Model Mech ; 15(9)2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35946435

RESUMO

TBX1 is a key regulator of pharyngeal apparatus (PhAp) development. Vitamin B12 (vB12) treatment partially rescues aortic arch patterning defects of Tbx1+/- embryos. Here, we show that it also improves cardiac outflow tract septation and branchiomeric muscle anomalies of Tbx1 hypomorphic mutants. At the molecular level, in vivo vB12 treatment enabled us to identify genes that were dysregulated by Tbx1 haploinsufficiency and rescued by treatment. We found that SNAI2, also known as SLUG, encoded by the rescued gene Snai2, identified a population of mesodermal cells that was partially overlapping with, but distinct from, ISL1+ and TBX1+ populations. In addition, SNAI2+ cells were mislocalized and had a greater tendency to aggregate in Tbx1+/- and Tbx1-/- embryos, and vB12 treatment restored cellular distribution. Adjacent neural crest-derived mesenchymal cells, which do not express TBX1, were also affected, showing enhanced segregation from cardiopharyngeal mesodermal cells. We propose that TBX1 regulates cell distribution in the core mesoderm and the arrangement of multiple lineages within the PhAp.


Assuntos
Síndrome de DiGeorge , Animais , Síndrome de DiGeorge/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/metabolismo , Camundongos , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Vitamina B 12
14.
Methods Mol Biol ; 2438: 231-250, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35147946

RESUMO

Epithelial cardiac progenitor cells of the second heart field (SHF) contribute to growth of the vertebrate heart tube by progressive addition of cells from the dorsal pericardial wall to the cardiac poles. Perturbation of SHF development, including defects in apicobasal or planar polarity, results in shortening of the heart tube and a spectrum of congenital heart defects. Here, we provide detailed protocols for fixed section and wholemount immunofluorescence and live imaging approaches to studying the epithelial properties of cardiac progenitors in the dorsal pericardial wall during mouse heart development. Whole-embryo culture and electroporation methods are also presented, allowing for pharmacological and genetic perturbation of SHF development, as well as image analysis approaches to quantify cell features across the progenitor cell epithelium. These protocols are broadly applicable to the study of epithelia in the early embryo.


Assuntos
Embrião de Mamíferos , Coração , Animais , Epitélio , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Organogênese , Pericárdio , Células-Tronco
15.
Exp Cell Res ; 410(1): 112931, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34798131

RESUMO

Branchiomeric muscles of the head and neck originate in a population of cranial mesoderm termed cardiopharyngeal mesoderm that also contains progenitor cells contributing to growth of the embryonic heart. Retrospective lineage analysis has shown that branchiomeric muscles share a clonal origin with parts of the heart, indicating the presence of common heart and head muscle progenitor cells in the early embryo. Genetic lineage tracing and functional studies in the mouse, as well as in Ciona and zebrafish, together with recent experiments using single cell transcriptomics and multipotent stem cells, have provided further support for the existence of bipotent head and heart muscle progenitor cells. Current challenges concern defining where and when such common progenitor cells exist in mammalian embryos and how alternative myogenic derivatives emerge in cardiopharyngeal mesoderm. Addressing these questions will provide insights into mechanisms of cell fate acquisition and the evolution of vertebrate musculature, as well as clinical insights into the origins of muscle restricted myopathies and congenital defects affecting craniofacial and cardiac development.


Assuntos
Desenvolvimento Embrionário/genética , Coração/crescimento & desenvolvimento , Mesoderma/crescimento & desenvolvimento , Desenvolvimento Muscular/genética , Animais , Diferenciação Celular/genética , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Cabeça/crescimento & desenvolvimento , Camundongos , Músculo Esquelético/crescimento & desenvolvimento , Células-Tronco/citologia , Peixe-Zebra/genética
16.
Cardiovasc Res ; 118(12): 2625-2637, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34755840

RESUMO

AIMS: Promoting cardiomyocyte renewal represents a major therapeutic approach for heart regeneration and repair. Our study aims to investigate the relevance of FGF10 as a potential target for heart regeneration. METHODS AND RESULTS: Our results first reveal that Fgf10 levels are up-regulated in the injured ventricle after MI. Adult mice with reduced Fgf10 expression subjected to MI display impaired cardiomyocyte proliferation and enhanced cardiac fibrosis, leading to a worsened cardiac function and remodelling post-MI. In contrast, conditional Fgf10 overexpression post-MI revealed that, by enhancing cardiomyocyte proliferation and preventing scar-promoting myofibroblast activation, FGF10 preserves cardiac remodelling and function. Moreover, FGF10 activates major regenerative pathways including the regulation of Meis1 expression levels, the Hippo signalling pathway and a pro-glycolytic metabolic switch. Finally, we demonstrate that elevated FGF10 levels in failing human hearts correlate with reduced fibrosis and enhanced cardiomyocyte proliferation. CONCLUSIONS: Altogether, our study shows that FGF10 promotes cardiac regeneration and repair through two cellular mechanisms: elevating cardiomyocyte renewal and limiting fibrosis. This study thus identifies FGF10 as a clinically relevant target for heart regeneration and repair in man.


Assuntos
Infarto do Miocárdio , Miócitos Cardíacos , Animais , Proliferação de Células , Células Cultivadas , Fator 10 de Crescimento de Fibroblastos/metabolismo , Fibrose , Humanos , Camundongos , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Regeneração
17.
Nat Commun ; 12(1): 6645, 2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34789765

RESUMO

The poles of the heart and branchiomeric muscles of the face and neck are formed from the cardiopharyngeal mesoderm within the pharyngeal apparatus. They are disrupted in patients with 22q11.2 deletion syndrome, due to haploinsufficiency of TBX1, encoding a T-box transcription factor. Here, using single cell RNA-sequencing, we now identify a multilineage primed population within the cardiopharyngeal mesoderm, marked by Tbx1, which has bipotent properties to form cardiac and branchiomeric muscle cells. The multilineage primed cells are localized within the nascent mesoderm of the caudal lateral pharyngeal apparatus and provide a continuous source of cardiopharyngeal mesoderm progenitors. Tbx1 regulates the maturation of multilineage primed progenitor cells to cardiopharyngeal mesoderm derivatives while restricting ectopic non-mesodermal gene expression. We further show that TBX1 confers this balance of gene expression by direct and indirect regulation of enriched genes in multilineage primed progenitors and downstream pathways, partly through altering chromatin accessibility, the perturbation of which can lead to congenital defects in individuals with 22q11.2 deletion syndrome.


Assuntos
Região Branquial/citologia , Mesoderma/citologia , Miocárdio/citologia , Proteínas com Domínio T/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Região Branquial/embriologia , Região Branquial/metabolismo , Diferenciação Celular , Linhagem da Célula , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Coração/embriologia , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Análise de Célula Única , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteínas com Domínio T/genética
18.
J Cardiovasc Dev Dis ; 8(8)2021 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-34436237

RESUMO

The rapid propagation of electrical activity through the ventricular conduction system (VCS) controls spatiotemporal contraction of the ventricles. Cardiac conduction defects or arrhythmias in humans are often associated with mutations in key cardiac transcription factors that have been shown to play important roles in VCS morphogenesis in mice. Understanding of the mechanisms of VCS development is thus crucial to decipher the etiology of conduction disturbances in adults. During embryogenesis, the VCS, consisting of the His bundle, bundle branches, and the distal Purkinje network, originates from two independent progenitor populations in the primary ring and the ventricular trabeculae. Differentiation into fast-conducting cardiomyocytes occurs progressively as ventricles develop to form a unique electrical pathway at late fetal stages. The objectives of this review are to highlight the structure-function relationship between VCS morphogenesis and conduction defects and to discuss recent data on the origin and development of the VCS with a focus on the distal Purkinje fiber network.

19.
Circ Res ; 128(3): 360-362, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33539223
20.
Cell Stem Cell ; 28(2): 230-240.e6, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33176168

RESUMO

Organoids are powerful models for studying tissue development, physiology, and disease. However, current culture systems disrupt the inductive tissue-tissue interactions needed for the complex morphogenetic processes of native organogenesis. Here, we show that mouse embryonic stem cells (mESCs) can be coaxed to robustly undergo fundamental steps of early heart organogenesis with an in-vivo-like spatiotemporal fidelity. These axially patterned embryonic organoids (gastruloids) mimic embryonic development and support the generation of cardiovascular progenitors, including first and second heart fields. The cardiac progenitors self-organize into an anterior domain reminiscent of a cardiac crescent before forming a beating cardiac tissue near a putative primitive gut-like tube, from which it is separated by an endocardial-like layer. These findings unveil the surprising morphogenetic potential of mESCs to execute key aspects of organogenesis through the coordinated development of multiple tissues. This platform could be an excellent tool for studying heart development in unprecedented detail and throughput.


Assuntos
Organogênese , Organoides , Animais , Desenvolvimento Embrionário , Coração , Camundongos , Células-Tronco Embrionárias Murinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA