Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
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
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.
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
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.

7.
Development ; 147(3)2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-32014863

RESUMO

Cardiopharyngeal mesoderm (CPM) gives rise to muscles of the head and heart. Using genetic lineage analysis in mice, we show that CPM develops into a broad range of pharyngeal structures and cell types encompassing musculoskeletal and connective tissues. We demonstrate that CPM contributes to medial pharyngeal skeletal and connective tissues associated with both branchiomeric and somite-derived neck muscles. CPM and neural crest cells (NCC) make complementary mediolateral contributions to pharyngeal structures, in a distribution established in the early embryo. We further show that biallelic expression of the CPM regulatory gene Tbx1, haploinsufficient in 22q11.2 deletion syndrome patients, is required for the correct patterning of muscles with CPM-derived connective tissue. Our results suggest that CPM plays a patterning role during muscle development, similar to that of NCC during craniofacial myogenesis. The broad lineage contributions of CPM to pharyngeal structures provide new insights into congenital disorders and evolution of the mammalian pharynx.


Assuntos
Tecido Conjuntivo/embriologia , Desenvolvimento Muscular/genética , Faringe/embriologia , Somitos/fisiologia , Animais , Padronização Corporal/genética , Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Transgênicos , Crista Neural/metabolismo , Faringe/citologia , Somitos/citologia , Proteínas com Domínio T/metabolismo
8.
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
9.
Hum Mol Genet ; 28(14): 2295-2308, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31180501

RESUMO

Tbx1, the major candidate gene for DiGeorge or 22q11.2 deletion syndrome, is required for efficient incorporation of cardiac progenitors of the second heart field (SHF) into the heart. However, the mechanisms by which TBX1 regulates this process are still unclear. Here, we have used two independent models, mouse embryos and cultured cells, to define the role of TBX1 in establishing morphological and dynamic characteristics of SHF in the mouse. We found that loss of TBX1 impairs extracellular matrix (ECM)-integrin-focal adhesion (FA) signaling in both models. Mosaic analysis in embryos suggested that this function is non-cell autonomous, and, in cultured cells, loss of TBX1 impairs cell migration and FAs. Additionally, we found that ECM-mediated integrin signaling is disrupted upon loss of TBX1. Finally, we show that interfering with the ECM-integrin-FA axis between E8.5 and E9.5 in mouse embryos, corresponding to the time window within which TBX1 is required in the SHF, causes outflow tract dysmorphogenesis. Our results demonstrate that TBX1 is required to maintain the integrity of ECM-cell interactions in the SHF and that this interaction is critical for cardiac outflow tract development. More broadly, our data identifies a novel TBX1 downstream pathway as an important player in SHF tissue architecture and cardiac morphogenesis.


Assuntos
Matriz Extracelular/metabolismo , Coração/embriologia , Proteínas com Domínio T/fisiologia , Animais , Adesão Celular , Comunicação Celular , Movimento Celular , Polaridade Celular/genética , Células Cultivadas , Adesões Focais/genética , Adesões Focais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mioblastos/citologia , Mioblastos/metabolismo , Organogênese , Transdução de Sinais , Proteínas com Domínio T/genética
10.
Nature ; 520(7548): 466-73, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25903628

RESUMO

It has been more than 30 years since the publication of the new head hypothesis, which proposed that the vertebrate head is an evolutionary novelty resulting from the emergence of neural crest and cranial placodes. Neural crest generates the skull and associated connective tissues, whereas placodes produce sensory organs. However, neither crest nor placodes produce head muscles, which are a crucial component of the complex vertebrate head. We discuss emerging evidence for a surprising link between the evolution of head muscles and chambered hearts - both systems arise from a common pool of mesoderm progenitor cells within the cardiopharyngeal field of vertebrate embryos. We consider the origin of this field in non-vertebrate chordates and its evolution in vertebrates.


Assuntos
Evolução Biológica , Região Branquial/embriologia , Cabeça/anatomia & histologia , Cabeça/embriologia , Coração/anatomia & histologia , Coração/embriologia , Vertebrados/anatomia & histologia , Vertebrados/embriologia , Animais , Região Branquial/anatomia & histologia , Região Branquial/citologia , Mesoderma/citologia , Modelos Biológicos , Músculos/anatomia & histologia , Músculos/citologia , Músculos/embriologia , Crista Neural/citologia
11.
PLoS Genet ; 14(7): e1007502, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29979676

RESUMO

Left ventricular non-compaction (LVNC) is a rare cardiomyopathy associated with a hypertrabeculated phenotype and a large spectrum of symptoms. It is still unclear whether LVNC results from a defect of ventricular trabeculae development and the mechanistic basis that underlies the varying severity of this pathology is unknown. To investigate these issues, we inactivated the cardiac transcription factor Nkx2-5 in trabecular myocardium at different stages of trabecular morphogenesis using an inducible Cx40-creERT2 allele. Conditional deletion of Nkx2-5 at embryonic stages, during trabecular formation, provokes a severe hypertrabeculated phenotype associated with subendocardial fibrosis and Purkinje fiber hypoplasia. A milder phenotype was observed after Nkx2-5 deletion at fetal stages, during trabecular compaction. A longitudinal study of cardiac function in adult Nkx2-5 conditional mutant mice demonstrates that excessive trabeculation is associated with complex ventricular conduction defects, progressively leading to strain defects, and, in 50% of mutant mice, to heart failure. Progressive impaired cardiac function correlates with conduction and strain defects independently of the degree of hypertrabeculation. Transcriptomic analysis of molecular pathways reflects myocardial remodeling with a larger number of differentially expressed genes in the severe versus mild phenotype and identifies Six1 as being upregulated in hypertrabeculated hearts. Our results provide insights into the etiology of LVNC and link its pathogenicity with compromised trabecular development including compaction defects and ventricular conduction system hypoplasia.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Insuficiência Cardíaca/genética , Ventrículos do Coração/embriologia , Proteína Homeobox Nkx-2.5/metabolismo , Miocárdio Ventricular não Compactado Isolado/genética , Morfogênese/genética , Animais , Modelos Animais de Doenças , Feminino , Fibrose , Perfilação da Expressão Gênica , Ventrículos do Coração/patologia , Proteína Homeobox Nkx-2.5/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Miocárdio Ventricular não Compactado Isolado/complicações , Miocárdio Ventricular não Compactado Isolado/diagnóstico , Miocárdio Ventricular não Compactado Isolado/patologia , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Miocárdio/patologia , Ramos Subendocárdicos/patologia , Deleção de Sequência , Índice de Gravidade de Doença , Regulação para Cima
13.
Hum Mol Genet ; 27(21): 3747-3760, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30016433

RESUMO

The arterial and venous poles of the mammalian heart are hotspots of congenital heart defects (CHD) such as those observed in 22q11.2 deletion (or DiGeorge) and Holt-Oram syndromes. These regions of the heart are derived from late differentiating cardiac progenitor cells of the Second Heart Field (SHF) located in pharyngeal mesoderm contiguous with the elongating heart tube. The T-box transcription factor Tbx1, encoded by the major 22q11.2 deletion syndrome gene, regulates SHF addition to both cardiac poles from a common progenitor population. Despite the significance of this cellular addition the mechanisms regulating the deployment of common progenitor cells to alternate cardiac poles remain poorly understood. Here we demonstrate that Tbx5, mutated in Holt-Oram syndrome and essential for venous pole development, is activated in Tbx1 expressing cells in the posterior region of the SHF at early stages of heart tube elongation. A subset of the SHF transcriptional program, including Tbx1 expression, is subsequently downregulated in Tbx5 expressing cells, generating a transcriptional boundary between Tbx1-positive arterial pole and Tbx5-positive venous pole progenitor cell populations. We show that normal downregulation of the definitive arterial pole progenitor cell program in the posterior SHF is dependent on both Tbx1 and Tbx5. Furthermore, retinoic acid (RA) signaling is required for Tbx5 activation in Tbx1-positive cells and blocking RA signaling at the time of Tbx5 activation results in atrioventricular septal defects at fetal stages. Our results reveal sequential steps of cardiac progenitor cell patterning and provide mechanistic insights into the origin of common forms of CHD.


Assuntos
Anormalidades Múltiplas/metabolismo , Vasos Coronários/metabolismo , Síndrome de DiGeorge/metabolismo , Cardiopatias Congênitas/metabolismo , Comunicação Interatrial/metabolismo , Deformidades Congênitas das Extremidades Inferiores/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Proteínas com Domínio T/metabolismo , Tretinoína/metabolismo , Deformidades Congênitas das Extremidades Superiores/metabolismo , Anormalidades Múltiplas/genética , Animais , Síndrome de DiGeorge/genética , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/genética , Defeitos dos Septos Cardíacos/genética , Defeitos dos Septos Cardíacos/metabolismo , Comunicação Interatrial/genética , Deformidades Congênitas das Extremidades Inferiores/genética , Camundongos , Camundongos Transgênicos , Deformidades Congênitas das Extremidades Superiores/genética
14.
Circ Res ; 122(1): 142-154, 2018 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-29301846

RESUMO

The vertebrate heart tube forms from epithelial progenitor cells in the early embryo and subsequently elongates by progressive addition of second heart field (SHF) progenitor cells from adjacent splanchnic mesoderm. Failure to maximally elongate the heart results in a spectrum of morphological defects affecting the cardiac poles, including outflow tract alignment and atrioventricular septal defects, among the most common congenital birth anomalies. SHF cells constitute an atypical apicobasally polarized epithelium with dynamic basal filopodia, located in the dorsal wall of the pericardial cavity. Recent studies have highlighted the importance of epithelial architecture and cell adhesion in the SHF, particularly for signaling events that control the progenitor cell niche during heart tube elongation. The 22q11.2 deletion syndrome gene Tbx1 regulates progenitor cell status through modulating cell shape and filopodial activity and is required for SHF contributions to both cardiac poles. Noncanonical Wnt signaling and planar cell polarity pathway genes control epithelial polarity in the dorsal pericardial wall, as progenitor cells differentiate in a transition zone at the arterial pole. Defects in these pathways lead to outflow tract shortening. Moreover, new biomechanical models of heart tube elongation have been proposed based on analysis of tissue-wide forces driving epithelial morphogenesis in the SHF, including regional cell intercalation, cell cohesion, and epithelial tension. Regulation of the epithelial properties of SHF cells is thus emerging as a key step during heart tube elongation, adding a new facet to our understanding of the mechanisms underlying both heart morphogenesis and congenital heart defects.


Assuntos
Polaridade Celular/fisiologia , Desenvolvimento Embrionário/fisiologia , Epitélio/embriologia , Epitélio/fisiologia , Coração/embriologia , Coração/fisiologia , Animais , Humanos
15.
Dev Biol ; 444 Suppl 1: S337-S351, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30292786

RESUMO

Transcription factors that coordinate migration, differentiation or proliferation of enteric nervous system (ENS) precursors are not well defined. To identify novel transcriptional regulators of ENS development, we performed microarray analysis at embryonic day (E) 17.5 and identified many genes that were enriched in the ENS compared to other bowel cells. We decided to investigate the T-box transcription factor Tbx3, which is prominently expressed in developing and mature ENS. Haploinsufficiency for TBX3 causes ulnar-mammary syndrome (UMS) in humans, a multi-organ system disorder. TBX3 also regulates several genes known to be important for ENS development. To test the hypothesis that Tbx3 is important for ENS development or function, we inactivated Tbx3 in all neural crest derivatives, including ENS progenitors using Wnt1-Cre and a floxed Tbx3 allele. Tbx3 fl/fl; Wnt1-Cre conditional mutant mice die shortly after birth with cleft palate and difficulty feeding. The ENS of mutants was well-organized with a normal density of enteric neurons and nerve fiber bundles, but small bowel glial cell density was reduced. Despite this, bowel motility appeared normal. Furthermore, although Tbx3 is expressed in cardiac neural crest, Tbx3 fl/fl; Wnt1-Cre mice had structurally normal hearts. Thus, loss of Tbx3 within neural crest has selective effects on Tbx3-expressing neural crest derivatives.


Assuntos
Sistema Nervoso Entérico/embriologia , Crista Neural/embriologia , Proteínas com Domínio T/fisiologia , Animais , Diferenciação Celular , Movimento Celular , Fissura Palatina/embriologia , Fissura Palatina/genética , Coração/embriologia , Intestinos/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Crista Neural/metabolismo , Crista Neural/fisiologia , Neurogênese , Neuroglia/fisiologia , Neurônios , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/genética , Proteína Wnt1
16.
Pediatr Cardiol ; 40(7): 1331-1338, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31342111

RESUMO

Left ventricular noncompaction (LVNC) is a genetically heterogeneous disorder the etiology of which is still debated. During fetal development, trabecular cardiomyocytes contribute extensively to the working myocardium and the ventricular conduction system. The impact of developmental defects in trabecular myocardium in the etiology of LVNC has been debated. Recently we generated new mouse models of LVNC by the conditional deletion of the key cardiac transcription factor encoding gene Nkx2-5 in trabecular myocardium at critical steps of trabecular development. These conditional mutant mice recapitulate pathological features similar to those observed in LVNC patients, including a hypertrabeculated left ventricle with deep endocardial recesses, subendocardial fibrosis, conduction defects, strain defects, and progressive heart failure. After discussing recent findings describing the respective contribution of trabecular and compact myocardium during ventricular morphogenesis, this review will focus on new data reflecting the link between trabecular development and LVNC.


Assuntos
Ventrículos do Coração/anormalidades , Miocárdio Ventricular não Compactado Isolado/genética , Animais , Modelos Animais de Doenças , Ventrículos do Coração/embriologia , Humanos , Miocárdio Ventricular não Compactado Isolado/fisiopatologia , Masculino , Camundongos , Miocárdio/patologia , Miócitos Cardíacos/patologia , Deleção de Sequência
17.
Circ Res ; 128(3): 360-362, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33539223
18.
Proc Natl Acad Sci U S A ; 112(5): 1446-51, 2015 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-25605943

RESUMO

Neck muscles constitute a transition zone between somite-derived skeletal muscles of the trunk and limbs, and muscles of the head, which derive from cranial mesoderm. The trapezius and sternocleidomastoid neck muscles are formed from progenitor cells that have expressed markers of cranial pharyngeal mesoderm, whereas other muscles in the neck arise from Pax3-expressing cells in the somites. Mef2c-AHF-Cre genetic tracing experiments and Tbx1 mutant analysis show that nonsomitic neck muscles share a gene regulatory network with cardiac progenitor cells in pharyngeal mesoderm of the second heart field (SHF) and branchial arch-derived head muscles. Retrospective clonal analysis shows that this group of neck muscles includes laryngeal muscles and a component of the splenius muscle, of mixed somitic and nonsomitic origin. We demonstrate that the trapezius muscle group is clonally related to myocardium at the venous pole of the heart, which derives from the posterior SHF. The left clonal sublineage includes myocardium of the pulmonary trunk at the arterial pole of the heart. Although muscles derived from the first and second branchial arches also share a clonal relationship with different SHF-derived parts of the heart, neck muscles are clonally distinct from these muscles and define a third clonal population of common skeletal and cardiac muscle progenitor cells within cardiopharyngeal mesoderm. By linking neck muscle and heart development, our findings highlight the importance of cardiopharyngeal mesoderm in the evolution of the vertebrate heart and neck and in the pathophysiology of human congenital disease.


Assuntos
Coração/embriologia , Músculo Esquelético/embriologia , Pescoço/embriologia , Animais , Redes Reguladoras de Genes , Camundongos , Camundongos Transgênicos , Somitos
19.
Hum Mol Genet ; 24(6): 1704-16, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25410658

RESUMO

Outflow tract (OFT) malformation accounts for ∼30% of human congenital heart defects and manifests frequently in TBX1 haplo-insufficiency associated DiGeorge (22q11.2 deletion) syndrome. OFT myocardium originates from second heart field (SHF) progenitors in the pharyngeal and splanchnic mesoderm (SpM), but how these progenitors are deployed to the OFT is unclear. We find that SHF progenitors in the SpM gradually gain epithelial character and are deployed to the OFT as a cohesive sheet. Wnt5a, a non-canonical Wnt, is expressed specifically in the caudal SpM and may regulate oriented cell intercalation to incorporate SHF progenitors into an epithelial-like sheet, thereby generating the pushing force to deploy SHF cells rostrally into the OFT. Using enhancer trap and Cre transgenes, our lineage tracing experiments show that in Wnt5a null mice, SHF progenitors are trapped in the SpM and fail to be deployed to the OFT efficiently, resulting in a reduction in the inferior OFT myocardial wall and its derivative, subpulmonary myocardium. Concomitantly, the superior OFT and subaortic myocardium are expanded. Finally, in chick embryos, blocking the Wnt5a function in the caudal SpM perturbs polarized elongation of SHF progenitors, and compromises their deployment to the OFT. Collectively, our results highlight a critical role for Wnt5a in deploying SHF progenitors from the SpM to the OFT. Given that Wnt5a is a putative transcriptional target of Tbx1, and the similar reduction of subpulmonary myocardium in Tbx1 mutant mice, our results suggest that perturbing Wnt5a-mediated SHF deployment may be an important pathogenic mechanism contributing to OFT malformations in DiGeorge syndrome.


Assuntos
Síndrome de DiGeorge/genética , Células-Tronco Embrionárias/patologia , Coração/embriologia , Proteínas Wnt/genética , Animais , Embrião de Galinha , Síndrome de DiGeorge/etiologia , Deleção de Genes , Camundongos , Camundongos Knockout , Miocárdio/patologia , Proteína Wnt-5a
20.
Development ; 141(22): 4320-31, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25371366

RESUMO

Elongation of the vertebrate heart occurs by progressive addition of second heart field (SHF) cardiac progenitor cells from pharyngeal mesoderm to the poles of the heart tube. The importance of these cells in the etiology of congenital heart defects has led to extensive research into the regulation of SHF deployment by signaling pathways and transcription factors. However, the basic cellular features of these progenitor cells, including epithelial polarity, cell shape and cell dynamics, remain poorly characterized. Here, using immunofluorescence, live imaging and embryo culture, we demonstrate that SHF cells constitute an atypical, apicobasally polarized epithelium in the dorsal pericardial wall, characterized by apical monocilia and dynamic actin-rich basal filopodia. We identify the 22q11.2 deletion syndrome gene Tbx1, required in the SHF for outflow tract development, as a regulator of the epithelial properties of SHF cells. Cell shape changes in mutant embryos include increased circularity, a reduced basolateral membrane domain and impaired filopodial activity, and are associated with elevated aPKCζ levels. Activation of aPKCζ in embryo culture similarly impairs filopodia activity and phenocopies proliferative defects and ectopic differentiation observed in the SHF of Tbx1 null embryos. Our results reveal that epithelial and progenitor cell status are coupled in the SHF, identifying control of cell shape as a regulatory step in heart tube elongation and outflow tract morphogenesis.


Assuntos
Polaridade Celular/fisiologia , Epitélio/embriologia , Coração/embriologia , Morfogênese/fisiologia , Pseudópodes/fisiologia , Proteínas com Domínio T/genética , Animais , Western Blotting , Síndrome de DiGeorge/genética , Imuno-Histoquímica , Camundongos , Proteína Quinase C/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA