Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39082789

RESUMEN

Skeletal muscles of the head and trunk originate in distinct lineages with divergent regulatory programs converging on activation of myogenic determination factors. Branchiomeric head and neck muscles share a common origin with cardiac progenitor cells in cardiopharyngeal mesoderm (CPM). The retinoic acid (RA) signalling pathway is required during a defined early time window for normal deployment of cells from posterior CPM to the heart. Here we show that blocking RA signalling in the early mouse embryo also results in selective loss of the trapezius neck muscle, without affecting other skeletal muscles. RA signalling is required for robust expression of myogenic determination factors in posterior CPM and subsequent expansion of the trapezius primordium. Lineage specific activation of a dominant negative RA receptor reveals that trapezius development is not regulated by direct RA signalling to myogenic progenitor cells in CPM, or through neural crest cells, but indirectly through the somitic lineage, closely apposed with posterior CPM in the early embryo. These findings suggest that trapezius development is dependent on precise spatiotemporal interactions between cranial and somitic mesoderm at the head/trunk interface.

2.
J Mol Cell Cardiol ; 195: 1-13, 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39038734

RESUMEN

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.

3.
Dev Biol ; 499: 10-21, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37060937

RESUMEN

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.


Asunto(s)
Corazón , Miocardio , Diferenciación Celular/fisiología , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Mesodermo/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos , Animales , Ratones
4.
Circ Res ; 131(10): 842-858, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-36205127

RESUMEN

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.


Asunto(s)
Cardiopatías Congénitas , PPAR gamma , Animales , Ratones , Corazón , Cardiopatías Congénitas/genética , Miocardio/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Factores de Transcripción/metabolismo , Receptores Notch/metabolismo
5.
Adv Exp Med Biol ; 1441: 77-85, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884705

RESUMEN

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).


Asunto(s)
Modelos Animales de Enfermedad , Cardiopatías Congénitas , Corazón , Animales , Cardiopatías Congénitas/fisiopatología , Cardiopatías Congénitas/patología , Corazón/embriología , Corazón/fisiopatología , Corazón/crecimiento & desarrollo , Humanos , Ratones , Morfogénesis
6.
Adv Exp Med Biol ; 1441: 645-659, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884739

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Ventrículo Derecho con Doble Salida , Transducción de Señal , Tetralogía de Fallot , Tetralogía de Fallot/genética , Tetralogía de Fallot/patología , Tetralogía de Fallot/fisiopatología , Tetralogía de Fallot/embriología , Animales , Ventrículo Derecho con Doble Salida/genética , Ventrículo Derecho con Doble Salida/patología , Ventrículo Derecho con Doble Salida/fisiopatología , Humanos , Cresta Neural/metabolismo , Cresta Neural/patología , Cresta Neural/embriología , Morfogénesis/genética
7.
Adv Exp Med Biol ; 1441: 103-124, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884707

RESUMEN

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.


Asunto(s)
Linaje de la Célula , Animales , Humanos , Diferenciación Celular/genética , Linaje de la Célula/genética , Corazón/fisiología , Miocardio/citología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Transducción de Señal , Células Madre/metabolismo , Células Madre/citología , Células Madre/fisiología
8.
Differentiation ; : 100741, 2023 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-38040515

RESUMEN

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.

9.
Development ; 147(3)2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-32014863

RESUMEN

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.


Asunto(s)
Tejido Conectivo/embriología , Desarrollo de Músculos/genética , Faringe/embriología , Somitos/fisiología , Animales , Tipificación del Cuerpo/genética , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Transgénicos , Cresta Neural/metabolismo , Faringe/citología , Somitos/citología , Proteínas de Dominio T Box/metabolismo
10.
Exp Cell Res ; 410(1): 112931, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34798131

RESUMEN

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.


Asunto(s)
Desarrollo Embrionario/genética , Corazón/crecimiento & desarrollo , Mesodermo/crecimiento & desarrollo , Desarrollo de Músculos/genética , Animales , Diferenciación Celular/genética , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Cabeza/crecimiento & desarrollo , Ratones , Músculo Esquelético/crecimiento & desarrollo , Células Madre/citología , Pez Cebra/genética
11.
Hum Mol Genet ; 28(14): 2295-2308, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31180501

RESUMEN

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.


Asunto(s)
Matriz Extracelular/metabolismo , Corazón/embriología , Proteínas de Dominio T Box/fisiología , Animales , Adhesión Celular , Comunicación Celular , Movimiento Celular , Polaridad Celular/genética , Células Cultivadas , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mioblastos/citología , Mioblastos/metabolismo , Organogénesis , Transducción de Señal , Proteínas de Dominio T Box/genética
12.
Nature ; 520(7548): 466-73, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25903628

RESUMEN

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.


Asunto(s)
Evolución Biológica , Región Branquial/embriología , Cabeza/anatomía & histología , Cabeza/embriología , Corazón/anatomía & histología , Corazón/embriología , Vertebrados/anatomía & histología , Vertebrados/embriología , Animales , Región Branquial/anatomía & histología , Región Branquial/citología , Mesodermo/citología , Modelos Biológicos , Músculos/anatomía & histología , Músculos/citología , Músculos/embriología , Cresta Neural/citología
13.
PLoS Genet ; 14(7): e1007502, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29979676

RESUMEN

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.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Insuficiencia Cardíaca/genética , Ventrículos Cardíacos/embriología , Proteína Homeótica Nkx-2.5/metabolismo , No Compactación Aislada del Miocardio Ventricular/genética , Morfogénesis/genética , Animales , Modelos Animales de Enfermedad , Femenino , Fibrosis , Perfilación de la Expresión Génica , Ventrículos Cardíacos/patología , Proteína Homeótica Nkx-2.5/genética , Proteínas de Homeodominio/metabolismo , Humanos , No Compactación Aislada del Miocardio Ventricular/complicaciones , No Compactación Aislada del Miocardio Ventricular/diagnóstico , No Compactación Aislada del Miocardio Ventricular/patología , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Ramos Subendocárdicos/patología , Eliminación de Secuencia , Índice de Severidad de la Enfermedad , Regulación hacia Arriba
15.
Hum Mol Genet ; 27(21): 3747-3760, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30016433

RESUMEN

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.


Asunto(s)
Anomalías Múltiples/metabolismo , Vasos Coronarios/metabolismo , Síndrome de DiGeorge/metabolismo , Cardiopatías Congénitas/metabolismo , Defectos del Tabique Interatrial/metabolismo , Deformidades Congénitas de las Extremidades Inferiores/metabolismo , Transducción de Señal , Células Madre/metabolismo , Proteínas de Dominio T Box/metabolismo , Tretinoina/metabolismo , Deformidades Congénitas de las Extremidades Superiores/metabolismo , Anomalías Múltiples/genética , Animales , Síndrome de DiGeorge/genética , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/genética , Defectos de los Tabiques Cardíacos/genética , Defectos de los Tabiques Cardíacos/metabolismo , Defectos del Tabique Interatrial/genética , Deformidades Congénitas de las Extremidades Inferiores/genética , Ratones , Ratones Transgénicos , Deformidades Congénitas de las Extremidades Superiores/genética
16.
Circ Res ; 122(1): 142-154, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29301846

RESUMEN

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.


Asunto(s)
Polaridad Celular/fisiología , Desarrollo Embrionario/fisiología , Epitelio/embriología , Epitelio/fisiología , Corazón/embriología , Corazón/fisiología , Animales , Humanos
17.
Dev Biol ; 444 Suppl 1: S337-S351, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30292786

RESUMEN

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.


Asunto(s)
Sistema Nervioso Entérico/embriología , Cresta Neural/embriología , Proteínas de Dominio T Box/fisiología , Animales , Diferenciación Celular , Movimiento Celular , Fisura del Paladar/embriología , Fisura del Paladar/genética , Corazón/embriología , Intestinos/embriología , Ratones , Ratones Endogámicos C57BL , Cresta Neural/metabolismo , Cresta Neural/fisiología , Neurogénesis , Neuroglía/fisiología , Neuronas , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/genética , Proteína Wnt1
18.
Pediatr Cardiol ; 40(7): 1331-1338, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31342111

RESUMEN

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.


Asunto(s)
Ventrículos Cardíacos/anomalías , No Compactación Aislada del Miocardio Ventricular/genética , Animales , Modelos Animales de Enfermedad , Ventrículos Cardíacos/embriología , Humanos , No Compactación Aislada del Miocardio Ventricular/fisiopatología , Masculino , Ratones , Miocardio/patología , Miocitos Cardíacos/patología , Eliminación de Secuencia
19.
Circ Res ; 128(3): 360-362, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33539223
20.
Proc Natl Acad Sci U S A ; 112(5): 1446-51, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25605943

RESUMEN

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.


Asunto(s)
Corazón/embriología , Músculo Esquelético/embriología , Cuello/embriología , Animales , Redes Reguladoras de Genes , Ratones , Ratones Transgénicos , Somitos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA