Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
BMC Biol ; 21(1): 55, 2023 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-36941669

RESUMO

BACKGROUND: The reactivation of genetic programs from early development is a common mechanism for injury-induced organ regeneration. T-box 3 (TBX3) is a member of the T-box family of transcription factors previously shown to regulate pluripotency and subsequent lineage commitment in a number of tissues, including limb and lung. TBX3 is also involved in lung and heart organogenesis. Here, we provide a comprehensive and thorough characterization of TBX3 and its role during pancreatic organogenesis and regeneration. RESULTS: We interrogated the level and cell specificity of TBX3 in the developing and adult pancreas at mRNA and protein levels at multiple developmental stages in mouse and human pancreas. We employed conditional mutagenesis to determine its role in murine pancreatic development and in regeneration after the induction of acute pancreatitis. We found that Tbx3 is dynamically expressed in the pancreatic mesenchyme and epithelium. While Tbx3 is expressed in the developing pancreas, its absence is likely compensated by other factors after ablation from either the mesenchymal or epithelial compartments. In an adult model of acute pancreatitis, we found that a lack of Tbx3 resulted in increased proliferation and fibrosis as well as an enhanced inflammatory gene programs, indicating that Tbx3 has a role in tissue homeostasis and regeneration. CONCLUSIONS: TBX3 demonstrates dynamic expression patterns in the pancreas. Although TBX3 is dispensable for proper pancreatic development, its absence leads to altered organ regeneration after induction of acute pancreatitis.


Assuntos
Pancreatite , Adulto , Humanos , Animais , Camundongos , Doença Aguda , Pancreatite/genética , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Pâncreas/metabolismo , Organogênese/genética
2.
Development ; 147(22)2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-33046506

RESUMO

FGF8 signaling plays diverse roles in inner ear development, acting at multiple stages from otic placode induction to cellular differentiation in the organ of Corti. As a secreted morphogen with diverse functions, Fgf8 expression is likely to be spatially restricted and temporally dynamic throughout inner ear development. We evaluated these characteristics using genetic labeling mediated by Fgf8mcm gene-targeted mice and determined that Fgf8 expression is a specific and early marker of Type-I vestibular hair cell identity. Fgf8mcm expression initiates at E11.5 in the future striolar region of the utricle, labeling hair cells following EdU birthdating, and demonstrates that sub-type identity is determined shortly after terminal mitosis. This early fate specification is not apparent using markers or morphological criteria that are not present before birth in the mouse. Although analyses of Fgf8 conditional knockout mice did not reveal developmental phenotypes, the restricted pattern of Fgf8 expression suggests that functionally redundant FGF ligands may contribute to vestibular hair cell differentiation and supports a developmental model in which Type-I and Type-II hair cells develop in parallel rather than from an intermediate precursor.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Células Ciliadas Vestibulares/metabolismo , Sáculo e Utrículo/embriologia , Animais , Fator 8 de Crescimento de Fibroblasto/genética , Células Ciliadas Vestibulares/citologia , Camundongos , Camundongos Knockout , Sáculo e Utrículo/citologia
3.
Proc Natl Acad Sci U S A ; 117(27): 15712-15723, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32561646

RESUMO

The mechanisms used by embryos to pattern tissues across their axes has fascinated developmental biologists since the founding of embryology. Here, using single-cell technology, we interrogate complex patterning defects and define a Hedgehog (Hh)-fibroblast growth factor (FGF) signaling axis required for anterior mesoderm lineage development during gastrulation. Single-cell transcriptome analysis of Hh-deficient mesoderm revealed selective deficits in anterior mesoderm populations, culminating in defects to anterior embryonic structures, including the pharyngeal arches, heart, and anterior somites. Transcriptional profiling of Hh-deficient mesoderm during gastrulation revealed disruptions to both transcriptional patterning of the mesoderm and FGF signaling for mesoderm migration. Mesoderm-specific Fgf4/Fgf8 double-mutants recapitulated anterior mesoderm defects and Hh-dependent GLI transcription factors modulated enhancers at FGF gene loci. Cellular migration defects during gastrulation induced by Hh pathway antagonism were mitigated by the addition of FGF4 protein. These findings implicate a multicomponent signaling hierarchy activated by Hh ligands from the embryonic node and executed by FGF signals in nascent mesoderm to control anterior mesoderm patterning.


Assuntos
Fator 4 de Crescimento de Fibroblastos/genética , Fator 8 de Crescimento de Fibroblasto/genética , Gastrulação/genética , Proteína GLI1 em Dedos de Zinco/genética , Animais , Padronização Corporal/genética , Linhagem da Célula/genética , Embrião de Galinha , Fatores de Crescimento de Fibroblastos/genética , Gástrula/crescimento & desenvolvimento , Gástrula/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas Hedgehog/genética , Mesoderma/crescimento & desenvolvimento , Mesoderma/metabolismo , Camundongos , Transdução de Sinais/genética , Análise de Célula Única , Transcriptoma/genética
4.
Dev Dyn ; 251(9): 1613-1627, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35506352

RESUMO

The limb phenotypes of Tbx2 and Tbx3 mutants are distinct: loss of Tbx2 results in isolated duplication of digit 4 in the hindlimb while loss of Tbx3 results in anterior polydactyly and posterior oligodactly in the forelimb. In the face of such disparate phenotypes, we sought to determine whether Tbx2 and Tbx3 have functional redundancy during development of the mouse limb. We found that sequential loss of alleles generates defects that are not simply additive of those observed in single mutants and that multiple structures in both the forelimb and hindlimb display compound sensitivity to decreased gene dosage.


Assuntos
Extremidades , Proteínas com Domínio T/metabolismo , Animais , Membro Posterior , Camundongos , Fenótipo , Proteínas com Domínio T/genética
5.
Dev Dyn ; 251(10): 1711-1727, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35618654

RESUMO

BACKGROUND: Asymmetries in craniofacial anomalies are commonly observed. In the facial skeleton, the left side is more commonly and/or severely affected than the right. Such asymmetries complicate treatment options. Mechanisms underlying variation in disease severity between individuals as well as within individuals (asymmetries) are still relatively unknown. RESULTS: Developmental reductions in fibroblast growth factor 8 (Fgf8) have a dosage dependent effect on jaw size, shape, and symmetry. Further, Fgf8 mutants have directionally asymmetric jaws with the left side being more affected than the right. Defects in lower jaw development begin with disruption to Meckel's cartilage, which is discontinuous. All skeletal elements associated with the proximal condensation are dysmorphic, exemplified by a malformed and misoriented malleus. At later stages, Fgf8 mutants exhibit syngnathia, which falls into two broad categories: bony fusion of the maxillary and mandibular alveolar ridges and zygomatico-mandibular fusion. All of these morphological defects exhibit both inter- and intra-specimen variation. CONCLUSIONS: We hypothesize that these asymmetries are linked to heart development resulting in higher levels of Fgf8 on the right side of the face, which may buffer the right side to developmental perturbations. This mouse model may facilitate future investigations of mechanisms underlying human syngnathia and facial asymmetry.


Assuntos
Região Branquial , Coração , Animais , Fator 8 de Crescimento de Fibroblasto/genética , Humanos , Anormalidades Maxilomandibulares , Maxila , Camundongos , Anormalidades da Boca
6.
Development ; 146(4)2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30787001

RESUMO

Congenital heart disease (CHD) is the most common type of birth defect. In recent years, research has focussed on identifying the genetic causes of CHD. However, only a minority of CHD cases can be attributed to single gene mutations. In addition, studies have identified different environmental stressors that promote CHD, but the additive effect of genetic susceptibility and environmental factors is poorly understood. In this context, we have investigated the effects of short-term gestational hypoxia on mouse embryos genetically predisposed to heart defects. Exposure of mouse embryos heterozygous for Tbx1 or Fgfr1/Fgfr2 to hypoxia in utero increased the incidence and severity of heart defects while Nkx2-5+/- embryos died within 2 days of hypoxic exposure. We identified the molecular consequences of the interaction between Nkx2-5 and short-term gestational hypoxia, which suggest that reduced Nkx2-5 expression and a prolonged hypoxia-inducible factor 1α response together precipitate embryo death. Our study provides insight into the causes of embryo loss and variable penetrance of monogenic CHD, and raises the possibility that cases of foetal death and CHD in humans could be caused by similar gene-environment interactions.


Assuntos
Interação Gene-Ambiente , Cardiopatias Congênitas/genética , Coração/embriologia , Proteína Homeobox Nkx-2.5/genética , Proteínas de Homeodomínio/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Animais , Apoptose , Proliferação de Células , Embrião de Mamíferos/metabolismo , Feminino , Predisposição Genética para Doença , Coração/diagnóstico por imagem , Heterozigoto , Proteína Homeobox Nkx-2.5/fisiologia , Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxigênio/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Proteínas com Domínio T/genética , Fatores de Tempo
7.
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
8.
Pediatr Cardiol ; 40(7): 1388-1400, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31372681

RESUMO

In this article, we provide a brief summary of work by us and others to discover the molecular underpinnings of early conduction system development and function. We focus on how the multifunctional protein Tbx3 contributes to acquisition and homeostasis of the tissue-specific properties of the sinoatrial and atrioventricular nodes. We also provide unpublished, preliminary findings supporting the role of Tbx3-regulated alternative RNA processing in the developing conduction system.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Sistema de Condução Cardíaco/metabolismo , Animais , Nó Atrioventricular/fisiopatologia , Expressão Gênica , Sistema de Condução Cardíaco/embriologia , Humanos , RNA/metabolismo , Proteínas com Domínio T
9.
PLoS Genet ; 10(3): e1004247, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24675841

RESUMO

TBX3 is a member of the T-box family of transcription factors with critical roles in development, oncogenesis, cell fate, and tissue homeostasis. TBX3 mutations in humans cause complex congenital malformations and Ulnar-mammary syndrome. Previous investigations into TBX3 function focused on its activity as a transcriptional repressor. We used an unbiased proteomic approach to identify TBX3 interacting proteins in vivo and discovered that TBX3 interacts with multiple mRNA splicing factors and RNA metabolic proteins. We discovered that TBX3 regulates alternative splicing in vivo and can promote or inhibit splicing depending on context and transcript. TBX3 associates with alternatively spliced mRNAs and binds RNA directly. TBX3 binds RNAs containing TBX binding motifs, and these motifs are required for regulation of splicing. Our study reveals that TBX3 mutations seen in humans with UMS disrupt its splicing regulatory function. The pleiotropic effects of TBX3 mutations in humans and mice likely result from disrupting at least two molecular functions of this protein: transcriptional regulation and pre-mRNA splicing.


Assuntos
Anormalidades Múltiplas/genética , Processamento Alternativo/genética , Doenças Mamárias/genética , Mapas de Interação de Proteínas/genética , Proteínas com Domínio T/genética , Ulna/anormalidades , Anormalidades Múltiplas/patologia , Animais , Doenças Mamárias/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Mutação , Malformações do Sistema Nervoso , Proteômica/métodos , Precursores de RNA/genética , RNA Mensageiro/genética , Proteínas com Domínio T/biossíntese , Ulna/patologia
10.
Dev Biol ; 390(1): 68-79, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24613616

RESUMO

The vertebrate heart develops from mesoderm and requires inductive signals secreted from early endoderm. During embryogenesis, Nkx2.5 acts as a key transcription factor and plays essential roles for heart formation from Drosophila to human. In mice, Nkx2.5 is expressed in the early first heart field, second heart field pharyngeal mesoderm, as well as pharyngeal endodermal cells underlying the second heart field. Currently, the specific requirements for Nkx2.5 in the endoderm versus mesoderm with regard to early heart formation are incompletely understood. Here, we performed tissue-specific deletion in mice to dissect the roles of Nkx2.5 in the pharyngeal endoderm and mesoderm. We found that heart development appeared normal after endodermal deletion of Nkx2.5 whereas mesodermal deletion engendered cardiac defects almost identical to those observed on Nkx2.5 null embryos (Nkx2.5(-/-)). Furthermore, re-expression of Nkx2.5 in the mesoderm rescued Nkx2.5(-/-) heart defects. Our findings reveal that Nkx2.5 in the mesoderm is essential while endodermal expression is dispensable for early heart formation in mammals.


Assuntos
Coração/embriologia , Proteínas de Homeodomínio/genética , Mesoderma/metabolismo , Miocárdio/metabolismo , Fatores de Transcrição/genética , Animais , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/ultraestrutura , Endoderma/embriologia , Endoderma/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Mesoderma/embriologia , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Varredura , Faringe/embriologia , Faringe/metabolismo , Gravidez , Fatores de Tempo , Fatores de Transcrição/metabolismo
11.
Proc Natl Acad Sci U S A ; 109(3): E154-63, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22203979

RESUMO

TBX3 is critical for human development: mutations in TBX3 cause congenital anomalies in patients with ulnar-mammary syndrome. Data from mice and humans suggest multiple roles for Tbx3 in development and function of the cardiac conduction system. The mechanisms underlying the functional development, maturation, and maintenance of the conduction system are not well understood. We tested the requirements for Tbx3 in these processes. We generated a unique series of Tbx3 hypomorphic and conditional mouse mutants with varying levels and locations of Tbx3 activity within the heart, and developed techniques for evaluating in vivo embryonic conduction system function. Disruption of Tbx3 function in different regions of the developing heart causes discrete phenotypes and lethal arrhythmias: sinus pauses and bradycardia indicate sinoatrial node dysfunction, whereas preexcitation and atrioventricular block reveal abnormalities in the atrioventricular junction. Surviving Tbx3 mutants are at increased risk for sudden death. Arrhythmias induced by knockdown of Tbx3 in adults reveal its requirement for conduction system homeostasis. Arrhythmias in Tbx3-deficient embryos are accompanied by disrupted expression of multiple ion channels despite preserved expression of previously described conduction system markers. These findings indicate that Tbx3 is required for the conduction system to establish and maintain its correct molecular identity and functional properties. In conclusion, Tbx3 is required for the functional development, maturation, and homeostasis of the conduction system in a highly dosage-sensitive manner. TBX3 and its regulatory targets merit investigation as candidates for human arrhythmias.


Assuntos
Arritmias Cardíacas/fisiopatologia , Dosagem de Genes , Sistema de Condução Cardíaco/fisiopatologia , Homeostase/genética , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética , Alelos , Animais , Animais Recém-Nascidos , Arritmias Cardíacas/complicações , Arritmias Cardíacas/diagnóstico por imagem , Arritmias Cardíacas/patologia , Bloqueio Atrioventricular/complicações , Bloqueio Atrioventricular/diagnóstico por imagem , Bloqueio Atrioventricular/patologia , Bloqueio Atrioventricular/fisiopatologia , Nó Atrioventricular/patologia , Nó Atrioventricular/fisiopatologia , Conexina 43/metabolismo , Eletrocardiografia , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Sistema de Condução Cardíaco/anormalidades , Sistema de Condução Cardíaco/diagnóstico por imagem , Sistema de Condução Cardíaco/patologia , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos , Mutação/genética , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Recombinação Genética/genética , Análise de Sobrevida , Proteínas com Domínio T/metabolismo , Ultrassonografia
12.
PLoS One ; 19(2): e0293072, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38349913

RESUMO

We performed a forward genetic screen to discover peptides that specifically target breast cancer cells using a Penetratin tagged, random 15mer peptide library. We identified a group of novel peptides that specifically inhibited the proliferation and survival of breast cancer cells without affecting normal primary mammary epithelial cells or fibroblasts. The intrinsic apoptotic pathway is activated by these peptides in the face of abnormal expression of numerous cell cycle regulatory genes. Associated alterations in histone marks, nuclear structure, and levels of critical RNA binding proteins vary in a peptide specific manner. This study demonstrates a novel method for the discovery of new potential therapeutic peptides.


Assuntos
Neoplasias da Mama , Biblioteca de Peptídeos , Humanos , Feminino , Proliferação de Células , Peptídeos/química , Mama/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo
13.
Dev Biol ; 361(1): 125-36, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22040872

RESUMO

The interaction between the cranial neural crest (NC) and the epibranchial placode is critical for the formation of parasympathetic and visceral sensory ganglia, respectively. However, the molecular mechanism that controls this intercellular interaction is unknown. Here we show that the spatiotemporal expression of Fibroblast growth factor 8 (Fgf8) is strategically poised to control this cellular relationship. A global reduction of Fgf8 in hypomorph embryos leads to an early loss of placode-derived sensory ganglia and reduced number of NC-derived postganglionic (PG) neurons. The latter finding is associated with the early loss of NC cells by apoptosis. This loss occurs concurrent with the interaction between the NC and placode-derived ganglia. Conditional knockout of Fgf8 in the anterior mesoderm shows that this tissue source of Fgf8 has a specific influence on the formation of PG neurons. Unlike the global reduction of Fgf8, mesodermal loss of Fgf8 leads to a deficiency in PG neurons that is independent of NC apoptosis or defects in placode-derived ganglia. We further examined the differentiation of PG precursors by using a quantitative approach to measure the intensity of Phox2b, a PG neuronal determinant. We found reduced numbers and immature state of PG precursors emerging from the placode-derived ganglia en route to their terminal target areas. Our findings support the view that global expression of Fgf8 is required for early NC survival and differentiation of placode-derived sensory neurons, and reveal a novel role for mesodermal Fgf8 on the early differentiation of the NC along the parasympathetic PG lineage.


Assuntos
Diferenciação Celular/fisiologia , Fator 8 de Crescimento de Fibroblasto/metabolismo , Mesoderma/metabolismo , Crista Neural/embriologia , Neurônios Aferentes/fisiologia , Fibras Parassimpáticas Pós-Ganglionares/embriologia , Animais , Apoptose/fisiologia , Proteínas de Homeodomínio/metabolismo , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Fatores de Transcrição/metabolismo
14.
Invest Ophthalmol Vis Sci ; 64(5): 1, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37126314

RESUMO

Purpose: Familial exudative vitreoretinopathy (FEVR) and Norrie disease are examples of genetic disorders in which the retinal vasculature fails to fully form (hypovascular), leading to congenital blindness. While studying the role of a factor expressed during retinal development, T-box factor Tbx3, we discovered that optic cup loss of Tbx3 caused the retina to become hypovascular. The purpose of this study was to characterize how loss of Tbx3 affects retinal vasculature formation. Methods: Conditional removal of Tbx3 from both retinal progenitors and astrocytes was done using the optic cup-Cre recombinase driver BAC-Dkk3-Cre and was analyzed using standard immunohistochemical techniques. Results: With Tbx3 loss, the retinas were hypovascular, as seen in patients with retinopathy of prematurity (ROP) and FEVR. Retinal vasculature failed to form the stereotypic tri-layered plexus in the dorsal-temporal region. Astrocyte precursors were reduced in number and failed to form a lattice at the dorsal-temporal edge. We next examined retinal ganglion cells, as they have been shown to play a critical role in retinal angiogenesis. We found that melanopsin expression and Islet1/2-positive retinal ganglion cells were reduced in the dorsal half of the retina. In previous studies, the loss of melanopsin has been linked to hyaloid vessel persistence, which we also observed in the Tbx3 conditional knockout (cKO) retinas, as well as in infants with ROP or FEVR. Conclusions: To the best of our knowledge, these studies are the first demonstration that Tbx3 is required for normal mammalian eye formation. Together, the results provide a potential genetic model for retinal hypovascular diseases.


Assuntos
Degeneração Retiniana , Retinopatia da Prematuridade , Camundongos , Animais , Recém-Nascido , Humanos , Retina , Células Ganglionares da Retina , Vasos Retinianos , Vitreorretinopatias Exsudativas Familiares , Mamíferos , Proteínas com Domínio T
15.
Dev Biol ; 356(2): 383-97, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21664901

RESUMO

Heart development requires contributions from, and coordinated signaling interactions between, several cell populations, including splanchnic and pharyngeal mesoderm, postotic neural crest and the proepicardium. Here we report that Fgf3 and Fgf10, which are expressed dynamically in and near these cardiovascular progenitors, have redundant and dosage sensitive requirements in multiple aspects of early murine cardiovascular development. Embryos with Fgf3(-/+);Fgf10(-/-), Fgf3(-/-);Fgf10(-/+) and Fgf3(-/-);Fgf10(-/-) genotypes formed an allelic series of increasing severity with respect to embryonic survival, with double mutants dead by E11.5. Morphologic analysis of embryos with three mutant alleles at E11.5-E13.5 and double mutants at E9.5-E11.0 revealed multiple cardiovascular defects affecting the outflow tract, ventricular septum, atrioventricular cushions, ventricular myocardium, dorsal mesenchymal protrusion, pulmonary arteries, epicardium and fourth pharyngeal arch artery. Assessment of molecular markers in E8.0-E10.5 double mutants revealed abnormalities in each progenitor population, and suggests that Fgf3 and Fgf10 are not required for specification of cardiovascular progenitors, but rather for their normal developmental coordination. These results imply that coding or regulatory mutations in FGF3 or FGF10 could contribute to human congenital heart defects.


Assuntos
Vasos Coronários/fisiologia , Fator 10 de Crescimento de Fibroblastos/fisiologia , Fator 3 de Crescimento de Fibroblastos/fisiologia , Coração/embriologia , Neovascularização Fisiológica , Animais , Feminino , Fator 8 de Crescimento de Fibroblasto/genética , Camundongos , Crista Neural/anormalidades , Gravidez , Proteínas com Domínio T/genética
16.
Dev Biol ; 356(2): 308-22, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21600894

RESUMO

Systemic loss-of-function studies have demonstrated that Pax3 transcription factor expression is essential for dorsal neural tube, early neural crest and muscle cell lineage morphogenesis. Cardiac neural crest cells participate in both remodeling of the pharyngeal arch arteries and outflow tract septation during heart development, but the lineage specific role of Pax3 in neural crest function has not yet been determined. To gain insight into the requirement of Pax3 within the neural crest, we conditionally deleted Pax3 in both the premigratory and migratory neural crest populations via Wnt1-Cre and Ap2α-Cre and via P0-Cre in only the migratory neural crest, and compared these phenotypes to the pulmonary atresia phenotype observed following the systemic loss of Pax3. Surprisingly, using Wnt1-Cre deletion there are no resultant heart defects despite the loss of Pax3 from the premigratory and migratory neural crest. In contrast, earlier premigratory and migratory Ap2α-Cre mediated deletion resulted in double outlet right ventricle alignment heart defects. In order to assess the tissue-specific contribution of neural crest to heart development, genetic ablation of neural crest lineage using a Wnt1-Cre-activated diphtheria toxin fragment-A cell-killing system was employed. Significantly, ablation of Wnt1-Cre-expressing neural crest cells resulted in fully penetrant persistent truncus arteriosus malformations. Combined, the data show that Pax3 is essential for early neural crest progenitor formation, but is not required for subsequent cardiac neural crest progeny morphogenesis involving their migration to the heart or septation of the outflow tract.


Assuntos
Coração/embriologia , Morfogênese , Miocárdio/metabolismo , Crista Neural/embriologia , Fatores de Transcrição Box Pareados/fisiologia , Animais , Linhagem da Célula , Movimento Celular , Feminino , Integrases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/citologia , Miócitos Cardíacos/citologia , Fator de Transcrição PAX3 , Proteína Wnt1/fisiologia
17.
Development ; 136(18): 3173-83, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19700621

RESUMO

Elucidating the gene regulatory networks that govern pharyngeal arch artery (PAA) development is an important goal, as such knowledge can help to identify new genes involved in cardiovascular disease. The transcription factor Tbx1 plays a vital role in PAA development and is a major contributor to cardiovascular disease associated with DiGeorge syndrome. In this report, we used various genetic approaches to reveal part of a signalling network by which Tbx1 controls PAA development in mice. We investigated the crucial role played by the homeobox-containing transcription factor Gbx2 downstream of Tbx1. We found that PAA formation requires the pharyngeal surface ectoderm as a key signalling centre from which Gbx2, in response to Tbx1, triggers essential directional cues to the adjacent cardiac neural crest cells (cNCCs) en route to the caudal PAAs. Abrogation of this signal generates cNCC patterning defects leading to PAA abnormalities. Finally, we showed that the Slit/Robo signalling pathway is activated during cNCC migration and that components of this pathway are affected in Gbx2 and Tbx1 mutant embryos at the time of PAA development. We propose that the spatiotemporal control of this tightly orchestrated network of genes participates in crucial aspects of PAA development.


Assuntos
Artérias/embriologia , Padronização Corporal/fisiologia , Região Branquial , Movimento Celular/fisiologia , Ectoderma , Proteínas de Homeodomínio/metabolismo , Crista Neural/citologia , Proteínas com Domínio T/metabolismo , Animais , Artérias/anormalidades , Artérias/anatomia & histologia , Região Branquial/anormalidades , Região Branquial/irrigação sanguínea , Região Branquial/embriologia , Ectoderma/anatomia & histologia , Ectoderma/embriologia , Ectoderma/metabolismo , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Glicoproteínas/metabolismo , Coração/embriologia , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais/fisiologia , Proteínas com Domínio T/genética , Proteínas Roundabout
18.
Circ Res ; 106(3): 495-503, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20035084

RESUMO

RATIONALE: The genes encoding fibroblast growth factor (FGF) 8 and 10 are expressed in the anterior part of the second heart field that constitutes a population of cardiac progenitor cells contributing to the arterial pole of the heart. Previous studies of hypomorphic and conditional Fgf8 mutants show disrupted outflow tract (OFT) and right ventricle (RV) development, whereas Fgf10 mutants do not have detectable OFT defects. OBJECTIVES: Our aim was to investigate functional overlap between Fgf8 and Fgf10 during formation of the arterial pole. METHODS AND RESULTS: We generated mesodermal Fgf8; Fgf10 compound mutants with MesP1Cre. The OFT/RV morphology in these mutants was affected with variable penetrance; however, the incidence of embryos with severely affected OFT/RV morphology was significantly increased in response to decreasing Fgf8 and Fgf10 gene dosage. Fgf8 expression in the pharyngeal arch ectoderm is important for development of the pharyngeal arch arteries and their derivatives. We now show that Fgf8 deletion in the mesoderm alone leads to pharyngeal arch artery phenotypes and that these vascular phenotypes are exacerbated by loss of Fgf10 function in the mesodermal core of the arches. CONCLUSIONS: These results show functional overlap of FGF8 and FGF10 signaling from second heart field mesoderm during development of the OFT/RV, and from pharyngeal arch mesoderm during pharyngeal arch artery formation, highlighting the sensitivity of these key aspects of cardiovascular development to FGF dosage.


Assuntos
Região Branquial/irrigação sanguínea , Coração Fetal/crescimento & desenvolvimento , Fator 10 de Crescimento de Fibroblastos/fisiologia , Fator 8 de Crescimento de Fibroblasto/fisiologia , Cardiopatias Congênitas/embriologia , Animais , Região Branquial/anormalidades , Região Branquial/embriologia , Cruzamentos Genéticos , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/deficiência , Fator 10 de Crescimento de Fibroblastos/genética , Fator 8 de Crescimento de Fibroblasto/biossíntese , Fator 8 de Crescimento de Fibroblasto/deficiência , Fator 8 de Crescimento de Fibroblasto/genética , Deleção de Genes , Dosagem de Genes , Genótipo , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/patologia , Ventrículos do Coração/anormalidades , Ventrículos do Coração/embriologia , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos Mutantes , Obstrução do Fluxo Ventricular Externo/embriologia , Obstrução do Fluxo Ventricular Externo/genética
19.
Dev Biol ; 347(1): 92-108, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20727874

RESUMO

The fibroblast growth factor, FGF8, has been shown to be essential for vertebrate cardiovascular, craniofacial, brain and limb development. Here we report that Fgf8 function is required for normal progression through the late fetal stages of lung development that culminate in alveolar formation. Budding, lobation and branching morphogenesis are unaffected in early stage Fgf8 hypomorphic and conditional mutant lungs. Excess proliferation during fetal development disrupts distal airspace formation, mesenchymal and vascular remodeling, and Type I epithelial cell differentiation resulting in postnatal respiratory failure and death. Our findings reveal a previously unknown, critical role for Fgf8 function in fetal lung development and suggest that this factor may also contribute to postnatal alveologenesis. Given the high number of premature infants with alveolar dysgenesis and lung dysplasia, and the accumulating evidence that short-term benefits of available therapies may be outweighed by long-term detrimental effects on postnatal alveologenesis, the therapeutic implications of identifying a factor or pathway that can be targeted to stimulate normal alveolar development are profound.


Assuntos
Feto/embriologia , Feto/metabolismo , Fator 8 de Crescimento de Fibroblasto/metabolismo , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Diferenciação Celular , Proliferação de Células , Embrião de Mamíferos/metabolismo , Epitélio/metabolismo , Epitélio/patologia , Fator 8 de Crescimento de Fibroblasto/deficiência , Fator 8 de Crescimento de Fibroblasto/genética , Regulação da Expressão Gênica no Desenvolvimento , Integrases/metabolismo , Pulmão/anormalidades , Pulmão/irrigação sanguínea , Mesoderma/metabolismo , Mesoderma/patologia , Camundongos , Mutação/genética
20.
Dev Biol ; 342(1): 11-25, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20346941

RESUMO

Fibroblast growth factors (FGFs) are secreted molecules that activate the RAS/mitogen-activated protein kinase (MAPK) signaling pathway. In zebrafish development, FGF signaling is responsible for establishing dorsal polarity, maintaining the isthmic organizer, and cardiac ventricle formation. Because several ETS factors are known transcriptional mediators of MAPK signaling, we hypothesized that these factors function to mediate FGF signaling processes. In zebrafish, the simultaneous knock-down of three Pea3 ETS proteins, Etv5, Erm, and Pea3, produced phenotypes reminiscent of embryos deficient in FGF signaling. Morphant embryos displayed both cardiac and left/right patterning defects as well as disruption of the isthmic organizer. Furthermore, the expression of FGF target genes was abolished in Pea3 ETS depleted embryos. To understand how FGF signaling and ETS factors control gene expression, transcriptional regulation of dusp6 was studied in mouse and zebrafish. Conserved Pea3 ETS binding sites were identified within the Dusp6 promoter, and reporter assays showed that one of these sites is required for dusp6 induction by FGFs. We further demonstrated the interaction of Pea3 ETS factors with the Dusp6 promoter both in vitro and in vivo. These results revealed the requirement of ETS factors in transducing FGF signals in developmental processes.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Fatores de Transcrição/metabolismo , Peixe-Zebra/metabolismo , Animais , Sítios de Ligação/genética , Diferenciação Celular/genética , Fatores de Crescimento de Fibroblastos/genética , Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oncogenes , Proteínas Proto-Oncogênicas c-ets/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética , Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa