Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
PLoS Genet ; 9(1): e1003189, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23341776

RESUMO

Vertebrate organ development relies on the precise spatiotemporal orchestration of proliferation rates and differentiation patterns in adjacent tissue compartments. The underlying integration of patterning and cell cycle control during organogenesis is insufficiently understood. Here, we have investigated the function of the patterning T-box transcription factor gene Tbx2 in lung development. We show that lungs of Tbx2-deficient mice are markedly hypoplastic and exhibit reduced branching morphogenesis. Mesenchymal proliferation was severely decreased, while mesenchymal differentiation into fibrocytes was prematurely induced. In the epithelial compartment, proliferation was reduced and differentiation of alveolar epithelial cells type 1 was compromised. Prior to the observed cellular changes, canonical Wnt signaling was downregulated, and Cdkn1a (p21) and Cdkn1b (p27) (two members of the Cip/Kip family of cell cycle inhibitors) were strongly induced in the Tbx2-deficient lung mesenchyme. Deletion of both Cdkn1a and Cdkn1b rescued, to a large degree, the growth deficits of Tbx2-deficient lungs. Prolongation of Tbx2 expression into adulthood led to hyperproliferation and maintenance of mesenchymal progenitor cells, with branching morphogenesis remaining unaffected. Expression of Cdkn1a and Cdkn1b was ablated from the lung mesenchyme in this gain-of-function setting. We further show by ChIP experiments that Tbx2 directly binds to Cdkn1a and Cdkn1b loci in vivo, defining these two genes as direct targets of Tbx2 repressive activity in the lung mesenchyme. We conclude that Tbx2-mediated regulation of Cdkn1a and Cdkn1b represents a crucial node in the network integrating patterning information and cell cycle regulation that underlies growth, differentiation, and branching morphogenesis of this organ.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Pulmão , Proteínas com Domínio T , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Mesoderma , Camundongos , Morfogênese , Transdução de Sinais , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética
2.
PLoS Genet ; 9(4): e1003467, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23633963

RESUMO

Vertebrate limb outgrowth is driven by a positive feedback loop that involves Sonic hedgehog (Shh) and Gremlin1 (Grem1) in the posterior limb bud mesenchyme and Fibroblast growth factors (Fgfs) in the overlying epithelium. Proper spatio-temporal control of these signaling activities is required to avoid limb malformations such as polydactyly. Here we show that, in Tbx2-deficient hindlimbs, Shh/Fgf4 signaling is prolonged, resulting in increased limb bud size and duplication of digit 4. In turn, limb-specific Tbx2 overexpression leads to premature termination of this signaling loop with smaller limbs and reduced digit number as phenotypic manifestation. We show that Tbx2 directly represses Grem1 in distal regions of the posterior limb mesenchyme allowing Bone morphogenetic protein (Bmp) signaling to abrogate Fgf4/9/17 expression in the overlying epithelium. Since Tbx2 itself is a target of Bmp signaling, our data identify a growth-inhibiting positive feedback loop (Bmp/Tbx2/Grem1). We propose that proliferative expansion of Tbx2-expressing cells mediates self-termination of limb bud outgrowth due to their refractoriness to Grem1 induction.


Assuntos
Fatores de Crescimento de Fibroblastos/genética , Proteínas Hedgehog/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Botões de Extremidades/crescimento & desenvolvimento , Proteínas com Domínio T/genética , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Citocinas , Epitélio/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Botões de Extremidades/metabolismo , Mesoderma/crescimento & desenvolvimento , Mesoderma/metabolismo , Camundongos , Fenótipo , Transdução de Sinais , Proteínas com Domínio T/metabolismo
3.
Dev Biol ; 391(1): 54-65, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24727668

RESUMO

Otic fibrocytes tether the cochlear duct to the surrounding otic capsule but are also critically involved in maintenance of ion homeostasis in the cochlea, thus, perception of sound. The molecular pathways that regulate the development of this heterogenous group of cells from mesenchymal precursors are poorly understood. Here, we identified epithelial Wnt7a and Wnt7b as possible ligands of Fzd-mediated ß-catenin (Ctnnb1)-dependent (canonical) Wnt signaling in the adjacent undifferentiated periotic mesenchyme (POM). Mice with a conditional deletion of Ctnnb1 in the POM exhibited a complete failure of fibrocyte differentiation, a severe reduction of mesenchymal cells surrounding the cochlear duct, loss of pericochlear spaces, a thickening and partial loss of the bony capsule and a secondary disturbance of cochlear duct coiling shortly before birth. Analysis at earlier stages revealed that radial patterning of the POM in two domains with highly condensed cartilaginous precursors and more loosely arranged inner mesenchymal cells occurred normally but that proliferation in the inner domain was reduced and cytodifferentiation failed. Cells with mis/overexpression of a stabilized form of Ctnnb1 in the entire POM mesenchyme sorted to the inner mesenchymal compartment and exhibited increased proliferation. Our analysis suggests that Wnt signals from the cochlear duct epithelium are crucial to induce differentiation and expansion of fibrocyte precursor cells. Our findings emphasize the importance of epithelial-mesenchymal signaling in inner ear development.


Assuntos
Orelha Interna/embriologia , Orelha Interna/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Animais , Diferenciação Celular , Proliferação de Células , Ducto Coclear/metabolismo , Orelha Interna/citologia , Transição Epitelial-Mesenquimal , Feminino , Deleção de Genes , Masculino , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Fatores de Tempo , beta Catenina/metabolismo
4.
Nat Genet ; 38(6): 674-81, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16682973

RESUMO

The molecular basis of nephronophthisis, the most frequent genetic cause of renal failure in children and young adults, and its association with retinal degeneration and cerebellar vermis aplasia in Joubert syndrome are poorly understood. Using positional cloning, we here identify mutations in the gene CEP290 as causing nephronophthisis. It encodes a protein with several domains also present in CENPF, a protein involved in chromosome segregation. CEP290 (also known as NPHP6) interacts with and modulates the activity of ATF4, a transcription factor implicated in cAMP-dependent renal cyst formation. NPHP6 is found at centrosomes and in the nucleus of renal epithelial cells in a cell cycle-dependent manner and in connecting cilia of photoreceptors. Abrogation of its function in zebrafish recapitulates the renal, retinal and cerebellar phenotypes of Joubert syndrome. Our findings help establish the link between centrosome function, tissue architecture and transcriptional control in the pathogenesis of cystic kidney disease, retinal degeneration, and central nervous system development.


Assuntos
Fator 4 Ativador da Transcrição/genética , Antígenos de Neoplasias/genética , Mutação , Proteínas de Neoplasias/genética , Animais , Proteínas de Ciclo Celular , Proteínas do Citoesqueleto , Feminino , Ligação Genética , Humanos , Hibridização In Situ , Masculino , Linhagem , Síndrome , Peixe-Zebra
5.
Cell Mol Life Sci ; 69(8): 1377-89, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22130515

RESUMO

A key step in heart development is the coordinated development of the atrioventricular canal (AVC), the constriction between the atria and ventricles that electrically and physically separates the chambers, and the development of the atrioventricular valves that ensure unidirectional blood flow. Using knock-out and inducible overexpression mouse models, we provide evidence that the developmentally important T-box factors Tbx2 and Tbx3, in a functionally redundant manner, maintain the AVC myocardium phenotype during the process of chamber differentiation. Expression profiling and ChIP-sequencing analysis of Tbx3 revealed that it directly interacts with and represses chamber myocardial genes, and induces the atrioventricular pacemaker-like phenotype by activating relevant genes. Moreover, mutant mice lacking 3 or 4 functional alleles of Tbx2 and Tbx3 failed to form atrioventricular cushions, precursors of the valves and septa. Tbx2 and Tbx3 trigger development of the cushions through a regulatory feed-forward loop with Bmp2, thus providing a mechanism for the co-localization and coordination of these important processes in heart development.


Assuntos
Coxins Endocárdicos/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas com Domínio T/metabolismo , Animais , Sequência de Bases , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Coxins Endocárdicos/metabolismo , Humanos , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Miocárdio/metabolismo , Ratos , Proteínas com Domínio T/genética , Regulação para Cima
6.
Dev Biol ; 359(1): 95-107, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21925491

RESUMO

In the cochlea, sensory transduction depends on the endocochlear potential (EP) and the unique composition of the endolymph, both of which are maintained by a highly specialized epithelium at the cochlear lateral wall, the stria vascularis. The generation of the EP by the stria vascularis, in turn, relies on the insulation of an intrastrial extracellular compartment by epithelial basal cells. Despite the physiological importance of basal cells, their cellular origin and the molecular pathways that lead to their differentiation are unclear. Here, we show by genetic lineage tracing in the mouse that basal cells exclusively derive from the otic mesenchyme. Conditional deletion of E-cadherin in the otic mesenchyme and its descendants does not abrogate the transition from mesenchymal precursors to epithelial basal cells. Rather, dedifferentiation of intermediate cells, altered morphology of basal and marginal cells and hearing impairment due to decreased EP in E-cadherin mutant mice demonstrate an essential role of E-cadherin in terminal basal cell differentiation and their interaction with other strial cell types to establish and maintain the functional architecture of the stria vascularis.


Assuntos
Caderinas/genética , Estria Vascular/fisiologia , Animais , Sequência de Bases , Primers do DNA , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Estria Vascular/citologia
7.
Hum Mol Genet ; 19(24): 4918-29, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20881014

RESUMO

Congenital ureter anomalies, including hydroureter, affect up to 1% of the newborn children. Despite the prevalence of these developmental abnormalities in young children, the underlying molecular causes are only poorly understood. Here, we show that the high mobility group domain transcription factor Sox9 plays an important role in ureter development in the mouse. Transient Sox9 expression was detected in the undifferentiated ureteric mesenchyme and inactivation of Sox9 in this domain resulted in strong proximal hydroureter formation due to functional obstruction. Loss of Sox9 did not affect condensation, proliferation and apoptosis of the undifferentiated mesenchyme, but perturbed cyto-differentiation into smooth muscle cells (SMCs). Expression of genes encoding extracellular matrix (ECM) components was strongly reduced, suggesting that deficiency in ECM composition and/or signaling may underlie the observed defects. Prolonged expression of Sox9 in the ureteric mesenchyme led to increased deposition of ECM components and SMC dispersal. Furthermore, Sox9 genetically interacts with the T-box transcription factor 18 gene (Tbx18) during ureter development at two levels--as a downstream mediator of Tbx18 function and in a converging pathway. Together, our results argue that obstructive uropathies in campomelic dysplasia patients that are heterozygous for mutations in and around SOX9 arise from a primary requirement of Sox9 in the development of the ureteric mesenchyme.


Assuntos
Diferenciação Celular , Hidronefrose/genética , Hidronefrose/patologia , Mesoderma/patologia , Miócitos de Músculo Liso/patologia , Fatores de Transcrição SOX9/genética , Ureter/patologia , Animais , Diferenciação Celular/genética , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Matriz Extracelular/genética , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Rim/metabolismo , Rim/patologia , Mesoderma/metabolismo , Camundongos , Mutação/genética , Miócitos de Músculo Liso/metabolismo , Fatores de Transcrição SOX9/metabolismo , Ureter/crescimento & desenvolvimento , Ureter/metabolismo
8.
Histochem Cell Biol ; 137(3): 355-66, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22205279

RESUMO

Neph proteins are evolutionarily conserved members of the immunoglobulin superfamily of adhesion proteins and regulate morphogenesis and patterning of different tissues. They share a common protein structure consisting of extracellular immunoglobulin-like domains, a transmembrane region, and a carboxyl terminal cytoplasmic tail required for signaling. Neph orthologs have been widely characterized in invertebrates where they mediate such diverse processes as neural development, synaptogenesis, or myoblast fusion. Vertebrate Neph proteins have been described first at the glomerular filtration barrier of the kidney. Recently, there has been accumulating evidence suggesting a function of Neph proteins also outside the kidney. Here we demonstrate that Neph1, Neph2, and Neph3 are expressed differentially in various tissues during ontogenesis in mouse and chicken. Neph1 and Neph2 were found to be amply expressed in the central nervous system while Neph3 expression remained localized to the cerebellum anlage and the spinal cord. Outside the nervous system, Neph mRNAs were also differentially expressed in branchial arches, somites, heart, lung bud, and apical ectodermal ridge. Our findings support the concept that vertebrate Neph proteins, similarly to their Drosophila and C. elegans orthologs, provide guidance cues for cell recognition and tissue patterning in various organs which may open interesting perspectives for future research on Neph1-3 controlled morphogenesis.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Imunoglobulinas/genética , Proteínas de Membrana/genética , Animais , Região Branquial/embriologia , Região Branquial/fisiologia , Cerebelo/embriologia , Cerebelo/fisiologia , Embrião de Galinha , Galinhas , Ectoderma/embriologia , Ectoderma/fisiologia , Feminino , Coração/embriologia , Coração/fisiologia , Humanos , Pulmão/embriologia , Pulmão/fisiologia , Camundongos , Camundongos Endogâmicos , Filogenia , Gravidez , Somitos/embriologia , Somitos/fisiologia , Especificidade da Espécie , Medula Espinal/embriologia , Medula Espinal/fisiologia
9.
Circ Res ; 106(7): 1212-20, 2010 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-20185795

RESUMO

RATIONALE: The cardiac venous pole is a common focus of congenital malformations and atrial arrhythmias, yet little is known about the cellular and molecular mechanisms that regulate its development. The systemic venous return myocardium (sinus node and sinus horns) forms only late in cardiogenesis from a pool of pericardial mesenchymal precursor cells. OBJECTIVE: To analyze the cellular and molecular mechanisms directing the formation of the fetal sinus horns. METHODS AND RESULTS: We analyzed embryos deficient for the Wt1 (Wilms tumor 1) gene and observed a failure to form myocardialized sinus horns. Instead, the cardinal veins become embedded laterally in the pleuropericardial membranes that remain tethered to the lateral body wall by the persisting subcoelomic mesenchyme, a finding that correlates with decreased apoptosis in this region. We show by expression analysis and lineage tracing studies that Wt1 is expressed in the subcoelomic mesenchyme surrounding the cardinal veins, but that this Wt1-positive mesenchyme does not contribute cells to the sinus horn myocardium. Expression of the Raldh2 (aldehyde dehydrogenase family 1, subfamily A2) gene was lost from this mesenchyme in Wt1(-/-) embryos. Phenotypic analysis of Raldh2 mutant mice rescued from early cardiac defects by retinoic acid food supply revealed defects of the venous pole and pericardium highly similar to those of Wt1(-/-) mice. CONCLUSIONS: Pericardium and sinus horn formation are coupled and depend on the expansion and correct temporal release of pleuropericardial membranes from the underlying subcoelomic mesenchyme. Wt1 and downstream Raldh2/retinoic acid signaling are crucial regulators of this process. Thus, our results provide novel insight into the genetic and cellular pathways regulating the posterior extension of the mammalian heart and the formation of its coelomic lining.


Assuntos
Seio Coronário/metabolismo , Mesoderma/metabolismo , Pericárdio/metabolismo , Pleura/metabolismo , Transdução de Sinais , Nó Sinoatrial/metabolismo , Tretinoína/metabolismo , Proteínas WT1/metabolismo , Aldeído Oxirredutases/genética , Aldeído Oxirredutases/metabolismo , Animais , Apoptose , Linhagem da Célula , Seio Coronário/embriologia , Morte Fetal , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Idade Gestacional , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Pericárdio/embriologia , Fenótipo , Pleura/embriologia , Transdução de Sinais/genética , Nó Sinoatrial/embriologia , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Proteínas WT1/deficiência , Proteínas WT1/genética
10.
Dev Biol ; 342(1): 51-62, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20346939

RESUMO

Sox9 encodes an HMG-domain transcription factor that is critically required in numerous developmental processes such as chondrogenesis and otic placode formation. Here, we show that Sox9 is expressed in the mesenchyme surrounding the developing cochlea in the mouse suggesting that Sox9 may also control development of the otic fibrocyte compartment and the surrounding otic capsule. Tissue-specific inactivation of Sox9 in the periotic mesenchyme using a Tbx18(Cre) mouse line results in arrest of early chondrogenesis and consequently, in a lack of cochlear otic capsule formation. Furthermore, loss of Sox9 severely compromises expansion, differentiation and remodeling of the otic fibrocyte compartment. Early cell proliferation defects in the entire periotic mesenchyme of Sox9-deficient inner ears suggest a cell-autonomous function of Sox9 for the development of the inner mesenchymal compartment. Abnormal cochlear duct morphogenesis in Sox9 mutants including disruption of the coiling process is tightly associated with the onset of mesenchymal defects whereas the absence of major differentiation defects in the otic epithelium suggests that Sox9-dependent mesenchymal signals primarily control epithelial morphogenesis.


Assuntos
Cóclea/embriologia , Orelha Interna/citologia , Orelha/embriologia , Mesoderma/citologia , Fatores de Transcrição SOX9/fisiologia , Animais , Diferenciação Celular , Condrogênese/fisiologia , Epitélio/fisiologia , Camundongos , Camundongos Transgênicos , Morfogênese/fisiologia , Transdução de Sinais/fisiologia
11.
Circ Res ; 105(5): 442-52, 2009 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-19661464

RESUMO

RATIONALE: T-box transcription factors play critical roles in the coordinated formation of the working chambers and the atrioventricular canal (AVC). Tbx2 patterns embryonic myocardial cells to form the AVC and suppresses their differentiation into chamber myocardium. Tbx20-deficient embryos, which fail to form chambers, ectopically express Tbx2 throughout the entire heart tube, providing a potential mechanism for the function of Tbx20 in chamber differentiation. OBJECTIVE: To identify the mechanism of Tbx2 suppression by Tbx20 and to investigate the involvement of Tbx2 in Tbx20-mediated chamber formation. METHODS AND RESULTS: We generated Tbx20 and Tbx2 single and double knockout embryos and observed that loss of Tbx2 did not rescue the Tbx20-deficient heart from failure to form chambers. However, Tbx20 is required to suppress Tbx2 in the developing chambers, a prerequisite to localize its strong differentiation-inhibiting activity to the AVC. We identified a bone morphogenetic protein (Bmp)/Smad-dependent Tbx2 enhancer conferring AVC-restricted expression and Tbx20-dependent chamber suppression of Tbx2 in vivo. Unexpectedly, we found in transfection and localization studies in vitro that both Tbx20 and mutant isoforms of Tbx20 unable to bind DNA attenuate Bmp/Smad-dependent activation of Tbx2 by binding Smad1 and Smad5 and sequestering them from Smad4. CONCLUSIONS: Our data suggest that Tbx20 directly interferes with Bmp/Smad signaling to suppress Tbx2 expression in the chambers, thereby confining Tbx2 expression to the prospective AVC region.


Assuntos
Diferenciação Celular , Átrios do Coração/metabolismo , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/genética , Coxins Endocárdicos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Células HeLa , Átrios do Coração/embriologia , Ventrículos do Coração/embriologia , Humanos , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Mutação , Regiões Promotoras Genéticas , Transdução de Sinais/genética , Proteína Smad1/metabolismo , Proteína Smad4/metabolismo , Proteína Smad5/metabolismo , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética , Ativação Transcricional , Transfecção
12.
Dev Biol ; 336(2): 145-55, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19769959

RESUMO

Formation of the mammalian secondary palate is a highly regulated and complex process. Impairment of the underlying cellular and molecular programs often results in cleft palate, a common birth defect in mammals. Here we report that Tbx2 and Tbx3, two closely related genes encoding T-box transcription factors, are expressed in the mesenchyme of the mouse palatal structures during development. Mice homozygous mutant for Tbx2 and mice double heterozygous for Tbx2 and Tbx3 exhibit a cleft palate phenotype arguing for an important contribution of Tbx2 and Tbx3 to palatogenesis. In Tbx2-deficient embryos, the bilateral primordial palatal shelves form but are smaller and retarded in the outgrowth process. They do not make contact but retain the potential to fuse. Development of other craniofacial structures appears normal, suggesting that impaired palate formation in Tbx2-mutant mice is caused by a primary defect in the palatal shelf mesenchyme. This is further supported by increased cell proliferation and apoptosis accompanied by increased expression of Bmp4 and CyclinD1 in Tbx2-deficient palatal shelves. Hence, Tbx2 and Tbx3 function overlappingly to control growth of the palatal shelf mesenchyme.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Palato/embriologia , Proteínas com Domínio T/genética , Animais , Proteína Morfogenética Óssea 4/genética , Ciclina D1/genética , Hibridização In Situ , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas com Domínio T/fisiologia
13.
Hepatology ; 49(3): 969-78, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19140222

RESUMO

UNLABELLED: After specification of the hepatic endoderm, mammalian liver organogenesis progresses through a series of morphological stages that culminate in the migration of hepatocytes into the underlying mesenchyme to populate the hepatic lobes. Here, we show that in the mouse the transcriptional repressor Tbx3, a member of the T-box protein family, is required for the transition from a hepatic diverticulum with a pseudo-stratified epithelium to a cell-emergent liver bud. In Tbx3-deficient embryos, proliferation in the hepatic epithelium is severely reduced, hepatoblasts fail to delaminate, and cholangiocyte rather than hepatocyte differentiation occurs. Molecular analyses suggest that the primary function of Tbx3 is to maintain expression of hepatocyte transcription factors, including hepatic nuclear factor 4a (Hnf4a) and CCAAT/enhancer binding protein (C/EBP), alpha (Cebpa), and to repress expression of cholangiocyte transcription factors such as Onecut1 (Hnf6) and Hnf1b. CONCLUSION: Tbx3 controls liver bud expansion by suppressing cholangiocyte and favoring hepatocyte differentiation in the liver bud.


Assuntos
Ductos Biliares Extra-Hepáticos/embriologia , Diferenciação Celular/fisiologia , Células Epiteliais/citologia , Fígado/embriologia , Fígado/metabolismo , Organogênese/fisiologia , Proteínas com Domínio T/metabolismo , Animais , Ductos Biliares Extra-Hepáticos/citologia , Ductos Biliares Extra-Hepáticos/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Células Epiteliais/metabolismo , Feminino , Fator 1-beta Nuclear de Hepatócito/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Fator 6 Nuclear de Hepatócito/metabolismo , Fígado/citologia , Camundongos , Camundongos Knockout , Gravidez , Proteínas com Domínio T/genética
14.
J Clin Invest ; 116(3): 663-74, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16511601

RESUMO

Congenital malformations of the urinary tract are a major cause of renal failure in children and young adults. They are often caused by physical obstruction or by functional impairment of the peristaltic machinery of the ureter. The underlying molecular and cellular defects are, however, poorly understood. Here we present the phenotypic characterization of a new mouse model for congenital ureter malformation that revealed the molecular pathway important for the formation of the functional mesenchymal coating of the ureter. The gene encoding the T-box transcription factor Tbx18 was expressed in undifferentiated mesenchymal cells surrounding the distal ureter stalk. In Tbx18-/- mice, prospective ureteral mesenchymal cells largely dislocalized to the surface of the kidneys. The remaining ureteral mesenchymal cells showed reduced proliferation and failed to differentiate into smooth muscles, but instead became fibrous and ligamentous tissue. Absence of ureteral smooth muscles resulted in a short hydroureter and hydronephrosis at birth. Our analysis also showed that the ureteral mesenchyme derives from a distinct cell population that is separated early in kidney development from that of other mesenchymal cells of the renal system.


Assuntos
Diferenciação Celular/fisiologia , Mesoderma/fisiologia , Fatores de Transcrição/fisiologia , Ureter/embriologia , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Modelos Animais de Doenças , Triagem de Portadores Genéticos , Hidronefrose/genética , Hidronefrose/patologia , Mesoderma/citologia , Mesoderma/patologia , Camundongos , Camundongos Knockout , Técnicas de Cultura de Órgãos , Fenótipo , Proteínas com Domínio T , Fatores de Transcrição/biossíntese , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Ureter/anormalidades , Ureter/citologia , Ureter/patologia , Urotélio/metabolismo , Urotélio/patologia
15.
Mech Dev ; 122(2): 131-44, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15652702

RESUMO

During early limb development several signaling centers coordinate limb bud outgrowth as well as patterning. Members of the T-box gene family of transcriptional regulators are crucial players in these processes by activating and interpreting these signaling pathways. Here, we show that Tbx15, a member of this gene family, is expressed during limb development, first in the mesenchyme of the early limb bud, then during early endochondral bone development in prehypertrophic chondrocytes of cartilaginous templates. Expression is also found in mesenchymal precursor cells and prehypertrophic chondrocytes, respectively, during development of skeletal elements of the vertebral column and the head. Analysis of Tbx15 null mutant mice indicates a role of Tbx15 in the development of skeletal elements throughout the body. Mutants display a general reduction of bone size and changes of bone shape. In the forelimb skeleton, the scapula lacks the central region of the blade. Cartilaginous templates are already reduced in size and show a transient delay in ossification in mutant embryos. Mutants show a significantly reduced proliferation of prehypertrophic chondrocytes as well as of mesenchymal precursor cells. These data suggest that Tbx15 plays an important role in the development of the skeleton of the limb, vertebral column and head by controlling the number of mesenchymal precursor cells and chondrocytes.


Assuntos
Osso e Ossos/metabolismo , Extremidades/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação da Expressão Gênica , Mesoderma/metabolismo , Proteínas com Domínio T/biossíntese , Proteínas com Domínio T/fisiologia , Alelos , Animais , Apoptose , Padronização Corporal , Desenvolvimento Ósseo , Cartilagem/metabolismo , Proliferação de Células , Condrócitos/metabolismo , Primers do DNA/metabolismo , DNA Complementar/metabolismo , Éxons , Genótipo , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Mutação , Reação em Cadeia da Polimerase , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica
16.
J Biol Chem ; 283(37): 25372-25380, 2008 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-18644785

RESUMO

The compartmentalization of somites along their anterior-posterior axis is crucial to the segmental organization of the vertebral column. Anterior-posterior somite polarity is generated in the anterior presomitic mesoderm by Mesp2 and Delta/Notch signaling and is further maintained by two transcriptional regulators, Uncx4.1 and Tbx18, acting in the posterior and anterior somite compartment, respectively. Here, we report that the paired box transcription factor Pax3 cooperates with the T-box protein Tbx18 in maintaining anterior somite half identity. Our findings that both genes are co-expressed in the anterior presomitic mesoderm and in early somites, that Pax3 and Tbx18 proteins physically interact, and that the loss of Pax3 gene function enhances the vertebral defects (i.e. the gain of vertebral elements derived from posterior somite halves in Tbx18 mutant mice) suggests that the two proteins cooperatively regulate the gene expression program necessary for maintaining anterior-posterior somite polarity. Genetic interaction of Pax3 with Tbx18 and the closely related T-box gene Tbx15 was also observed in the development of the scapula blade, indicating an additional cooperative function for these genes in the paraxial mesoderm.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Mutação , Fator de Transcrição PAX3 , Escápula/embriologia , Somitos/metabolismo , Fatores de Tempo , Fatores de Transcrição/metabolismo , Técnicas do Sistema de Duplo-Híbrido
17.
Differentiation ; 74(4): 167-73, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16683987

RESUMO

Adult liver tissue regeneration may recapitulate molecular events of liver organogenesis. As gaps in our understanding of the fundamental processes that govern development and regeneration of the liver still exist, we studied gene expression in the developing liver at embryonic day 9.5 post coitum (E d9.5 p.c.). Microarray data from E d9.5 p.c. as well as previously published data from embryonic day 11.5 post coitum (E d11.5 p.c.) and embryonic day 13.5 post coitum (E d13.5 p.c.) were subjected to cluster analysis. This led to the identification of 130 genes which were characterized by continuous expression at all stages of liver development with peak expression of 44 genes at E d9.5 p.c. Five of these genes, previously not known to be associated with early liver development or with adult liver regeneration were selected for further analysis. The expression of the genes was studied by real-time polymerase chain reaction at 0, 2, 4, 6, 12, 24 and 48 hr after partial hepatectomy in the adult liver. Two of the genes, growth arrest protein 43 (GAP43) and paired-like homeodomain transcription factor 2 (Pitx2) were exclusively detected at 24 hr, whereas the genes Twist1, Midkine, and zinc finger protein of cerebellum 1 (Zic1) each showed a specific expression profile in the regenerating liver with peak expressions at 4, 24, and 6 hr, respectively. In summary, we were able to identify novel genes, that may act as regulators during liver formation as well as in the regeneration phase of adult liver. This information may contribute to the development of new targets for the treatment of liver diseases in the future.


Assuntos
Regulação da Expressão Gênica , Regeneração Hepática/genética , Fígado/embriologia , Fígado/metabolismo , Animais , Análise por Conglomerados , Embrião de Mamíferos/anatomia & histologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Hepatectomia , Hibridização In Situ , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos
18.
Development ; 132(12): 2697-707, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15901664

RESUMO

Tbx20, a member of the T-box family of transcriptional regulators, shows evolutionary conserved expression in the developing heart. In the mouse, Tbx20 is expressed in the cardiac crescent, then in the endocardium and myocardium of the linear and looped heart tube before it is restricted to the atrioventricular canal and outflow tract in the multi-chambered heart. Here, we show that Tbx20 is required for progression from the linear heart tube to a multi-chambered heart. Mice carrying a targeted mutation of Tbx20 show early embryonic lethality due to hemodynamic failure. A linear heart tube with normal anteroposterior patterning is established in the mutant. The tube does not elongate, indicating a defect in recruitment of mesenchyme from the secondary heart field, even though markers of the secondary heart field are not affected. Furthermore, dorsoventral patterning of the tube, formation of working myocardium, looping, and further differentiation and morphogenesis fail. Instead, Tbx2, Bmp2 and vinexin alpha (Sh3d4), genes normally restricted to regions of primary myocardium and lining endocardium, are ectopically expressed in the linear heart tube of Tbx20 mutant embryos. Because Tbx2 is both necessary and sufficient to repress chamber differentiation (Christoffels et al., 2004a; Harrelson et al., 2004), Tbx20 may ensure progression to a multi-chambered heart by repressing Tbx2 in the myocardial precursor cells of the linear heart tube destined to form the chambers.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Coração/anatomia & histologia , Coração/embriologia , Miocárdio/citologia , Miocárdio/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Apoptose , Padronização Corporal/genética , Diferenciação Celular , Proliferação de Células , Camundongos , Camundongos Knockout , Mutação/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética
19.
Genes Dev ; 18(10): 1209-21, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15155583

RESUMO

The compartmentalization of somites along their anterior-posterior (AP) axis is pivotal to the segmental organization of the vertebrate axial skeleton and the peripheral nervous system. Anterior and posterior somite halves contribute to different vertebral elements. They are also characterized by different proliferation rates and properties with respect to neural crest cell migration and spinal nerve passage. AP-somite polarity is generated in the anterior presomitic mesoderm by Mesp2 and Delta/Notch signaling. Here, we demonstrate that maintenance of AP-somite polarity is mediated by the T-box transcription factor Tbx18. Mice deficient for Tbx18 show expansion of pedicles with transverse processes and proximal ribs, elements derived from the posterior lateral sclerotome. AP-somite polarity is established in Tbx18 mutant embryos but is not maintained. During somite maturation, posterior somite compartments expand most likely because of posterior cells invading the anterior somite half. In the anterior lateral sclerotome, Tbx18 acts as an antiapoptotic factor. Ectopic expression experiments suggest that Tbx18 can promote anterior at the expense of posterior somite compartments. In summary, Tbx18 appears to act downstream of Mesp2 and Delta/Notch signaling to maintain the separation of anterior and posterior somite compartments.


Assuntos
Padronização Corporal/fisiologia , Somitos/citologia , Fatores de Transcrição/fisiologia , Animais , Sequência de Bases , Padronização Corporal/genética , DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Marcadores Genéticos , Hibridização In Situ , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Nervos Periféricos/embriologia , Coluna Vertebral/anormalidades , Coluna Vertebral/embriologia , Proteínas com Domínio T , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA