Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Development ; 143(23): 4462-4473, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27770010

RESUMO

Holoprosencephaly (HPE) is defined as the incomplete separation of the two cerebral hemispheres. The pathology of HPE is variable and, based on the severity of the defect, HPE is divided into alobar, semilobar, and lobar. Using a novel hypomorphic Six3 allele, we demonstrate in mice that variability in Six3 dosage results in different HPE phenotypes. Furthermore, we show that whereas the semilobar phenotype results from severe downregulation of Shh expression in the rostral diencephalon ventral midline, the alobar phenotype is caused by downregulation of Foxg1 expression in the anterior neural ectoderm. Consistent with these results, in vivo activation of the Shh signaling pathway rescued the semilobar phenotype but not the alobar phenotype. Our findings show that variations in Six3 dosage result in different forms of HPE.


Assuntos
Cérebro/embriologia , Proteínas do Olho/genética , Haploinsuficiência/genética , Holoprosencefalia/genética , Proteínas de Homeodomínio/genética , Proteínas do Tecido Nervoso/genética , Animais , Linhagem Celular , Cérebro/anormalidades , Diencéfalo/embriologia , Diencéfalo/metabolismo , Ectoderma/metabolismo , Fatores de Transcrição Forkhead/biossíntese , Células HEK293 , Proteínas Hedgehog/biossíntese , Proteínas Hedgehog/metabolismo , Holoprosencefalia/patologia , Humanos , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/biossíntese , Transdução de Sinais/fisiologia , Proteína Homeobox SIX3
2.
J Clin Invest ; 120(10): 3568-77, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20890044

RESUMO

Retinal degeneration causes vision impairment and blindness in humans. If one day we are to harness the potential of stem cell-based cell replacement therapies to treat these conditions, it is imperative that we better understand normal retina development. Currently, the genes and mechanisms that regulate the specification of the neuroretina during vertebrate eye development remain unknown. Here, we identify sine oculis-related homeobox 3 (Six3) as a crucial player in this process in mice. In Six3 conditional-mutant mouse embryos, specification of the neuroretina was abrogated, but that of the retinal pigmented epithelium was normal. Conditional deletion of Six3 did not affect the initial development of the optic vesicle but did arrest subsequent neuroretina specification. Ectopic rostral expansion of Wnt8b expression was the major response to Six3 deletion and the leading cause for the specific lack of neuroretina, as ectopic Wnt8b expression in transgenic embryos was sufficient to suppress neuroretina specification. Using chromatin immunoprecipitation assays, we identified Six3-responsive elements in the Wnt8b locus and demonstrated that Six3 directly repressed Wnt8b expression in vivo. Our findings provide a molecular framework to the program leading to neuroretina differentiation and may be relevant for the development of novel strategies aimed at characterizing and eventually treating different abnormalities in eye formation.


Assuntos
Proteínas do Olho/fisiologia , Proteínas de Homeodomínio/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Placa Neural/embriologia , Retina/embriologia , Proteínas Wnt/antagonistas & inibidores , Animais , Camundongos , Camundongos Transgênicos , Fatores de Transcrição SOXB1/fisiologia , Proteínas Wnt/fisiologia , beta Catenina/fisiologia , Proteína Homeobox SIX3
3.
PLoS Biol ; 8(8)2010 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-20808958

RESUMO

The dentate gyrus has an important role in learning and memory, and adult neurogenesis in the subgranular zone of the dentate gyrus may play a role in the acquisition of new memories. The homeobox gene Prox1 is expressed in the dentate gyrus during embryonic development and adult neurogenesis. Here we show that Prox1 is necessary for the maturation of granule cells in the dentate gyrus during development and for the maintenance of intermediate progenitors during adult neurogenesis. We also demonstrate that Prox1-expressing intermediate progenitors are required for adult neural stem cell self-maintenance in the subgranular zone; thus, we have identified a previously unknown non-cell autonomous regulatory feedback mechanism that controls adult neurogenesis in this region of the mammalian brain. Finally, we show that the ectopic expression of Prox1 induces premature differentiation of neural stem cells.


Assuntos
Diferenciação Celular , Giro Denteado/citologia , Proteínas de Homeodomínio/farmacologia , Células-Tronco Neurais/citologia , Neurogênese/efeitos dos fármacos , Proteínas Supressoras de Tumor/farmacologia , Células-Tronco Adultas/citologia , Animais , Encéfalo/embriologia , Giro Denteado/embriologia , Giro Denteado/metabolismo , Feminino , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Neurônios/citologia , Neurônios/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
4.
Genes Dev ; 24(7): 696-707, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20360386

RESUMO

The homeobox gene Prox1 is crucial for mammalian lymphatic vascular development. In the absence of Prox1, lymphatic endothelial cells (LECs) are not specified. The maintenance of LEC identity also requires the constant expression of Prox1. However, the mechanisms controlling the expression of this gene in LECs remain poorly understood. The SRY-related gene Sox18 is required to induce Prox1 expression in venous LEC progenitors. Although Sox18 is also expressed in embryonic arteries, these vessels do not express Prox1, nor do they give rise to LECs. This finding suggests that some venous endothelial cell-specific factor is required for the activation of Prox1. Here we demonstrate that the nuclear hormone receptor Coup-TFII is necessary for the activation of Prox1 in embryonic veins by directly binding a conserved DNA domain in the regulatory region of Prox1. In addition, we show that the direct interaction between nuclear hormone receptors and Prox1 is also necessary for the maintenance of Prox1 expression during early stages of LEC specification and differentiation.


Assuntos
Fator II de Transcrição COUP/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Fator II de Transcrição COUP/genética , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Células Endoteliais/citologia , Deleção de Genes , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Proteínas Supressoras de Tumor/genética , Veias/embriologia
5.
Dev Cell ; 15(2): 236-47, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18694563

RESUMO

Holoprosencephaly (HPE), the most common forebrain malformation, is characterized by an incomplete separation of the cerebral hemispheres. Mutations in the homeobox gene SIX3 account for 1.3% of all cases of human HPE. Using zebrafish-based assays, we have now determined that HPE-associated Six3 mutant proteins function as hypomorphs. Haploinsufficiency of Six3 caused by deletion of one allele of Six3 or by replacement of wild-type Six3 with HPE-associated Six3 mutant alleles was sufficient to recapitulate in mouse models most of the phenotypic features of human HPE. We demonstrate that Shh is a direct target of Six3 in the rostral diencephalon ventral midline (RDVM). Reduced amounts of functional Six3 protein fail to activate Shh expression in the mutant RDVM and ultimately lead to HPE. These results identify Six3 as a direct regulator of Shh expression and reveal a crossregulatory loop between Shh and Six3 in the ventral forebrain.


Assuntos
Haploidia , Proteínas Hedgehog/metabolismo , Holoprosencefalia/patologia , Proteínas do Tecido Nervoso/deficiência , Prosencéfalo/patologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Apoptose , Padronização Corporal , Proliferação de Células , Diencéfalo/anormalidades , Diencéfalo/metabolismo , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/patologia , Embrião de Mamíferos/ultraestrutura , Embrião não Mamífero/anormalidades , Embrião não Mamífero/patologia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fenótipo , Prosencéfalo/embriologia , Transdução de Sinais , Somitos/embriologia , Somitos/metabolismo , Telencéfalo/anormalidades , Telencéfalo/metabolismo , Proteínas de Peixe-Zebra/genética , Proteína Homeobox SIX3
6.
Development ; 135(3): 441-50, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18094027

RESUMO

The homeobox gene Six3 represses Wnt1 transcription. It is also required in the anterior neural plate for the development of the mammalian rostral forebrain. We have now determined that at the 15- to 17-somite stage, the prospective diencephalon is the most-anterior structure in the Six3-null brain, and Wnt1 expression is anteriorly expanded. Consequently, the brain caudalizes, and at the 22- to 24-somite stage, the prospective thalamic territory is the most-anterior structure. At around E11.0, the pretectum replaces this structure. Analysis of Six3;Wnt1 double-null mice revealed that Six3-mediated repression of Wnt1 is necessary for the formation of the rostral diencephalon and that Six3 activity is required for the formation of the telencephalon. These results provide insight into the mechanisms that establish anteroposterior identity in the developing mammalian brain.


Assuntos
Padronização Corporal , Diencéfalo/embriologia , Proteínas do Tecido Nervoso/deficiência , Animais , Diencéfalo/metabolismo , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/metabolismo , Somitos/embriologia , Somitos/metabolismo , Telencéfalo/embriologia , Telencéfalo/metabolismo , Tálamo/embriologia , Tálamo/metabolismo , Proteínas Wnt/metabolismo , Proteína Homeobox SIX3
7.
Genes Dev ; 21(19): 2422-32, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17908929

RESUMO

The origin of the mammalian lymphatic vasculature has been debated for more than 100 years. Whether lymphatic endothelial cells have a single or dual, venous or mesenchymal origin remains controversial. To resolve this debate, we performed Cre/loxP-based lineage-tracing studies using mouse strains expressing Cre recombinase under the control of the Tie2, Runx1, or Prox1 promoter elements. These studies, together with the analysis of Runx1-mutant embryos lacking definitive hematopoiesis, conclusively determined that from venous-derived lymph sacs, lymphatic endothelial cells sprouted, proliferated, and migrated to give rise to the entire lymphatic vasculature, and that hematopoietic cells did not contribute to the developing lymph sacs. We conclude that the mammalian lymphatic system has a solely venous origin.


Assuntos
Linhagem da Célula , Células Endoteliais/citologia , Vasos Linfáticos/embriologia , Veias/citologia , Animais , Movimento Celular , Proliferação de Células , Subunidade alfa 2 de Fator de Ligação ao Core/análise , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Estruturas Embrionárias/química , Estruturas Embrionárias/citologia , Estruturas Embrionárias/efeitos dos fármacos , Células Endoteliais/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Proteínas de Homeodomínio/análise , Proteínas de Homeodomínio/genética , Integrases/genética , Camundongos , Camundongos Transgênicos , Receptor TIE-2/análise , Receptor TIE-2/genética , Tamoxifeno/farmacologia , Proteínas Supressoras de Tumor/análise , Proteínas Supressoras de Tumor/genética , Veias/embriologia
8.
Gene Expr Patterns ; 7(3): 252-7, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17084678

RESUMO

Recently, sequence analyses have identified a large number of opposite strand transcripts in the vertebrate genome. Although the transcripts appear to be spliced and polyadenylated, many of them are predicted to represent noncoding RNAs. High levels of noncoding transcripts of the Six3 Opposite Strand (Six3OS) were recently identified in the embryonic and postnatal retina of the mouse. In this study, we expanded those initial expression analyses, elucidated in detail the developmental expression profile of mouse Six3OS in the brain and visual system, and compared it with that of Six3. Our results show that Six3OS expression overlaps extensively with that of Six3 and is not altered in Six3-null embryos.


Assuntos
Desenvolvimento Embrionário/genética , Proteínas do Olho/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas do Tecido Nervoso/genética , RNA Antissenso/genética , RNA não Traduzido/genética , Animais , Encéfalo/embriologia , Embrião de Mamíferos/metabolismo , Perfilação da Expressão Gênica , Camundongos , Mutação , Retina/embriologia , Retina/metabolismo , Proteína Homeobox SIX3
9.
EMBO J ; 25(21): 5214-28, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17036046

RESUMO

During kidney development and in response to inductive signals, the metanephric mesenchyme aggregates, becomes polarized, and generates much of the epithelia of the nephron. As such, the metanephric mesenchyme is a renal progenitor cell population that must be replenished as epithelial derivatives are continuously generated. The molecular mechanisms that maintain the undifferentiated state of the metanephric mesenchymal precursor cells have not yet been identified. In this paper, we report that functional inactivation of the homeobox gene Six2 results in premature and ectopic differentiation of mesenchymal cells into epithelia and depletion of the progenitor cell population within the metanephric mesenchyme. Failure to renew the mesenchymal cells results in severe renal hypoplasia. Gain of Six2 function in cortical metanephric mesenchymal cells was sufficient to prevent their epithelial differentiation in an organ culture assay. We propose that in the developing kidney, Six2 activity is required for maintaining the mesenchymal progenitor population in an undifferentiated state by opposing the inductive signals emanating from the ureteric bud.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Homeodomínio/metabolismo , Córtex Renal/embriologia , Células-Tronco Mesenquimais/metabolismo , Néfrons/embriologia , Fatores de Transcrição/metabolismo , Animais , Córtex Renal/citologia , Células-Tronco Mesenquimais/citologia , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Néfrons/citologia , Organogênese/fisiologia , Fatores de Transcrição/deficiência , Urotélio/citologia , Urotélio/embriologia
10.
EMBO J ; 25(22): 5383-95, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17066077

RESUMO

The homeobox gene Six3 regulates forebrain development. Here we show that Six3 is also crucial for lens formation. Conditional deletion of mouse Six3 in the presumptive lens ectoderm (PLE) disrupted lens formation. In the most severe cases, lens induction and specification were defective, and the lens placode and lens were absent. In Six3-mutant embryos, Pax6 was downregulated, and Sox2 was absent in the lens preplacodal ectoderm. Using ChIP, electrophoretic mobility shift assay, and luciferase reporter assays, we determined that Six3 activates Pax6 and Sox2 expression. Misexpression of mouse Six3 into chick embryos promoted the ectopic expansion of the ectodermal Pax6 expression domain. Our results position Six3 at the top of the regulatory pathway leading to lens formation. We conclude that Six3 directly activates Pax6 and probably also Sox2 in the PLE and regulates cell autonomously the earliest stages of mammalian lens induction.


Assuntos
Proteínas do Olho/biossíntese , Proteínas do Olho/fisiologia , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/fisiologia , Cristalino/embriologia , Proteínas do Tecido Nervoso/fisiologia , Fatores de Transcrição Box Pareados/biossíntese , Proteínas Repressoras/biossíntese , Animais , Embrião de Galinha , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas do Olho/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Cristalino/anormalidades , Cristalino/metabolismo , Camundongos , Mutação , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Proteínas Repressoras/genética , Fatores de Transcrição SOXB1 , Transdução de Sinais , Transativadores/genética , Transativadores/metabolismo , Proteína Homeobox SIX3
11.
Hum Mol Genet ; 14(14): 2027-34, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15930016

RESUMO

Fragile X syndrome (FXS) is almost always caused by silencing of the FMR1 gene. The defects observed in FXS indicate that the normal FMR1 gene has a range of functions and plays a particularly prominent role during development. However, the mechanisms regulating FMR1 expression in vivo are not known. Here, we have tested the role of the transcription factor AP-2alpha in regulating Fmr1 expression. Chromatin immunoprecipitation showed that AP-2alpha associates with the Fmr1 promoter in vivo. Furthermore, Fmr1 transcript levels are reduced >4-fold in homozygous null AP-2alpha mutant mice at embryonic day 18.5 when compared with normal littermates. Notably, AP-2alpha exhibits a strong gene dosage effect, with heterozygous mice showing approximately 2-fold reduction in Fmr1 levels. Examination of conditional AP-2alpha mutant mice indicates that this transcription factor plays a major role in regulating Fmr1 expression in embryos, but not in adults. We further investigated the role of AP-2alpha in the developmental regulation of Fmr1 expression using the Xenopus animal cap assay. Over-expression of a dominant-negative AP-2alpha in Xenopus embryos led to reduced Fmr1 levels. Moreover, exogenous wild-type AP-2alpha rescued Fmr1 expression in embryos where endogenous AP-2alpha had been suppressed. We conclude that AP-2alpha associates with the Fmr1 promoter in vivo and selectively regulates Fmr1 transcription during embryonic development.


Assuntos
Síndrome do Cromossomo X Frágil/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Transcrição Gênica/fisiologia , Animais , Sequência de Bases , Northern Blotting , DNA , Células HeLa , Humanos , Hibridização In Situ , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Xenopus
12.
Genes Dev ; 17(3): 368-79, 2003 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-12569128

RESUMO

In vertebrate embryos, formation of anterior neural structures requires suppression of Wnt signals emanating from the paraxial mesoderm and midbrain territory. In Six3(-/-) mice, the prosencephalon was severely truncated, and the expression of Wnt1 was rostrally expanded, a finding that indicates that the mutant head was posteriorized. Ectopic expression of Six3 in chick and fish embryos, together with the use of in vivo and in vitro DNA-binding assays, allowed us to determine that Six3 is a direct negative regulator of Wnt1 expression. These results, together with those of phenotypic rescue of headless/tcf3 zebrafish mutants by mouse Six3, demonstrate that regionalization of the vertebrate forebrain involves repression of Wnt1 expression by Six3 within the anterior neuroectoderm. Furthermore, these results support the hypothesis that a Wnt signal gradient specifies posterior fates in the anterior neural plate.


Assuntos
Proteínas de Homeodomínio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Prosencéfalo/embriologia , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Peixe-Zebra , Animais , Ectoderma/fisiologia , Proteínas do Olho , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Prosencéfalo/anormalidades , Proteínas Wnt , Proteína Wnt1 , Peixe-Zebra/genética , Proteína Homeobox SIX3
13.
Development ; 129(12): 2835-49, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12050133

RESUMO

Recent findings suggest that Six3, a member of the evolutionarily conserved So/Six homeodomain family, plays an important role in vertebrate visual system development. However, little is known about the molecular mechanisms by which this function is accomplished. Although several members of the So/Six gene family interact with members of the eyes absent (Eya) gene family and function as transcriptional activators, Six3 does not interact with any known member of the Eya family. Here, we report that Grg4 and Grg5, mouse counterparts of the Drosophila transcriptional co-repressor Groucho, interact with mouse Six3 and its closely related member Six6, which may also be involved in vertebrate eye development. The specificity of the interaction was validated by co-immunoprecipitation of Six3 and Grg4 complexes from cell lines. We also show that the interaction between Six3 and Grg5 requires the Q domain of Grg5 and a conserved phenylalanine residue present in an eh1-like motif located in the Six domain of Six3. The pattern of Grg5 expression in the mouse ventral forebrain and developing optic vesicles overlapped that previously reported for Six3 and Six6. Using PCR, we identified a specific DNA motif that is bound by Six3 and we demonstrated that Six3 acts as a potent transcriptional repressor upon its interaction with Groucho-related members. We also demonstrated that this interaction is required for Six3 auto repression. The biological significance of this interaction in the retina and lens was assessed by overexpression experiments using either wild type full-length Six3 cDNA or a mutated form of this gene in which the interaction with Groucho proteins was disrupted. Overexpression of wild type Six3 by in vivo retroviral infection of newborn rat retinae led to an altered photoreceptor phenotype, while the in ovo electroporation of chicken embryos resulted in failure of lens placode invagination and production of delta-crystallin-negative cells within the placode. These specific alterations were not seen when the mutated form of Six3 cDNA was used in similar experimental approaches, indicating that Six3 interaction with Groucho proteins plays an essential role in vertebrate eye development.


Assuntos
Olho/crescimento & desenvolvimento , Proteínas de Homeodomínio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Repressoras/metabolismo , Motivos de Aminoácidos , Animais , Animais Recém-Nascidos , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Linhagem Celular , Embrião de Galinha , Proteínas Correpressoras , Sequência Conservada , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas do Olho , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Mutação , Proteínas do Tecido Nervoso/genética , Testes de Precipitina , Regiões Promotoras Genéticas , Prosencéfalo/embriologia , Prosencéfalo/metabolismo , Ratos , Proteínas Repressoras/genética , Retina/crescimento & desenvolvimento , Fatores de Transcrição , Proteína Homeobox SIX3
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...