Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 147(4)2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-31988185

RESUMO

Organogenesis requires precise interactions between a developing tissue and its environment. In vertebrates, the developing eye is surrounded by a complex extracellular matrix as well as multiple mesenchymal cell populations. Disruptions to either the matrix or periocular mesenchyme can cause defects in early eye development, yet in many cases the underlying mechanism is unknown. Here, using multidimensional imaging and computational analyses in zebrafish, we establish that cell movements in the developing optic cup require neural crest. Ultrastructural analysis reveals that basement membrane formation around the developing eye is also dependent on neural crest, but only specifically around the retinal pigment epithelium. Neural crest cells produce the extracellular matrix protein nidogen: impairing nidogen function disrupts eye development, and, strikingly, expression of nidogen in the absence of neural crest partially restores optic cup morphogenesis. These results demonstrate that eye formation is regulated in part by extrinsic control of extracellular matrix assembly.This article has an associated 'The people behind the papers' interview.


Assuntos
Membrana Basal/embriologia , Olho/embriologia , Crista Neural/embriologia , Alelos , Animais , Sistemas CRISPR-Cas , Proteínas de Ligação ao Cálcio/fisiologia , Movimento Celular , Eletroforese Capilar , Matriz Extracelular/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Fatores de Transcrição Forkhead/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Mesoderma/embriologia , Microscopia Eletrônica de Transmissão , Morfogênese , Mutação , Crista Neural/citologia , Organogênese , Retina/embriologia , Epitélio Pigmentado da Retina/embriologia , Transdução de Sinais , Fator de Transcrição AP-2/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/fisiologia
2.
Development ; 146(13)2019 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31160420

RESUMO

Renal functional units known as nephrons undergo patterning events during development that create a segmental array of cellular compartments with discrete physiological identities. Here, from a forward genetic screen using zebrafish, we report the discovery that transcription factor AP-2 alpha (tfap2a) coordinates a gene regulatory network that activates the terminal differentiation program of distal segments in the pronephros. We found that tfap2a acts downstream of Iroquois homeobox 3b (irx3b), a distal lineage transcription factor, to operate a circuit consisting of tfap2b, irx1a and genes encoding solute transporters that dictate the specialized metabolic functions of distal nephron segments. Interestingly, this regulatory node is distinct from other checkpoints of differentiation, such as polarity establishment and ciliogenesis. Thus, our studies reveal insights into the genetic control of differentiation, where tfap2a is essential for regulating a suite of segment transporter traits at the final tier of zebrafish pronephros ontogeny. These findings have relevance for understanding renal birth defects, as well as efforts to recapitulate nephrogenesis in vivo to facilitate drug discovery and regenerative therapies.


Assuntos
Rim/embriologia , Néfrons/embriologia , Organogênese/genética , Fator de Transcrição AP-2/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Padronização Corporal/genética , Diferenciação Celular/genética , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Genes de Troca/fisiologia , Rim/metabolismo , Néfrons/metabolismo , Pronefro/embriologia , Pronefro/crescimento & desenvolvimento , Pronefro/metabolismo , Fator de Transcrição AP-2/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
3.
BMC Urol ; 21(1): 36, 2021 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-33691672

RESUMO

BACKGROUND: WW Domain Containing Oxidoreductase (WWOX) belongs to the unusual tumor suppressors, whose molecular function is not fully understood in bladder cancer, especially regarding interaction with Activator Protein 2 (AP-2) α/γ transcription factors. Thus, using lentiviral systems we created an in vitro model overexpressing or downregulating WWOX in CAL-29 cell line to assess invasiveness pathways. Surprisingly, while WWOX overexpression was accompanied with increased expression of both AP-2 factors, its downregulation only affected AP-2α level but not AP-2γ which remained high. METHODS: Using cellular models and unpaired t-test or Wilcoxon test, we investigated significant changes in biological processes: clonogenicity, extracellular matrix adhesion, metalloproteinases activity, 3D culture growth, proliferation, mitochondrial redox potential and invasiveness. Relative gene expression acquired through Real-Time qPCR has been analyzed by Welch's t-test. Additionally, using oncoprint analysis we distinguished groups for bioinformatics analyzes in order to perform a follow-up of in vitro experiments. RESULTS: Downregulation of WWOX in bladder cancer cell line intensified ability of single cell to grow into colony, mitochondrial redox potential and proliferation rate. Moreover, these cells shown elevated pro-MMP-2/9 activity but reduced adhesion to collagen I or laminin I, as well as distinct 3D culture growth. Through global in silico profiling we determined that WWOX alters disease-free survival of bladder cancer patients and modulates vital processes through AP-2 downstream effectors. CONCLUSIONS: Our research indicates that WWOX possesses tumor suppressor properties in bladder cancer but consecutive examination is required to entirely understand the contribution of AP-2γ or AP-2α.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fator de Transcrição AP-2/fisiologia , Proteínas Supressoras de Tumor/genética , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia , Oxidorredutase com Domínios WW/genética , Linhagem Celular Tumoral , Simulação por Computador , Humanos , Invasividade Neoplásica/genética
4.
Development ; 144(1): 33-37, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27888190

RESUMO

Epidermis and neural tissues differentiate from the ectoderm in animal embryos. Although epidermal fate is thought to be induced in vertebrate embryos, embryological evidence has indicated that no intercellular interactions during early stages are required for epidermal fate in ascidian embryos. To test this hypothesis, we determined the gene regulatory circuits for epidermal and neural specification in the ascidian embryo. These circuits started with Tfap2-r.b and Sox1/2/3, which are expressed in the ectodermal lineage immediately after zygotic genome activation. Tfap2-r.b expression was diminished in the neural lineages upon activation of fibroblast growth factor signaling, which is known to induce neural fate, and sustained only in the epidermal lineage. Tfap2-r.b specified the epidermal fate cooperatively with Dlx.b, which was activated by Sox1/2/3 This Sox1/2/3-Dlx.b circuit was also required for specification of the anterior neural fate. In the posterior neural lineage, Sox1/2/3 activated Nodal, which is required for specification of the posterior neural fate. Our findings support the hypothesis that the epidermal fate is specified autonomously in ascidian embryos.


Assuntos
Ciona intestinalis/embriologia , Ectoderma/embriologia , Fatores de Transcrição SOXB1/fisiologia , Fator de Transcrição AP-2/fisiologia , Urocordados/embriologia , Animais , Animais Geneticamente Modificados , Padronização Corporal/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Ciona intestinalis/genética , Ectoderma/metabolismo , Embrião não Mamífero , Epiderme/embriologia , Epiderme/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais/genética , Urocordados/genética
5.
Development ; 143(5): 787-98, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26811378

RESUMO

Loss of TFAP2C in mouse leads to developmental defects in the extra-embryonic compartment with lethality at embryonic day (E)7.5. To investigate the requirement of TFAP2C in later placental development, deletion of TFAP2C was induced throughout extra-embryonic ectoderm at E6.5, leading to severe placental abnormalities caused by reduced trophoblast population and resulting in embryonic retardation by E8.5. Deletion of TFAP2C in TPBPA(+) progenitors at E8.5 results in growth arrest of the junctional zone. TFAP2C regulates its target genes Cdkn1a (previously p21) and Dusp6, which are involved in repression of MAPK signaling. Loss of TFAP2C reduces activation of ERK1/2 in the placenta. Downregulation of Akt1 and reduced activation of phosphorylated AKT in the mutant placenta are accompanied by impaired glycogen synthesis. Loss of TFAP2C led to upregulation of imprinted gene H19 and downregulation of Slc38a4 and Ascl2. The placental insufficiency post E16.5 causes fetal growth restriction, with 19% lighter mutant pups. Knockdown of TFAP2C in human trophoblast choriocarcinoma JAr cells inhibited MAPK and AKT signaling. Thus, we present a model where TFAP2C in trophoblasts controls proliferation by repressing Cdkn1a and activating the MAPK pathway, further supporting differentiation of glycogen cells by activating the AKT pathway.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas da Gravidez/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição AP-2/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Cruzamentos Genéticos , Inibidor de Quinase Dependente de Ciclina p21/genética , Feminino , Deleção de Genes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Glicogênio/metabolismo , Hibridização In Situ , Integrases/genética , Integrases/fisiologia , Masculino , Camundongos , Fosforilação , Placenta/metabolismo , Gravidez , Proteínas da Gravidez/genética , Fator de Transcrição AP-2/genética , Transgenes , Trofoblastos/metabolismo
6.
EMBO J ; 33(4): 312-26, 2014 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-24451200

RESUMO

Androgen receptor (AR) binds male sex steroids and mediates physiological androgen actions in target tissues. ChIP-seq analyses of AR-binding events in murine prostate, kidney and epididymis show that in vivo AR cistromes and their respective androgen-dependent transcription programs are highly tissue specific mediating distinct biological pathways. This high order of tissue specificity is achieved by the use of exclusive collaborating factors in the three androgen-responsive tissues. We find two novel collaborating factors for AR signaling in vivo--Hnf4α (hepatocyte nuclear factor 4α) in mouse kidney and AP-2α (activating enhancer binding protein 2α) in mouse epididymis--that define tissue-specific AR recruitment. In mouse prostate, FoxA1 serves for the same purpose. FoxA1, Hnf4α and AP-2α motifs are over-represented within unique AR-binding loci, and the cistromes of these factors show substantial overlap with AR-binding events distinct to each tissue type. These licensing or pioneering factors are constitutively bound to chromatin and guide AR to specific genomic loci upon hormone exposure. Collectively, liganded receptor and its DNA-response elements are required but not sufficient for establishment of tissue-specific transcription programs.


Assuntos
Epididimo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Fator 4 Nuclear de Hepatócito/fisiologia , Rim/metabolismo , Próstata/metabolismo , Receptores Androgênicos/metabolismo , Testosterona/farmacologia , Fator de Transcrição AP-2/fisiologia , Animais , Linhagem Celular , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Epididimo/citologia , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos ICR , Orquiectomia , Especificidade de Órgãos , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Proteínas de Ligação a Tacrolimo/biossíntese , Proteínas de Ligação a Tacrolimo/genética , Testosterona/fisiologia , Transcrição Gênica
7.
PLoS Genet ; 11(8): e1005464, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26275148

RESUMO

Little is known about how the size of meristem cells is regulated and whether it participates in the control of meristem size in plants. Here, we report our findings on shoebox (shb), a mild gibberellin (GA) deficient rice mutant that has a short root meristem size. Quantitative analysis of cortical cell length and number indicates that shb has shorter, rather than fewer, cells in the root meristem until around the fifth day after sowing, from which the number of cortical cells is also reduced. These defects can be either corrected by exogenous application of bioactive GA or induced in wild-type roots by a dose-dependent inhibitory effect of paclobutrazol on GA biosynthesis, suggesting that GA deficiency is the primary cause of shb mutant phenotypes. SHB encodes an AP2/ERF transcription factor that directly activates transcription of the GA biosynthesis gene KS1. Thus, root meristem size in rice is modulated by SHB-mediated GA biosynthesis that regulates the elongation and proliferation of meristem cells in a developmental stage-specific manner.


Assuntos
Giberelinas/fisiologia , Meristema/crescimento & desenvolvimento , Oryza/crescimento & desenvolvimento , Proteínas de Plantas/fisiologia , Fator de Transcrição AP-2/fisiologia , Sequência de Bases , Sítios de Ligação , Proliferação de Células , Forma Celular , Regulação da Expressão Gênica no Desenvolvimento , Regulação da Expressão Gênica de Plantas , Meristema/citologia , Meristema/metabolismo , Oryza/citologia , Oryza/metabolismo , Reguladores de Crescimento de Plantas/fisiologia , Raízes de Plantas/citologia , Raízes de Plantas/crescimento & desenvolvimento , Raízes de Plantas/metabolismo
8.
Dev Biol ; 407(1): 75-89, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26277217

RESUMO

The species-specific morphology of digits in the tetrapod limb, including the length and number of metacarpal, metatarsal, and phalangeal bones, suggests that a common developmental mechanism for digit formation is modified in a species-specific manner. Here, we examined the function of the AP-2ß transcription factor in regulating digit length in the chicken autopod. Mutations in the gene encoding AP-2ß are associated with Char syndrome, a human autosomal dominant disorder. Char syndrome patients exhibit autopod skeletal defects, including loss of phalanges and shortened fingers, suggestive of a function for AP-2ß in normal digit development. The ectopic expression of two different dominant-negative forms of chick AP-2ß, equivalent to mutant forms associated with human Char syndrome, in the developing chick hindlimb bud resulted in defective digit formation, including reductions in the number and length of phalanges and metatarsals. A detailed analysis of the AP-2ß expression pattern in the limb bud indicated a correlation between the pattern/duration of AP-2ß expression in the limb mesenchyme and digit length in three amniote species, the chicken, mouse and gecko. In addition, we found that AP-2ß expression was downstream of Fgf signals from the apical ectodermal ridge, which is crucial in digit morphogenesis, and that excessive AP-2ß function resulted in dysregulated digit length. Taken together, these results suggest that AP-2ß functions as a novel transcriptional regulator for digit morphogenesis.


Assuntos
Extremidades/embriologia , Fator de Transcrição AP-2/fisiologia , Anormalidades Múltiplas/etiologia , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Embrião de Galinha , Galinhas , Permeabilidade do Canal Arterial/etiologia , Face/anormalidades , Fatores de Crescimento de Fibroblastos/fisiologia , Dedos/anormalidades , Proteínas Hedgehog/fisiologia , Humanos , Botões de Extremidades/embriologia , Botões de Extremidades/metabolismo , Camundongos , Morfogênese , Transdução de Sinais , Transcrição Gênica
9.
Proc Natl Acad Sci U S A ; 110(8): 2870-5, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23382213

RESUMO

The neural crest develops in vertebrate embryos within a discrete domain at the neural plate boundary and eventually gives rise to a migrating population of cells that differentiate into a multitude of derivatives. We have shown that the broad-complex, tramtrack and bric a brac (BTB) domain-containing factor potassium channel tetramerization domain containing 15 (Kctd15) inhibits neural crest formation, and we proposed that its function is to delimit the neural crest domain. Here we report that Kctd15 is a highly effective inhibitor of transcription factor activating enhancer binding protein 2 (AP-2) in zebrafish embryos and in human cells; AP-2 is known to be critical for several steps of neural crest development. Kctd15 interacts with AP-2α but does not interfere with its nuclear localization or binding to cognate sites in the genome. Kctd15 binds specifically to the activation domain of AP-2α and efficiently inhibits transcriptional activation by a hybrid protein composed of the regulatory protein Gal4 DNA binding and AP-2α activation domains. Mutation of one proline residue in the activation domain to an alanine (P59A) yields a protein that is highly active but largely insensitive to Kctd15. These results indicate that Kctd15 acts in the embryo at least in part by specifically binding to the activation domain of AP-2α, thereby blocking the function of this critical factor in the neural crest induction hierarchy.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Crista Neural/crescimento & desenvolvimento , Canais de Potássio/fisiologia , Fator de Transcrição AP-2/fisiologia , Imunoprecipitação da Cromatina , DNA/metabolismo , Células HEK293 , Humanos , Hibridização In Situ , Ligação Proteica , Fator de Transcrição AP-2/metabolismo
10.
EMBO J ; 30(13): 2569-81, 2011 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-21572391

RESUMO

Oestrogen receptor α (ERα) is key player in the progression of breast cancer. Recently, the cistrome and interactome of ERα were mapped in breast cancer cells, revealing the importance of spatial organization in oestrogen-mediated transcription. However, the underlying mechanism of this process is unclear. Here, we show that ERα binding sites (ERBS) identified from the Chromatin Interaction Analysis-Paired End DiTag of ERα are enriched for AP-2 motifs. We demonstrate the transcription factor, AP-2γ, which has been implicated in breast cancer oncogenesis, binds to ERBS in a ligand-independent manner. Furthermore, perturbation of AP-2γ expression impaired ERα DNA binding, long-range chromatin interactions, and gene transcription. In genome-wide analyses, we show that a large number of AP-2γ and ERα binding events converge together across the genome. The majority of these shared regions are also occupied by the pioneer factor, FoxA1. Molecular studies indicate there is functional interplay between AP-2γ and FoxA1. Finally, we show that most ERBS associated with long-range chromatin interactions are colocalized with AP-2γ and FoxA1. Together, our results suggest AP-2γ is a novel collaborative factor in ERα-mediated transcription.


Assuntos
Cromatina/metabolismo , Receptor alfa de Estrogênio/fisiologia , Fator de Transcrição AP-2/fisiologia , Transcrição Gênica , Sítios de Ligação/genética , Cromatina/genética , Imunoprecipitação da Cromatina , Análise por Conglomerados , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Análise em Microsséries , Modelos Biológicos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Ligação Proteica/fisiologia , RNA Interferente Pequeno/farmacologia , Elementos de Resposta/genética , Fator de Transcrição AP-2/antagonistas & inibidores , Fator de Transcrição AP-2/genética , Transcrição Gênica/genética , Transcrição Gênica/fisiologia , Células Tumorais Cultivadas
11.
Dev Dyn ; 243(10): 1298-309, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24753151

RESUMO

BACKGROUND: Transcription factors are critical in regulating lens development. The AP-2 family of transcription factors functions in differentiation, cell growth and apoptosis, and in lens and eye development. AP-2α, in particular, is important in early lens development, and when conditionally deleted at the placode stage defective separation of the lens vesicle from the surface ectoderm results. AP-2α's role during later stages of lens development is unknown. To address this, the MLR10-Cre transgene was used to delete AP-2α from the lens epithelium beginning at embryonic day (E) 10.5. RESULTS: The loss of AP-2α after lens vesicle separation resulted in morphological defects beginning at E18.5. By P4, a small highly vacuolated lens with a multilayered epithelium was evident in the MLR10-AP-2α mutants. Epithelial cells appeared elongated and expressed fiber cell specific ßB1 and γ-crystallins. Epithelial cell polarity and lens cell adhesion was disrupted and accompanied by the misexpression of ZO-1, N-Cadherin, and ß-catenin. Cell death was observed in the mutant lens epithelium between postnatal day (P) 14 and P30, and correlated with altered arrangements of cells within the epithelium. CONCLUSIONS: Our findings demonstrate that AP-2α continues to be required after lens vesicle separation to maintain a normal lens epithelial cell phenotype and overall lens integrity and to ensure correct fiber cell differentiation.


Assuntos
Cristalino/fisiologia , Fator de Transcrição AP-2/fisiologia , Animais , Catarata/genética , Adesão Celular/genética , Diferenciação Celular/genética , Polaridade Celular/genética , Embrião de Mamíferos , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Epitélio/metabolismo , Epitélio/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Cristalino/embriologia , Camundongos , Camundongos Transgênicos , Fenótipo
12.
Biochemistry (Mosc) ; 79(11): 1226-36, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25540008

RESUMO

The JcERF1 gene, which is related to the ERF family (ethylene responsive factor coding genes), was isolated and characterized from the oil tree Jatropha curcas. The JcERF1 protein contains conserved an AP2/EREBP DNA-binding domain of 58 amino acid residues. The JcERF1 gene could be induced by abscisic acid, high salinity, hormones, and osmotic stress, suggesting that JcERF1 is regulated by certain components of the stress-signaling pathway. The full-length and C-terminus of JcERF1 driven by the GAL4 promoter functioned effectively as a transactivator in yeast, while its N-terminus was completely inactive. Transient expression analysis using a JcERF1-mGFP fusion gene in onion epidermal cells revealed that the JcERF1 protein is targeted to the nucleus. Transgenic tobacco plants carrying CaMV35S::JcERF1 fragments were shown to be much more salt tolerant compared to wild-type plants. Our results indicate that JcERF1 is a new member of the ERF transcription factors family that may play an important role in tolerance to environmental stress.


Assuntos
Jatropha/genética , Nicotiana/genética , Tolerância ao Sal , Fator de Transcrição AP-2/fisiologia , Núcleo Celular/química , Expressão Gênica , Cebolas/genética , Cebolas/fisiologia , Epiderme Vegetal/genética , Epiderme Vegetal/fisiologia , Plantas Geneticamente Modificadas/fisiologia , Nicotiana/fisiologia , Fator de Transcrição AP-2/análise , Fator de Transcrição AP-2/genética
13.
Cancer Metastasis Rev ; 31(3-4): 621-32, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22684365

RESUMO

The progression of melanoma toward the metastatic phenotype occurs in a defined stepwise manner. While many molecular changes take place early in melanoma development, progression toward the malignant phenotype, most notably during the transition from the radial growth phase (RGP) to the vertical growth phase (VGP) involves deregulated expression of several transcription factors. For example, the switch from RGP to VGP is associated with the loss of the transcription factor AP2α and gain of transcriptional activity of cAMP-responsive element binding protein. Together with the upregulation of microphthalmia-associated transcription factor, activating transcription factor 2, nuclear factor kappa B, and other transcription factors, these changes lead to dysregulated expression or function of important cellular adhesion molecules, matrix degrading enzymes, survival factors, as well as other factors leading to metastatic melanoma. Additionally, recent evidence suggests that microRNAs and RNA editing machinery influence the expression of transcription factors or are regulated themselves by transcription factors. Many of the downstream signaling molecules regulated by transcription factors, such as protease activated receptor-1, interleukin-8, and MCAM/MUC18 represent new treatment prospects.


Assuntos
Melanoma/secundário , Fatores de Transcrição/fisiologia , Fator 1 Ativador da Transcrição/fisiologia , Fator 2 Ativador da Transcrição/fisiologia , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/tratamento farmacológico , Fator de Transcrição Associado à Microftalmia/fisiologia , NF-kappa B/fisiologia , Edição de RNA , Fator de Transcrição AP-2/fisiologia
14.
Dev Cell ; 14(1): 132-9, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18160346

RESUMO

While endocytosis can regulate morphogen distribution, its precise role in shaping these gradients is unclear. Even more enigmatic is the role of retromer, a complex that shuttles proteins between endosomes and the Golgi apparatus, in Wnt gradient formation. Here we report that DPY-23, the C. elegans mu subunit of the clathrin adaptor AP-2 that mediates the endocytosis of membrane proteins, regulates Wnt function. dpy-23 mutants display Wnt phenotypes, including defects in neuronal migration, neuronal polarity, and asymmetric cell division. DPY-23 acts in Wnt-expressing cells to promote these processes. MIG-14, the C. elegans homolog of the Wnt-secretion factor Wntless, also acts in these cells to control Wnt function. In dpy-23 mutants, MIG-14 accumulates at or near the plasma membrane. By contrast, MIG-14 accumulates in intracellular compartments in retromer mutants. Based on our observations, we propose that intracellular trafficking of MIG-14 by AP-2 and retromer plays an important role in Wnt secretion.


Assuntos
Complexo 2 de Proteínas Adaptadoras/fisiologia , Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/fisiologia , Proteínas de Transporte/fisiologia , Fator de Transcrição AP-2/fisiologia , Proteínas Wnt/fisiologia , Complexo 2 de Proteínas Adaptadoras/genética , Animais , Axônios/fisiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Transporte/genética , Endocitose , Homeostase , Peptídeos e Proteínas de Sinalização Intracelular , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo
15.
EMBO J ; 28(22): 3591-601, 2009 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-19798054

RESUMO

Overexpression of the activator protein (AP)-2gamma transcription factor in breast tumours has been identified as an independent predictor of poor outcome and failure of hormone therapy. To understand further the function of AP-2gamma in breast carcinoma, we have used an RNA interference and gene expression profiling strategy with the MCF-7 cell line as a model. Gene expression changes between control and silenced cells implicate AP-2gamma in the control of cell cycle progression and developmental signalling. A function for AP-2gamma in cell cycle control was verified using flow cytometry: AP-2gamma silencing led to a partial G1/S arrest and induction of the cyclin-dependent kinase inhibitor, p21cip/CDKN1A. Reporter and chromatin immunoprecipitation assays demonstrated a direct, functional interaction by AP-2gamma at the CDKN1A proximal promoter. AP-2gamma silencing coincided with acquisition of an active chromatin conformation at the CDKN1A locus and increased gene expression. These data provide a mechanism whereby AP-2gamma overexpression can promote breast epithelial proliferation and, coupled with previously published data, suggest how loss of oestrogen regulation of AP-2gamma may contribute to the failure of hormone therapy in patients.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/genética , Fator de Transcrição AP-2/fisiologia , Ciclo Celular/genética , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Células Tumorais Cultivadas
16.
Genome Res ; 20(4): 458-72, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20176728

RESUMO

Trophoblast stem cells (TS cells), derived from the trophectoderm (TE) of blastocysts, require transcription factors (TFs) and external signals (FGF4, INHBA/NODAL/TGFB1) for self-renewal. While many reports have focused on TF networks that regulate embryonic stem cell (ES cell) self-renewal and pluripotency, little is know about TF networks that regulate self-renewal in TS cells. To further understand transcriptional networks in TS cells, we used chromatin immunoprecipitation with DNA microarray hybridization (ChIP-chip) analysis to investigate targets of the TFs-TCFAP2C, EOMES, ETS2, and GATA3-and a chromatin remodeling factor, SMARCA4. We then evaluated the transcriptional states of target genes using transcriptome analysis and genome-wide analysis of histone H3 acetylation (AcH3). Our results describe previously unknown transcriptional networks in TS cells, including TF occupancy of genes involved in ES cell self-renewal and pluripotency, co-occupancy of TCFAP2C, SMARCA4, and EOMES at a significant number of genes, and transcriptional regulatory circuitry within the five factors. Moreover, RNAi depletion of Tcfap2c, Smarca4, and Eomes transcripts resulted in a loss of normal colony morphology and down-regulation of TS cell-specific genes, suggesting an important role for TCFAP2C, SMARCA4, and EOMES in TS cell self-renewal. Through genome-wide mapping and global expression analysis of five TF target genes, our data provide a comprehensive analysis of transcriptional networks that regulate TS cell self-renewal.


Assuntos
DNA Helicases/fisiologia , Redes Reguladoras de Genes , Proteínas Nucleares/fisiologia , Células-Tronco/fisiologia , Proteínas com Domínio T/fisiologia , Fator de Transcrição AP-2/fisiologia , Fatores de Transcrição/fisiologia , Trofoblastos/fisiologia , Animais , Sequência de Bases , Proliferação de Células , Células Cultivadas , Imunoprecipitação da Cromatina , DNA Helicases/genética , DNA Helicases/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Células-Tronco/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fator de Transcrição AP-2/genética , Fator de Transcrição AP-2/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Trofoblastos/citologia , Trofoblastos/metabolismo
17.
Am J Pathol ; 180(2): 661-71, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22182699

RESUMO

Interferon (IFN)-γ plays crucial roles in regulating both innate and adaptive immunity. The existence of IFN-γ receptor 1 (IFNGR1) molecules on the cell surface is a prerequisite to the initiation of IFN-γ signaling; low expression of IFNGR1 leads to a functional blockade of IFN-γ signaling. However, the molecular mechanisms by which IFNGR1 expression is controlled are unclear. In the present study, we demonstrated that IFNGR1 expression was reduced or lost in breast cancer. Heterogeneous IFNGR1 immunoreactivity appeared to be associated with the morphological heterogeneity of breast cancer, and loss of IFNGR1 expression was predominantly observed in poorly differentiated areas. We identified the functional activating protein (AP)-2 and specificity protein (SP)-1 sites within the IFNGR1 promoter. Ectopic expression of AP-2α drastically repressed the expression of IFNGR1 and hindered IFN-γ signaling, whereas AP-2α gene silencing elevated IFNGR1 levels. Overexpression of SP-1 effectively antagonized the repressive effects of AP-2α. Simultaneous recruitment of both transcription factors to the AP-2 and SP-1 motifs, respectively, in the IFNGR1 promoter was demonstrated, implying that AP-2α and SP-1 may synergistically modulate IFNGR1 transcription. Moreover, AP-2α overexpression in AP-2-deficient SW480 cells remarkably inhibited Stat1 phosphorylation and the anti-proliferative effects of IFN-γ, whereas knockdown of the AP-2α expression dramatically enhanced the sensitivities of HeLa cells highly expressing AP-2 to IFN-γ, indicating that dysregulation of AP-2α expression is associated with impaired IFN-γ actions in cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Interferon gama/antagonistas & inibidores , Receptores de Interferon/metabolismo , Fator de Transcrição Sp1/fisiologia , Fator de Transcrição AP-2/fisiologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Inativação Gênica/fisiologia , Humanos , Fosforilação , RNA Interferente Pequeno/fisiologia , Receptores de Interferon/genética , Fator de Transcrição STAT1/antagonistas & inibidores , Transdução de Sinais/fisiologia , Fator de Transcrição Sp1/metabolismo , Transfecção , Regulação para Cima , Receptor de Interferon gama
18.
Neuropsychobiology ; 68(2): 79-90, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23881096

RESUMO

BACKGROUND: The transcription factor AP-2ß has been shown to impact clinical and neuropsychological properties. Apparently, it regulates the transcription of genes that code for molecules which are part of the catecholaminergic transmission system. This investigation focuses on possible effects of the transcription factor AP-2ß intron 2 polymorphism on cognitive performance parameters. METHODS: This hypothesis-driven investigation examined the effects and interactions of the transcription factor AP-2ß intron 2 polymorphism, the Val158Met catechol-O-methyltransferase (COMT) polymorphism, and the variable number of tandem repeat polymorphism of monoamine oxidase A (MAOA) on cognitive performance parameters within a group of 200 healthy women (age: mean ± SD, 23.93 ± 3.33 years). RESULTS: The AP-2ß polymorphism significantly influenced cognitive performance (in particular, the Trail Making Test part B), whereas the MAOA and COMT polymorphisms did not. However, there was an interaction effect of the AP-2ß × MAOA × COMT genotypes on the decision bias ß of the degraded-stimulus version of the continuous performance task. Only the Val158Met COMT polymorphism showed an influence on personality questionnaires (openness and self-transcendence; NEO Five-Factor Inventory, Temperament and Character Inventory). CONCLUSION: The transcription factor AP-2ß intron 2 polymorphism had more influence on cognition than the MAOA and COMT polymorphisms. Possibly, the AP-2ß genotype might influence cognition through pathways other than those that regulate MAOA and COMT transcription. Interactions of transcription factor AP-2ß, COMT, and MAOA polymorphisms suggest higher leverage effects of transcription factor AP-2ß in subjects with high dopamine availability.


Assuntos
Catecol O-Metiltransferase/fisiologia , Cognição/fisiologia , Monoaminoxidase/fisiologia , Fator de Transcrição AP-2/fisiologia , Adulto , Catecol O-Metiltransferase/genética , Feminino , Voluntários Saudáveis , Humanos , Monoaminoxidase/genética , Testes Neuropsicológicos , Inventário de Personalidade , Polimorfismo Genético , Fator de Transcrição AP-2/genética
19.
Dev Biol ; 353(1): 38-49, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21377456

RESUMO

Defects in closure of embryonic tissues such as the neural tube, body wall, face and eye lead to severe birth defects. Cell adhesion is hypothesized to contribute to closure of the neural tube and body wall; however, potential molecular regulators of this process have not been identified. Here we identify an ENU-induced mutation in mice that reveals a molecular pathway of embryonic closure. Line2F homozygous mutant embryos fail to close the neural tube, body wall, face, and optic fissure, and they also display defects in lung and heart development. Using a new technology of genomic sequence capture and high-throughput sequencing of a 2.5Mb region of the mouse genome, we discovered a mutation in the grainyhead-like 2 gene (Grhl2). Microarray analysis revealed Grhl2 affects the expression of a battery of genes involved in cell adhesion and E-cadherin protein is drastically reduced in tissues that require Grhl2 function. The tissue closure defects in Grhl2 mutants are similar to that of AP-2α null mutants and AP-2α has been shown to bind to the promoter of E-cadherin. Therefore, we tested for a possible interaction between these genes. However, we find that Grhl2 and AP-2α do not regulate each other's expression, E-cadherin expression is normal in AP-2α mutants during neural tube closure, and Grhl2;AP-2α trans-heterozygous embryos are morphologically normal. Taken together, our studies point to a complex regulation of neural tube fusion and highlight the importance of comparisons between these two models to understand more fully the molecular pathways of embryonic tissue closure.


Assuntos
Moléculas de Adesão Celular/análise , Crista Neural/embriologia , Tubo Neural/embriologia , Fatores de Transcrição/fisiologia , Proteínas ADAM/análise , Proteína ADAMTS1 , Animais , Antígenos de Neoplasias/análise , Padronização Corporal , Caderinas/análise , Diferenciação Celular , Proliferação de Células , Molécula de Adesão da Célula Epitelial , Ensaios de Triagem em Larga Escala , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Fator de Transcrição AP-2/fisiologia
20.
Reprod Biomed Online ; 25(1): 12-20, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22560121

RESUMO

In recent years, knowledge regarding the genetic and epigenetic programmes governing specification, maintenance and differentiation of the extraembryonic lineage has advanced substantially. Establishment and analysis of mice deficient in genes implicated in trophoblast lineage and the option to generate and manipulate murine stem cell lines from the inner cell mass and the trophectoderm in vitro represent major advances. The activating enhancer binding protein 2 (AP2) family of transcription factors is expressed during mammalian development and in certain malignant diseases. This article summarizes the data regarding expression and function of murine Tcfap2 and human TFAP2 in extraembryonic development and differentiation. It also presents a model integrating Tcfap2c into the framework of trophoblast development and highlights the requirement of Tcfap2c to maintain trophoblast stem cells. With regard to human trophoblast cell-lineage restriction, the role of TFAP2C in lineage specification and maintenance is speculated upon. Furthermore, an overview of target genes of AP2 in mouse and human affecting placenta development and function is provided and the evidence suggesting that defects in regulating TFAP2 members might contribute to placental defects is discussed.


Assuntos
Fator de Transcrição AP-2/fisiologia , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Ectoderma/fisiologia , Feminino , Humanos , Camundongos , Camundongos Knockout , Gravidez , Células-Tronco/fisiologia , Trofoblastos/citologia , Trofoblastos/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA