Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Biochem Biophys Res Commun ; 523(2): 411-415, 2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-31870549

RESUMO

Pou3f2/Brn2 is a transcription factor that helps to determine the cellular identity of neocortical or hypothalamic neurons. Mammalian Pou3f2 contains three homopolymeric amino acids that are not present in amphibian Pou3f2. These amino acids contribute to monoamine function, which may play specific roles in mammalian development and behavior. Previous work has indicated that Pou3f2⊿ mice, which lack the homopolymeric amino acids, exhibited declined maternal activity and impaired object and spatial recognition. The current study, analyzed weight gain, brain development, home cage activity, social interaction, and response to novel objects in Pou3f2⊿ mice to determine which aspects of behavior were affected by monoamine dysregulation. Compared to their wild type counterparts, Pou3f2⊿ mice showed decreased social interaction and reduced home cage activity during their active phase. However, they showed normal weight gain, brain development, and responses to novelty. These results indicate that monoamine dysregulation in Pou3f2⊿ mice may specifically affect basal activity and social development, without altering non-social motivation.


Assuntos
Comportamento Animal/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Fatores do Domínio POU/fisiologia , Comportamento Social , Animais , Monoaminas Biogênicas/fisiologia , Encéfalo/crescimento & desenvolvimento , Comportamento Exploratório/fisiologia , Hipotálamo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neocórtex/fisiologia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Neurônios/fisiologia , Fatores do Domínio POU/química , Fatores do Domínio POU/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Aumento de Peso
2.
Int J Pediatr Otorhinolaryngol ; 129: 109790, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31786483

RESUMO

In this report, we describe a novel, probably pathogenic hemizygous variant c.870G > T (p.Lys290Asn) in the POU3F4 gene in two deaf brothers from one Italian family with identical inner ear abnormalities specific to X-linked deafness-2 (DFNX2). In addition, we performed homology modeling to predict the effect of the missense variant on the protein structure showing a possible disruption of the normal folding. The identification of pathogenic variants causing X-linked recessive deafness will improve molecular diagnosis, genetic counseling, and knowledge of the molecular epidemiology of hearing loss among Italian individuals. Taken together, we recommend preoperative gene mutation analysis in patients who have DFNX2 diagnosed on the basis of characteristic radiological findings, in order to provide with better prognostic information, the risk of recurrence, and improved rehabilitation options. Finally, the present work strengthens the hypothesis that DFNX-2 could be considered as a syndromic deafness, since mixed hearing loss is associated with other dysfunctions of the neuropsychological profile of the patients.


Assuntos
Surdez/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Perda Auditiva Condutiva/genética , Perda Auditiva Neurossensorial/genética , Fatores do Domínio POU/genética , Pré-Escolar , Feminino , Humanos , Lactente , Itália , Masculino , Modelos Moleculares , Mutação de Sentido Incorreto , Fatores do Domínio POU/química , Linhagem , Dobramento de Proteína , Irmãos
3.
Am J Hum Genet ; 105(2): 403-412, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31303265

RESUMO

POU3F3, also referred to as Brain-1, is a well-known transcription factor involved in the development of the central nervous system, but it has not previously been associated with a neurodevelopmental disorder. Here, we report the identification of 19 individuals with heterozygous POU3F3 disruptions, most of which are de novo variants. All individuals had developmental delays and/or intellectual disability and impairments in speech and language skills. Thirteen individuals had characteristic low-set, prominent, and/or cupped ears. Brain abnormalities were observed in seven of eleven MRI reports. POU3F3 is an intronless gene, insensitive to nonsense-mediated decay, and 13 individuals carried protein-truncating variants. All truncating variants that we tested in cellular models led to aberrant subcellular localization of the encoded protein. Luciferase assays demonstrated negative effects of these alleles on transcriptional activation of a reporter with a FOXP2-derived binding motif. In addition to the loss-of-function variants, five individuals had missense variants that clustered at specific positions within the functional domains, and one small in-frame deletion was identified. Two missense variants showed reduced transactivation capacity in our assays, whereas one variant displayed gain-of-function effects, suggesting a distinct pathophysiological mechanism. In bioluminescence resonance energy transfer (BRET) interaction assays, all the truncated POU3F3 versions that we tested had significantly impaired dimerization capacities, whereas all missense variants showed unaffected dimerization with wild-type POU3F3. Taken together, our identification and functional cell-based analyses of pathogenic variants in POU3F3, coupled with a clinical characterization, implicate disruptions of this gene in a characteristic neurodevelopmental disorder.


Assuntos
Regulação da Expressão Gênica , Mutação , Transtornos do Neurodesenvolvimento/etiologia , Fatores do Domínio POU/genética , Ativação Transcricional , Sequência de Aminoácidos , Criança , Feminino , Estudos de Associação Genética , Genótipo , Humanos , Masculino , Transtornos do Neurodesenvolvimento/patologia , Fatores do Domínio POU/química , Conformação Proteica , Homologia de Sequência
4.
Anticancer Agents Med Chem ; 19(3): 389-401, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30417795

RESUMO

BACKGROUND: BRN2 transcription factor is associated with the development of malignant melanoma. The cytotoxic activities and cell death mechanism against B16F10-Nex2 cells were determined with synthetic peptide R18H derived from the POU domain of the BRN2 transcription factor. OBJECTIVE: To determine the cell death mechanisms and in vivo activity of peptide R18H derived from the POU domain of the BRN2 transcription factor against B16F10-Nex2 cells. METHODS: Cell viability was determined by the MTT method. C57Bl/6 mice were challenged with B16F10-Nex2 cells and treated with R18H. To identify the type of cell death, we used TUNEL assay, Annexin V and PI, Hoechst, DHE, and determination of caspase activation and cytochrome c release. Transmission electron microscopy was performed to verify morphological alterations after peptide treatment. RESULTS: Peptide R18H displayed antitumor activity in the first hours of treatment and the EC50% was calculated for 2 and 24h, being 0.76 ± 0.045 mM and 0.559 ± 0.053 mM, respectively. After 24h apoptosis was evident, based on DNA degradation, chromatin condensation, increase of superoxide anion production, phosphatidylserine translocation, activation of caspases 3 and 8, and release of extracellular cytochrome c in B16F10-Nex2 cells. The peptide cytotoxic activity was not affected by necroptosis inhibitors and treated cells did not release LDH in the extracellular medium. Moreover, in vivo antitumor activity was observed following treatment with peptide R18H. CONCLUSION: Peptide R18H from BRN2 transcription factor induced apoptosis in B16F10-Nex2 and displayed antitumor activity in vivo.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas de Homeodomínio/química , Melanoma/tratamento farmacológico , Melanoma/patologia , Fatores do Domínio POU/química , Peptídeos/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Estrutura Molecular , Peptídeos/síntese química , Peptídeos/química , Relação Estrutura-Atividade
5.
Biomed Mater ; 13(1): 015022, 2017 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-28855426

RESUMO

Direct reprogramming of other somatic cells into neurons is an alternative strategy for the recovery of an injured nervous system. In this work, we developed a new non-viral gene carrier based on Porphyra yezoensis polysaccharide (PYP). After modification with ethylenediamine, the cationized PYP (Ed-PYP) was combined with plasmids encoding Ascl1, Brn2 and FoxA1 to form spherical nanoscale particles (Ed-PYP-pABF nanoparticles). Cytotoxicity assays proved that Ed-PYP-pABF nanoparticles had a better safety profile than Lipofectamine 2000 and polyetherimide. Characterization tests illustrated that the Ed-PYP-pABF nanoparticles at an Ed-PYP:pABF weight ratio of 40:1 is a potential candidate for gene delivery, which was further supported by Western blot and plasmid encoding enhanced green fluorescence protein transfection. Based on this transfection strategy, we co-delivered pABF to 3T6 cells using Ed-PYP. ELISA indicated that the levels of brain-derived neurotrophic factor, nerve growth factors and sonic hedgehog reached a maximum at 14 days after the last transfection. Immunofluorescence and Western blot further exhibited positive expression of neurofilament 200, Nestin, glial fibrillary acidic protein, growth associated protein-43, ß-3tubulin, and microtubule associated protein 2, proving the successful conversion of 3T6 cells into neurons. Taken together, these results illustrated that a natural polysaccharide-based gene co-delivery system is a promising strategy for neural reprogramming.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Fibroblastos/citologia , Técnicas de Transferência de Genes , Fator 3-alfa Nuclear de Hepatócito/química , Proteínas do Tecido Nervoso/química , Neurônios/citologia , Fatores do Domínio POU/química , Polissacarídeos/química , Animais , Cátions , Diferenciação Celular , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Camundongos , Nanopartículas/química , Polímeros/química , Porphyra
6.
Development ; 144(17): 3145-3155, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28760809

RESUMO

Grainy head (Grh) is a conserved transcription factor (TF) controlling epithelial differentiation and regeneration. To elucidate Grh functions we identified embryonic Grh targets by ChIP-seq and gene expression analysis. We show that Grh controls hundreds of target genes. Repression or activation correlates with the distance of Grh-binding sites to the transcription start sites of its targets. Analysis of 54 Grh-responsive enhancers during development and upon wounding suggests cooperation with distinct TFs in different contexts. In the airways, Grh-repressed genes encode key TFs involved in branching and cell differentiation. Reduction of the POU domain TF Ventral veins lacking (Vvl) largely ameliorates the airway morphogenesis defects of grh mutants. Vvl and Grh proteins additionally interact with each other and regulate a set of common enhancers during epithelial morphogenesis. We conclude that Grh and Vvl participate in a regulatory network controlling epithelial maturation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Genoma de Inseto , Fatores do Domínio POU/química , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Embrião não Mamífero/metabolismo , Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Imunidade Inata/genética , Morfogênese/genética , Especificidade de Órgãos/genética , Fatores do Domínio POU/metabolismo , Ligação Proteica , Domínios Proteicos , Sistema Respiratório/metabolismo , Elementos de Resposta/genética
7.
EMBO Rep ; 18(2): 319-333, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28007765

RESUMO

The transcription factor Oct4 is a core component of molecular cocktails inducing pluripotent stem cells (iPSCs), while other members of the POU family cannot replace Oct4 with comparable efficiency. Rather, group III POU factors such as Oct6 induce neural lineages. Here, we sought to identify molecular features determining the differential DNA-binding and reprogramming activity of Oct4 and Oct6. In enhancers of pluripotency genes, Oct4 cooperates with Sox2 on heterodimeric SoxOct elements. By re-analyzing ChIP-Seq data and performing dimerization assays, we found that Oct6 homodimerizes on palindromic OctOct more cooperatively and more stably than Oct4. Using structural and biochemical analyses, we identified a single amino acid directing binding to the respective DNA elements. A change in this amino acid decreases the ability of Oct4 to generate iPSCs, while the reverse mutation in Oct6 does not augment its reprogramming activity. Yet, with two additional amino acid exchanges, Oct6 acquires the ability to generate iPSCs and maintain pluripotency. Together, we demonstrate that cell type-specific POU factor function is determined by select residues that affect DNA-dependent dimerization.


Assuntos
Transdiferenciação Celular/genética , Reprogramação Celular/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Fatores do Domínio POU/química , Fatores do Domínio POU/metabolismo , Multimerização Proteica , Substituição de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Células-Tronco Embrionárias , Elementos Facilitadores Genéticos , Epigênese Genética , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Modelos Moleculares , Motivos de Nucleotídeos , Fatores de Transcrição de Octâmero/química , Fatores de Transcrição de Octâmero/genética , Fatores de Transcrição de Octâmero/metabolismo , Fatores do Domínio POU/genética , Regiões Promotoras Genéticas , Ligação Proteica , Conformação Proteica , Estabilidade Proteica , Transcriptoma
8.
Nucleic Acids Res ; 45(2): 832-845, 2017 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-27915232

RESUMO

Cooperative binding of transcription factors is known to be important in the regulation of gene expression programs conferring cellular identities. However, current methods to measure cooperativity parameters have been laborious and therefore limited to studying only a few sequence variants at a time. We developed Coop-seq (cooperativity by sequencing) that is capable of efficiently and accurately determining the cooperativity parameters for hundreds of different DNA sequences in a single experiment. We apply Coop-seq to 12 dimer pairs from the Sox and POU families of transcription factors using 324 unique sequences with changed half-site orientation, altered spacing and discrete randomization within the binding elements. The study reveals specific dimerization profiles of different Sox factors with Oct4. By contrast, Oct4 and the three neural class III POU factors Brn2, Brn4 and Oct6 assemble with Sox2 in a surprisingly indistinguishable manner. Two novel half-site configurations can support functional Sox/Oct dimerization in addition to known composite motifs. Moreover, Coop-seq uncovers a nucleotide switch within the POU half-site when spacing is altered, which is mirrored in genomic loci bound by Sox2/Oct4 complexes.


Assuntos
Fatores do Domínio POU/metabolismo , Fatores de Transcrição SOX/metabolismo , Animais , DNA/química , DNA/metabolismo , Camundongos , Modelos Moleculares , Fator 3 de Transcrição de Octâmero/química , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores do Domínio POU/química , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Fatores de Transcrição SOX/química , Fatores de Transcrição SOXB1/química , Fatores de Transcrição SOXB1/metabolismo
9.
J Mol Model ; 22(9): 228, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27568376

RESUMO

The incidence of skin cancer has increased in recent decades, and melanoma is the most aggressive form with the lowest chance of successful treatment. Currently, drug design projects are in progress, but available treatments against metastatic melanoma have not significantly increased survival, and few patients are cured. Thus, new therapeutic agents should be developed as more effective therapeutic options for melanoma. High levels of the BRN2 transcription factor have been related to melanoma development. However, neither the three-dimensional (3D) structure of BRN2 protein nor its POU domain has been determined experimentally. Construction of the BRN2 3D structure, and the study of its interaction with its DNA target, are important strategies for increasing the structural and functional knowledge of this protein. Thus, the aim of this work was to study the interaction between BRN2 and MORE DNA through in silico methods. The full-length BRN2 3D structure was built using the PHYRE2 and Swiss-Model programs, and molecular dynamics of this protein in complex with MORE DNA was simulated for 20 ns by the NAMD program. The BRN2 model obtained includes helix and loop regions, and the BRN2 POU domain shares structural similarity with other members of the transcription factor family. No significant conformational change of this protein occurred during dynamics simulation. These analyses revealed BRN2 residues important for the specific interaction with nucleotide bases and with more than one DNA nucleotide. This study may contribute to the design of inhibitors against BRN2 or MORE DNA as molecular targets of melanoma skin cancer. Graphical Abstract Model of complete Brn2 protein in complex with MORE DNA after building through comparative modeling and refinement by molecular dynamics simulation.


Assuntos
DNA/química , Proteínas de Homeodomínio/química , Melanoma/fisiopatologia , Fatores do Domínio POU/química , Simulação por Computador , Humanos
10.
Int J Biol Macromol ; 86: 701-8, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26854886

RESUMO

POU-M2 is a homeodomain transcription factor which plays important roles in the development and silk synthesis of Bombyx mori. In this study, we expressed, purified and characterized POU-M2 and studied its transcription regulation on fibroin heavy chain gene of Bombyx mori. Gel filtration showed POU-M2 existed as a dimer in solution. Far-UV circular dichroism spectra indicated POU-M2 had a well-defined α-helix structure and the α-helix content was about 26.4%. The thermal unfolding transition of POU-M2 was a cooperative process. Tm, ΔH and ΔS were 45.15 ± 0.2 °C, 138.4 ± 0.5 KJ/mol and 0.4349 ± 0.04 KJ/(mol·K), respectively. Western blotting analysis indicated the expression level of POU-M2 increased slightly from day 3 to day 7 of the fifth instar larvae in the posterior silk gland. POU-M2 was positioned in the nucleus of cells. The luciferase reporter assay demonstrated POU-M2 could stimulate the promoter activity of fibroin heavy chain gene, and the activation effect was dependent on the amount of POU-M2. Our study suggested POU-M2 may be involved in the transcriptional regulation of fibroin heavy chain gene. These findings expand toward a better understanding of the structure of POU-M2 and its function in silk synthesis of Bombyx mori.


Assuntos
Bombyx/metabolismo , Proteínas de Insetos/metabolismo , Fatores do Domínio POU/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Bombyx/genética , Biologia Computacional , Fibroínas/genética , Regulação da Expressão Gênica , Proteínas de Insetos/química , Proteínas de Insetos/genética , Fatores do Domínio POU/química , Fatores do Domínio POU/genética , Regiões Promotoras Genéticas/genética , Estrutura Secundária de Proteína , Seda/biossíntese
11.
BMC Genomics ; 16: 700, 2015 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-26377945

RESUMO

BACKGROUND: One of the major challenges in developmental biology is to understand the regulatory events that generate neuronal diversity. During Drosophila embryonic neural lineage development, cellular temporal identity is established in part by a transcription factor (TF) regulatory network that mediates a cascade of cellular identity decisions. Two of the regulators essential to this network are the POU-domain TFs Nubbin and Pdm-2, encoded by adjacent genes collectively known as pdm. The focus of this study is the discovery and characterization of cis-regulatory DNA that governs their expression. RESULTS: Phylogenetic footprinting analysis of a 125 kb genomic region that spans the pdm locus identified 116 conserved sequence clusters. To determine which of these regions function as cis-regulatory enhancers that regulate the dynamics of pdm gene expression, we tested each for in vivo enhancer activity during embryonic development and postembryonic neurogenesis. Our screen revealed 77 unique enhancers positioned throughout the noncoding region of the pdm locus. Many of these activated neural-specific gene expression during different developmental stages and many drove expression in overlapping patterns. Sequence comparisons of functionally related enhancers that activate overlapping expression patterns revealed that they share conserved elements that can be predictive of enhancer behavior. To facilitate data accessibility, the results of our analysis are catalogued in cisPatterns, an online database of the structure and function of these and other Drosophila enhancers. CONCLUSIONS: These studies reveal a diversity of modular enhancers that most likely regulate pdm gene expression during embryonic and adult development, highlighting a high level of temporal and spatial expression specificity. In addition, we discovered clusters of functionally related enhancers throughout the pdm locus. A subset of these enhancers share conserved elements including sequences that correspond to known TF DNA binding sites. Although comparative analysis of the nubbin and pdm-2 encoding sequences indicate that these two genes most likely arose from a duplication event, we found only partial evidence of sequence duplication between their enhancers, suggesting that after the putative duplication their cis-regulatory DNA diverged at a higher rate than their coding sequences.


Assuntos
Proteínas de Drosophila/genética , Drosophila/genética , Elementos Facilitadores Genéticos , Loci Gênicos , Proteínas de Homeodomínio/genética , Neurogênese/genética , Fatores do Domínio POU/genética , Animais , Sequência de Bases , Encéfalo/metabolismo , Análise por Conglomerados , Sequência Conservada , Proteínas de Drosophila/química , Regulação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Ordem dos Genes , Genes Reporter , Proteínas de Homeodomínio/química , Dados de Sequência Molecular , Especificidade de Órgãos/genética , Fatores do Domínio POU/química , Alinhamento de Sequência , Análise de Sequência de DNA , Transgenes
12.
BMC Med Genet ; 16: 9, 2015 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-25928534

RESUMO

BACKGROUND: Mutations in the POU3F4 gene cause X-linked deafness type 3 (DFN3), which is characterized by inner ear anomalies. METHODS: Three Turkish, one Ecuadorian, and one Nigerian families were included based on either inner ear anomalies detected in probands or X-linked family histories. Exome sequencing and/or Sanger sequencing were performed in order to identify the causative DNA variants in these families. RESULTS: Four novel, c.707A>C (p.(Glu236Ala)), c.772delG (p.(Glu258ArgfsX30)), c.902C>T (p.(Pro301Leu)), c.987T>C (p.(Ile308Thr)), and one previously reported mutation c.346delG (p.(Ala116ProfsX26)) in POU3F4, were identified. All mutations identified are predicted to affect the POU-specific or POU homeo domains of the protein and co-segregated with deafness in all families. CONCLUSIONS: Expanding the spectrum of POU3F4 mutations in different populations along with their associated phenotypes provides better understanding of their clinical importance and will be helpful in clinical evaluation and counseling of the affected individuals.


Assuntos
Surdez/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Fatores do Domínio POU/química , Fatores do Domínio POU/genética , Pré-Escolar , Estudos de Coortes , Surdez/etnologia , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/etnologia , Humanos , Masculino , Modelos Moleculares , Linhagem , Estrutura Terciária de Proteína
13.
Mol Biol Evol ; 31(12): 3136-47, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25261405

RESUMO

The POU genes represent a diverse class of animal-specific transcription factors that play important roles in neurogenesis, pluripotency, and cell-type specification. Although previous attempts have been made to reconstruct the evolution of the POU class, these studies have been limited by a small number of representative taxa, and a lack of sequences from basally branching organisms. In this study, we performed comparative analyses on available genomes and sequences recovered through "gene fishing" to better resolve the topology of the POU gene tree. We then used ancestral state reconstruction to map the most likely changes in amino acid evolution for the conserved domains. Our work suggests that four of the six POU families evolved before the last common ancestor of living animals-doubling previous estimates-and were followed by extensive clade-specific gene loss. Amino acid changes are distributed unequally across the gene tree, consistent with a neofunctionalization model of protein evolution. We consider our results in the context of early animal evolution, and the role of POU5 genes in maintaining stem cell pluripotency.


Assuntos
Fatores do Domínio POU/genética , Animais , Teorema de Bayes , Sequência Consenso , Evolução Molecular , Funções Verossimilhança , Modelos Moleculares , Dados de Sequência Molecular , Fatores do Domínio POU/química , Filogenia , Estrutura Terciária de Proteína
14.
Comput Math Methods Med ; 2013: 854710, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24324527

RESUMO

Biomolecular cooperativity is of great scientific interest due to its role in biological processes. Two transcription factors (TFs), Oct-4 and Sox-2, are crucial in transcriptional regulation of embryonic stem cells. In this paper, we analyze how Oct-1 (a similar POU factor) and Sox-2, interact cooperatively at their enhancer binding sites in collective motions. Normal mode analysis (NMA) is implemented to study the collective motions of two complexes with each involving these TFs and an enhancer. The special structure of Oct proteins is analyzed comprehensively, after which each Oct/Sox group is reassembled into two protein pairs. We subsequently propose a segmentation idea to extract the most correlated segments in each pair, using correlations of motion magnitude curves. The median analysis on these correlation values shows the intimacy of subunit POUS (Oct-1) and Sox-2. Using those larger-than-median correlation values, we conduct statistical studies and propose several protein-protein cooperative modes (S and D) coupled with their subtypes. Additional filters are applied and similar results are obtained. A supplementary study on the rotation angle curves reaches an agreement with these modes. Overall, these proposed cooperative modes provide useful information for us to understand the complicated interaction mechanism in the POU/HMG/DNA complexes.


Assuntos
Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Animais , Simulação por Computador , DNA/química , DNA/genética , DNA/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Elementos Facilitadores Genéticos , Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Grupo de Alta Mobilidade/metabolismo , Camundongos , Modelos Moleculares , Simulação de Dinâmica Molecular , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Fatores do Domínio POU/química , Fatores do Domínio POU/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas
15.
Mol Biol Evol ; 30(4): 844-52, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23197593

RESUMO

The FOXP2 gene is required for normal development of speech and language. By isolating and sequencing FOXP2 genomic DNA fragments from a 49,000-year-old Iberian Neandertal and 50 present-day humans, we have identified substitutions in the gene shared by all or nearly all present-day humans but absent or polymorphic in Neandertals. One such substitution is localized in intron 8 and affects a binding site for the transcription factor POU3F2, which is highly conserved among vertebrates. We find that the derived allele of this site is less efficient than the ancestral allele in activating transcription from a reporter construct. The derived allele also binds less POU3F2 dimers than POU3F2 monomers compared with the ancestral allele. Because the substitution in the POU3F2 binding site is likely to alter the regulation of FOXP2 expression, and because it is localized in a region of the gene associated with a previously described signal of positive selection, it is a plausible candidate for having caused a recent selective sweep in the FOXP2 gene.


Assuntos
Evolução Molecular , Fatores de Transcrição Forkhead/genética , Elementos Reguladores de Transcrição , Animais , Sequência de Bases , Sítios de Ligação , Sequência Conservada , Fatores de Transcrição Forkhead/metabolismo , Frequência do Gene , Células HeLa , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Humanos , Íntrons , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Homem de Neandertal/genética , Fatores do Domínio POU/química , Fatores do Domínio POU/metabolismo , Análise de Sequência de DNA , Ativação Transcricional
16.
PLoS One ; 7(4): e36420, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22558464

RESUMO

Formation of all metazoan bodies is controlled by a group of selector genes including homeobox genes, highly conserved across the entire animal kingdom. The homeobox genes from Pou and Six classes are key members of the regulation cascades determining development of sensory organs, nervous system, gonads and muscles. Besides using common bilaterian models, more attention has recently been targeted at the identification and characterization of these genes within the basal metazoan phyla. Cnidaria as a diploblastic sister group to bilateria with simple and yet specialized organs are suitable models for studies on the sensory organ origin and the associated role of homeobox genes. In this work, Pou and Six homeobox genes, together with a broad range of other sensory-specific transcription factors, were identified in the transcriptome of hydrozoan jellyfish Craspedacusta sowerbyi. Phylogenetic analyses of Pou and Six proteins revealed cnidarian-specific sequence motifs and contributed to the classification of individual factors. The majority of the Craspedacusta sowerbyi Pou and Six homeobox genes are predominantly expressed in statocysts, manubrium and nerve ring, the tissues with sensory and nervous activities. The described diversity and expression patterns of Pou and Six factors in hydrozoan jellyfish highlight their evolutionarily conserved functions. This study extends the knowledge of the cnidarian genome complexity and shows that the transcriptome of hydrozoan jellyfish is generally rich in homeodomain transcription factors employed in the regulation of sensory and nervous functions.


Assuntos
Variação Genética , Hidrozoários/genética , Fatores do Domínio POU/genética , Filogenia , Transcriptoma , Animais , Evolução Molecular , Feminino , Especificidade de Órgãos , Fatores do Domínio POU/química , Fatores do Domínio POU/metabolismo , Estrutura Terciária de Proteína
17.
J Neurochem ; 120(6): 899-912, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22225570

RESUMO

Aquaporin-4, a predominant water channel in the brain, is specifically expressed in astrocyte endfeet and plays a central role in water homeostasis, neuronal activity, and cell migration in the brain. It has two dominant isoforms called M1 and M23, whose mRNA is driven by distinct promoters located upstream of exons 0 and 1 of the aquaporin-4 gene, respectively. To identify cis-acting elements responsible for the astrocyte-specific transcription of M1 mRNA, the promoter activity of the 5'-flanking region upstream of exon 0 in primary cultured mouse astrocytes was examined by luciferase assay, and sequences, where nuclear factors bind, were identified by electrophoretic mobility shift assay. An astrocyte-specific activity enhancing transcription from the M1 promoter was observed within ∼2 kb from the transcriptional start sites of M1 mRNA. At least five elements clustered within the 286-bp region were found to function as a novel astrocyte-specific enhancer. Among the five elements, a consensus sequence of Pit-1/Oct/Unc-86 (POU) transcription factors was indispensable to the astrocyte-specific enhancer since disruption of the POU motif completely abolished the enhancer activity in astrocytes. However, the POU motif alone had little activity, indicating the requirement for cooperation with other upstream elements to exert full enhancer activity.


Assuntos
Aquaporina 4/genética , Sequência Consenso/fisiologia , Elementos Facilitadores Genéticos/fisiologia , Fatores do Domínio POU/química , Animais , Aquaporina 4/química , Astrócitos , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Éxons/fisiologia , Camundongos , Dados de Sequência Molecular , Fatores do Domínio POU/genética , Transfecção
18.
Pigment Cell Melanoma Res ; 23(3): 404-18, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20337985

RESUMO

POU3F2 is a POU-Homeodomain transcription factor expressed in neurons and melanoma cells. In melanoma lesions, cells expressing high levels of POU3F2 show enhanced invasive and metastatic capacity that can in part be explained by repression of Micropthalmia-associated Transcription Factor (MITF) expression via POU3F2 binding to its promoter. To identify other POU3F2 target genes that may be involved in modulating the properties of melanoma cells, we performed ChIP-chip experiments in 501Mel melanoma cells. 2108 binding loci located in the regulatory regions of 1700 potential target genes were identified. Bioinformatic and experimental assays showed the presence of known POU3F2-binding motifs, but also many AT-rich sequences with only partial similarity to the known motifs at the occupied loci. Functional analysis indicates that POU3F2 regulates the stem cell factor (Kit ligand, Kitl) promoter via a cluster of four closely spaced binding sites located in the proximal promoter. Our results suggest that POU3F2 may regulate the properties of melanoma cells via autocrine KIT ligand signalling.


Assuntos
Regulação Neoplásica da Expressão Gênica , Genoma Humano/genética , Estudo de Associação Genômica Ampla , Proteínas de Homeodomínio/genética , Melanoma/genética , Fatores do Domínio POU/genética , Regiões Promotoras Genéticas , Fator de Células-Tronco/genética , Motivos de Aminoácidos , Sequência de Bases , Sítios de Ligação , Antígenos CD36/genética , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Genes Neoplásicos/genética , Loci Gênicos/genética , Proteínas de Homeodomínio/química , Humanos , Melanoma/patologia , Fator de Transcrição Associado à Microftalmia/genética , Dados de Sequência Molecular , Fatores do Domínio POU/química , Ligação Proteica , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Ativação Transcricional/genética , Proteínas Wnt/genética
19.
J Biol Chem ; 285(11): 8408-21, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20064932

RESUMO

Xenopus Oct25 is a POU family subclass V (POU-V) transcription factor with a distinct domain structure. To investigate the contribution of different domains to the function of Oct25, we have performed gain of function analyses. Deletions of the N- or C-terminal regions and of the Hox domain (except its nuclear localization signal) result in mutants being indistinguishable from the wild type protein in the suppression of genes promoting germ layer formation. Deletion of the complete POU domain generates a mutant that has no effect on embryogenesis. However, disruption of the alpha-helical structures in the POU domain, even by a single amino acid mutation, causes reversal of protein function. Overexpression of such mutants leads to dorsalization of embryos and formation of secondary axial structures. The underlying mechanism is an enhanced transcription of genes coding for antagonists of the ligands for ventralizing bone morphogenetic protein and Wnt pathways. Corresponding deletion mutants of Xenopus Oct60, Oct91, or mouse Oct4 also exhibit such a dominant-negative effect. Therefore, our results reveal that the integrity of the POU domain is crucial for the function of POU-V transcription factors in the regulation of genes that promote germ layer formation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Fatores do Domínio POU , Proteínas de Xenopus , Xenopus/genética , Ativinas/metabolismo , Animais , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Células Cultivadas , Deleção de Genes , Humanos , Rim/citologia , Camundongos , Mutagênese/fisiologia , Proteína Nodal/metabolismo , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Fator 6 de Transcrição de Octâmero/metabolismo , Fatores do Domínio POU/química , Fatores do Domínio POU/genética , Fatores do Domínio POU/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Transdução de Sinais/fisiologia , Proteínas com Domínio T/metabolismo , Transcrição Gênica/fisiologia , Xenopus/embriologia , Proteínas de Xenopus/química , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , beta Catenina/metabolismo
20.
J Genet Genomics ; 37(12): 787-93, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21193157

RESUMO

We present the clinical and genetic findings for a Chinese family with X-linked non-syndromic hearing loss in which the affected males showed congenital profound sensorineural hearing impairment. In two affected brothers, the computer tomography of temporal bone showed bilateral dilation of the internal auditory canal with fistulous communication between the lateral canal and the basal cochlear turn, which is consistent with the typical DFNX2 phenotype. A missense mutation (c.647G→A) in the POU3F4 gene caused a substitution from glycine to glutamic acid at position 216 (p.G216E), and this mutation was found to consistently cosegregate with the deafness phenotype in the family. The mutation resulted in the loss of function of the POU3F4 by decreasing the affinity between the protein and DNA, as shown in silico by the structural analysis. Prenatal diagnosis of pregnant proband of this family revealed the c.647G→A mutation in DNA extracted from the amniotic fluid surrounding the fetus. The appropriate use of genetic testing and prenatal diagnosis plays a key role in reducing the recurrence of genetic defects in high-risk families.


Assuntos
Doenças Genéticas Ligadas ao Cromossomo X/genética , Perda Auditiva Neurossensorial/genética , Fatores do Domínio POU/genética , Povo Asiático/genética , DNA/genética , DNA/isolamento & purificação , Orelha Interna/diagnóstico por imagem , Orelha Interna/patologia , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/diagnóstico , Ácido Glutâmico/genética , Glicina/genética , Perda Auditiva Neurossensorial/diagnóstico , Humanos , Masculino , Modelos Moleculares , Mutação de Sentido Incorreto , Fatores do Domínio POU/química , Linhagem , Gravidez , Diagnóstico Pré-Natal , Osso Temporal/diagnóstico por imagem , Tomografia Computadorizada por Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA