Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Differentiation ; 115: 30-36, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32858261

RESUMO

Forkhead box (FOX) family transcription factors play essential roles in development, tissue homeostasis, and disease. Although the biology of several FOX proteins has been studied in depth, it is unclear to what extent these findings apply to even closely related family members, which frequently exert overlapping but non-redundant functions. To help address this question, we have generated a uniform, ready-to-use expression library of all 44 human FOX transcription factors with a convenient peptide tag for parallel screening assays. In addition, we have generated multiple universal forkhead box reporter plasmids, which can be used to monitor the transcriptional activity of most FOX proteins with high fidelity. As a proof-of-principle, we use our plasmid library to identify the DNA repair protein XRCC6/Ku70 as a selective FOX interaction partner and regulator of FOX transcriptional activity. We believe that these tools, which we make available via the Addgene plasmid repository, will considerably expedite the investigation of FOX protein biology.


Assuntos
Reparo do DNA/genética , Fatores de Transcrição Forkhead/genética , Homeostase/genética , Autoantígeno Ku/genética , Animais , Fatores de Transcrição Forkhead/classificação , Regulação da Expressão Gênica/genética , Células HCT116 , Humanos
2.
Viral Immunol ; 33(10): 652-655, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32634048

RESUMO

Regulatory T cell (Treg) lineage plays a central role in inflammation and autoimmunity control. Interleukin-10 (IL-10) has been described as a pleiotropic cytokine that is mainly released by CD4+ CD25+ FOXP3+ Treg cells and has a potent immunosuppressive activity. Forkhead box P3 (FOXP3) transcription factor expression is crucial for Treg to function as a suppressor cell, and FOXP3 gene single nucleotide variants (SNVs) have already been shown to influence on viral pathogenesis. This study was conducted to evaluate the plasmatic and cervical levels of IL-10 in human papillomavirus-infected and uninfected patients and investigate whether the FOXP3 intron -1 SNVs rs3761548 and rs2232365 might alter IL-10 secretion. SNVs were genotyped by the characterization of polymerase chain reaction (PCR) products based on sequence-specific enzymatic cleavage using restriction fragment length polymorphism (RFLP) method. IL-10 levels were determined by quantitative enzyme-linked immunosorbent assay (ELISA). In conclusion, the data indicate that there is no association between FOXP3 SNVs and circulating and cervical IL-10 levels. This finding provides a rationale that IL-10 gene activation is independent of FOXP3 transcription factor activities on Treg cells.


Assuntos
Fatores de Transcrição Forkhead/genética , Interleucina-10/análise , Interleucina-10/sangue , Polimorfismo de Nucleotídeo Único , Estudos de Casos e Controles , Feminino , Fatores de Transcrição Forkhead/classificação , Predisposição Genética para Doença , Genótipo , Humanos , Linfócitos T Reguladores/imunologia
3.
Int J Mol Sci ; 21(3)2020 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-31973091

RESUMO

Forkhead box class O family member proteins (FoxOs) are evolutionarily conserved transcription factors for their highly conserved DNA-binding domain. In mammalian species, all the four FoxO members, FoxO1, FoxO3, FoxO4, and FoxO6, are expressed in different organs. In bone, the first three members are extensively expressed and more studied. Bone development, remodeling, and homeostasis are all regulated by multiple cell lineages, including osteoprogenitor cells, chondrocytes, osteoblasts, osteocytes, osteoclast progenitors, osteoclasts, and the intercellular signaling among these bone cells. The disordered FoxOs function in these bone cells contribute to osteoarthritis, osteoporosis, or other bone diseases. Here, we review the current literature of FoxOs for their roles in bone cells, focusing on helping researchers to develop new therapeutic approaches and prevent or treat the related bone diseases.


Assuntos
Osso e Ossos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Osteócitos/metabolismo , Fatores de Transcrição/metabolismo , Doenças Ósseas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem da Célula , Condrogênese/fisiologia , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O3/metabolismo , Fatores de Transcrição Forkhead/classificação , Fatores de Transcrição Forkhead/genética , Células-Tronco Hematopoéticas , Osteoartrite/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese/fisiologia , Osteoporose/metabolismo , Transdução de Sinais
4.
Dev Comp Immunol ; 97: 38-44, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30905685

RESUMO

FOXO proteins are a subgroup of the forkhead family of transcription factors that play crucial roles in lifespan regulation. In addition, FOXO proteins are also involved in immune responses. After a systematic study of FOXO genes in channel catfish, Ictalurus punctatus, seven FOXO genes were identified and characterized, including FOXO1a, FOXO1b, FOXO3a, FOXO3b, FOXO4, FOXO6a and FOXO6b. Through phylogenetic and syntenic analyses, it was found that FOXO1, FOXO3 and FOXO6 were duplicated in the catfish genome, as in the zebrafish genome. Analysis of the relative rates of nonsynonymous (dN) and synonymous (dS) substitutions revealed that the FOXO genes were globally strongly constrained by negative selection. Differential expression patterns were observed in the majority of FOXO genes after Edwardsiella ictaluri and Flavobacterium columnare infections. After E. ictaluri infection, four FOXO genes with orthologs in mammal species were significantly upregulated, where FOXO6b was the most dramatically upregulated. However, after F. columnare infection, the expression levels of almost all FOXO genes were not significantly affected. These results suggested that either a pathogenesis-specific pattern or tissue-specific pattern existed in catfish after these two bacterial infections. Taken together, these findings indicated that FOXO genes may play important roles in immune responses to bacterial infections in catfish.


Assuntos
Infecções Bacterianas/genética , Doenças dos Peixes/genética , Proteínas de Peixes/genética , Fatores de Transcrição Forkhead/genética , Ictaluridae/genética , Família Multigênica , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Edwardsiella ictaluri/imunologia , Edwardsiella ictaluri/fisiologia , Doenças dos Peixes/imunologia , Doenças dos Peixes/microbiologia , Proteínas de Peixes/classificação , Proteínas de Peixes/imunologia , Flavobacterium/imunologia , Flavobacterium/fisiologia , Fatores de Transcrição Forkhead/classificação , Fatores de Transcrição Forkhead/imunologia , Perfilação da Expressão Gênica/métodos , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Ictaluridae/imunologia , Ictaluridae/microbiologia , Filogenia
5.
Int J Biol Sci ; 13(7): 815-827, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28808415

RESUMO

Many transcription factors play a key role in cellular differentiation and the delineation of cell phenotype. Transcription factors are regulated by phosphorylation, ubiquitination, acetylation/deacetylation and interactions between two or more proteins controlling multiple signaling pathways. These pathways regulate different physiological processes and pathological events, such as cancer and other diseases. The Forkhead box O (FOXO) is one subfamily of the fork head transcription factor family with important roles in cell fate decisions and this subfamily is also suggested to play a pivotal functional role as a tumor suppressor in a wide range of cancers. During apoptosis, FOXOs are involved in mitochondria-dependent and -independent processes triggering the expression of death receptor ligands like Fas ligand, TNF apoptosis ligand and Bcl­XL, bNIP3, Bim from Bcl-2 family members. Different types of growth factors like insulin play a vital role in the regulation of FOXOs. The most important pathway interacting with FOXO in different types of cancers is the PI3K/AKT pathway. Some other important pathways such as the Ras-MEK-ERK, IKK and AMPK pathways are also associated with FOXOs in tumorigenesis. Therapeutically targeting the FOXO signaling pathway(s) could lead to the discovery and development of efficacious agents against some cancers, but this requires an enhanced understanding and knowledge of FOXO transcription factors and their regulation and functioning. This review focused on the current understanding of cell biology of FOXO transcription factors which relates to their potential role as targets for the treatment and prevention of human cancers. We also discuss drugs which are currently being used for cancer treatment along with their target pathways and also point out some potential drawbacks of those drugs, which further signifies the need for development of new drug strategies in the field of cancer treatment.


Assuntos
Fatores de Transcrição Forkhead/classificação , Neoplasias/tratamento farmacológico , Transdução de Sinais/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Família Multigênica , Neoplasias/fisiopatologia
6.
Comput Biol Chem ; 64: 9-18, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27231827

RESUMO

Foxl2 belongs to forkhead/HNF-3-related family of transcription factors which is involved in ovarian differentiation and development. In present study, the Foxl2 mRNA was cloned from ovary of C. batrachus. The full length cDNA sequence of the Foxl2 was 1056bp which consists of 5' (41bp) and 3' (106bp) non-coding regions, as well as a 909bp of open reading frame (ORF) that encodes 302 amino acids. The putative protein was having the theoretical molecular weight (MW) of 34.018kD and a calculated isoelectric point (pI) of 9.38. There were 11 serine (Ser), 5 threonine (Thr), and 5 tyrosine (Tyr) phosphorylation sites and 2 putative N-glycosylation sites on the predicted protein. The ligand binding sites were predicted to be present on amino acids 42, 49, 50, 91, 92 and 95 respectively. The signal peptide analysis predicted that C. batrachus Foxl2 is a non-secretory protein. The hydropathy profile of Foxl2 protein revealed that this protein is hydrophilic in nature. Protein-protein interaction demonstrated that Foxl2 protein chiefly interacts with cytochrome P450 protein family. The mRNA transcript analysis of various tissues indicated that the C. batrachus Foxl2 mRNA was more expressed in the brain, pituitary and ovary in female while, the former two tissues and testis showed low expression in male. This study provides a basis for further structural and functional exploration of the Foxl2 from C. batrachus, including its deduced protein and its signal transduction function.


Assuntos
Peixes-Gato/genética , Biologia Computacional , Fatores de Transcrição Forkhead/genética , Expressão Gênica , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , DNA Complementar/genética , Feminino , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/classificação , Masculino , Filogenia , Homologia de Sequência de Aminoácidos
7.
Proc Natl Acad Sci U S A ; 110(30): 12349-54, 2013 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-23836653

RESUMO

The evolution of transcriptional regulatory networks entails the expansion and diversification of transcription factor (TF) families. The forkhead family of TFs, defined by a highly conserved winged helix DNA-binding domain (DBD), has diverged into dozens of subfamilies in animals, fungi, and related protists. We have used a combination of maximum-likelihood phylogenetic inference and independent, comprehensive functional assays of DNA-binding capacity to explore the evolution of DNA-binding specificity within the forkhead family. We present converging evidence that similar alternative sequence preferences have arisen repeatedly and independently in the course of forkhead evolution. The vast majority of DNA-binding specificity changes we observed are not explained by alterations in the known DNA-contacting amino acid residues conferring specificity for canonical forkhead binding sites. Intriguingly, we have found forkhead DBDs that retain the ability to bind very specifically to two completely distinct DNA sequence motifs. We propose an alternate specificity-determining mechanism whereby conformational rearrangements of the DBD broaden the spectrum of sequence motifs that a TF can recognize. DNA-binding bispecificity suggests a previously undescribed source of modularity and flexibility in gene regulation and may play an important role in the evolution of transcriptional regulatory networks.


Assuntos
Evolução Biológica , DNA/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Acanthamoeba castellanii/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/classificação , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Filogenia , Homologia de Sequência de Aminoácidos
8.
Cytogenet Genome Res ; 136(2): 123-30, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22286236

RESUMO

Using the genome sequence of the medaka, Oryzias latipes, we examined the genomic complement of Fox genes in this organism to gain insight into the evolutionary relationships and expression patterns of this gene family. We identified 31 Fox genes by searching for Forkhead domains in the medaka genome and by polymerase chain reaction (PCR) using primers designed from protein alignments. All the medaka Fox genes are encoded in 18 subclasses as follows: 5 Fox genes in subclass D; 3 Fox genes in subclass O; 2 Fox genes in each of subclass A, B, E, F, G, I, P, and Q, respectively; 1 Fox gene in each of subclass C, H, J, K, M, N, and R. The gene structures and general features are also discussed. In addition, we examined the expression patterns of some of these genes in different adult tissues and during embryonic development in medaka using quantitative PCR and in situ hybridization. The present study shows that Forkhead transcription factors play an important role during early embryonic development in medaka, and the results will enhance our knowledge in terms of Fox family evolution in different taxa.


Assuntos
Proteínas de Peixes/genética , Fatores de Transcrição Forkhead/genética , Família Multigênica , Oryzias/genética , Animais , Mapeamento Cromossômico , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Evolução Molecular , Feminino , Fatores de Transcrição Forkhead/classificação , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hibridização In Situ , Masculino , Dados de Sequência Molecular , Oryzias/embriologia , Oryzias/crescimento & desenvolvimento , Filogenia , Isoformas de Proteínas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Fatores de Tempo
9.
J Neurochem ; 122(1): 72-80, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21985339

RESUMO

FOXP2, a forkhead box-containing transcription factor, forms homo- or hetero-dimers with FOXP family members and localizes to the nucleus, while FOXP2(R553H), which contains a mutation related to speech/language disorders, features reduced DNA binding activity and both cytoplasmic and nuclear localization. In addition to being a loss-of-function mutation, it is possible that FOXP2(R553H) also may act as a gain-of-function mutation to inhibit the functions of FOXP2 isoforms including FOXP2Ex10+ lacking forkhead domain. Foxp2(R552H) knock-in mouse pups exhibit impaired ultrasonic vocalization and poor dendritic development in Purkinje cells. However, expressions of Foxp2 isoforms in the developing Purkinje are unclear. The appearance of 'apical cytoplasmic swelling' (mitochondria-rich regions that are the source of budding processes) correlates with dendritic development of Purkinje cells. In the present study, we focused on Foxp2 isoforms localizing to the apical cytoplasmic swelling and identified two isoforms lacking forkhead domain: Foxp2Ex12+ and Foxp2Ex15. They partly localized to the membrane fraction that includes mitochondria. Foxp2Ex12+ mainly localized to the apical cytoplasmic swelling in early developing Purkinje cells at the stellate stage (P2-P4). Mitochondrial localization of Foxp2Ex12+ in Purkinje cells was confirmed by immune-electron microscopic analysis. Foxp2Ex12+ may play a role in dendritic development in Purkinje cells.


Assuntos
Cerebelo/citologia , Cerebelo/crescimento & desenvolvimento , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Mitocôndrias/metabolismo , Células de Purkinje/ultraestrutura , Proteínas Repressoras/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Arginina/genética , Calbindinas , Citocromos c/metabolismo , Citoplasma/metabolismo , Dendritos/metabolismo , Dendritos/ultraestrutura , Feto , Fatores de Transcrição Forkhead/classificação , Histidina/genética , Humanos , Camundongos , Camundongos Transgênicos , Microscopia Imunoeletrônica , Mutação/genética , Isoformas de Proteínas/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Proteínas Repressoras/classificação , Proteína G de Ligação ao Cálcio S100/metabolismo , Frações Subcelulares/metabolismo , Ultrassom , Vocalização Animal/fisiologia
10.
Gene ; 489(1): 30-40, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21907770

RESUMO

In the human genome, members of the FoxC, FoxF, FoxL1, and FoxQ1 gene families are found in two paralagous clusters. One cluster contains the genes FOXQ1, FOXF2, FOXC1 and the second consists of FOXF1, FOXC2, and FOXL1. In jawed vertebrates these genes are known to be expressed in different pharyngeal tissues and all, except FoxQ1, are involved in patterning the early embryonic mesoderm. We have previously traced the evolution of this cluster in the bony vertebrates, and the gene content is identical in the dogfish, a member of the most basally branching lineage of the jawed vertebrates. Here we extend these analyses to jawless vertebrates. Using genomic searches and molecular approaches we have identified homologues of these genes from lampreys. We identify two FoxC genes, two FoxF genes, two FoxQ1 genes and single FoxL1 gene. We examine the embryonic expression of one predominantly mesodermally expressed gene family, FoxC, and the endodermally expressed member of the cluster, FoxQ1. We identified FoxQ1 transcripts in the pharyngeal endoderm, while the two FoxC genes are differentially expressed in the pharyngeal mesenchyme and ectoderm. Furthermore we identify conserved expression of lamprey FoxC genes in the paraxial and intermediate mesoderms. We interpret our results through a chordate-wide comparison of expression patterns and discuss gene content in the context of theories on the evolution of the vertebrate genome.


Assuntos
Evolução Molecular , Fatores de Transcrição Forkhead/genética , Lampreias/genética , Sequência de Aminoácidos , Animais , Fatores de Transcrição Forkhead/classificação , Perfilação da Expressão Gênica , Genômica , Mesoderma/metabolismo , Dados de Sequência Molecular , Família Multigênica , Filogenia
11.
Mol Biol Evol ; 28(1): 237-47, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20651048

RESUMO

The FoxP gene subfamily of transcription factors is defined by its characteristic 110 amino acid long DNA-binding forkhead domain and plays essential roles in vertebrate biology. Its four members, FoxP1-P4, have been extensively characterized functionally. FoxP1, FoxP2, and FoxP4 are involved in lung, heart, gut, and central nervous system (CNS) development. FoxP3 is necessary and sufficient for the specification of regulatory T cells (Tregs) of the adaptive immune system. In Drosophila melanogaster, in silico predictions identify one unique FoxP subfamily gene member (CG16899) with no described function. We characterized this gene and established that it generates by alternative splicing two isoforms that differ in the forkhead DNA-binding domain. In D. melanogaster, both isoforms are expressed in the embryonic CNS, but in hemocytes, only isoform A is expressed, hinting to a putative modulation through alternative splicing of FoxP1 function in immunity and/or other hemocyte-dependent processes. Furthermore, we show that in vertebrates, this novel alternative splicing pattern is conserved for FoxP1. In mice, this new FoxP1 isoform is expressed in brain, liver, heart, testes, thymus, and macrophages (equivalent in function to hemocytes). This alternative splicing pattern has arisen at the base of the Bilateria, probably through exon tandem duplication. Moreover, our phylogenetic analysis suggests that in vertebrates, FoxP1 is more related to the FoxP gene ancestral form and the other three paralogues, originated through serial duplications, which only retained one of the alternative exons. Also, the newly described isoform differs from the other in amino acids critical for DNA-binding specificity. The integrity of its fold is maintained, but the molecule has lost the direct hydrogen bonding to DNA bases leading to a putatively lower specificity and possibly affinity toward DNA. With the present comparative study, through the integration of experimental and in silico studies of the FoxP gene subfamily across the animal kingdom, we establish a new model for the FoxP gene in invertebrates and for the vertebrate FoxP1 paralogue. Furthermore, we present a scenario for the structural evolution of this gene class and reveal new previously unsuspected levels of regulation for FoxP1 in the vertebrate system.


Assuntos
Processamento Alternativo , Proteínas de Drosophila/genética , Evolução Molecular , Fatores de Transcrição Forkhead/genética , Duplicação Gênica , Isoformas de Proteínas/genética , Proteínas Repressoras/genética , Sequência de Aminoácidos , Animais , Proteínas de Drosophila/química , Proteínas de Drosophila/classificação , Drosophila melanogaster/genética , Éxons , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/classificação , Hemócitos/fisiologia , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Família Multigênica , Filogenia , Conformação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/classificação , Proteínas Repressoras/química , Proteínas Repressoras/classificação , Alinhamento de Sequência
12.
PLoS One ; 5(7): e11686, 2010 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-20657733

RESUMO

BACKGROUND: In the face of changing environmental conditions, the mechanisms underlying stress responses in diverse organisms are of increasing interest. In vertebrates, Drosophila, and Caenorhabditis elegans, FoxO transcription factors mediate cellular responses to stress, including oxidative stress and dietary restriction. Although FoxO genes have been identified in early-arising animal lineages including sponges and cnidarians, little is known about their roles in these organisms. METHODS/PRINCIPAL FINDINGS: We have examined the regulation of FoxO activity in members of the well-studied cnidarian genus Hydra. We find that Hydra FoxO is expressed at high levels in cells of the interstitial lineage, a cell lineage that includes multipotent stem cells that give rise to neurons, stinging cells, secretory cells and gametes. Using transgenic Hydra that express a FoxO-GFP fusion protein in cells of the interstitial lineage, we have determined that heat shock causes localization of the fusion protein to the nucleus. Our results also provide evidence that, as in bilaterian animals, Hydra FoxO activity is regulated by both Akt and JNK kinases. CONCLUSIONS: These findings imply that basic mechanisms of FoxO regulation arose before the evolution of bilaterians and raise the possibility that FoxO is involved in stress responses of other cnidarian species, including corals.


Assuntos
Cnidários/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sequência de Aminoácidos , Animais , Núcleo Celular/metabolismo , Cnidários/genética , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/classificação , Fatores de Transcrição Forkhead/genética , Proteínas de Fluorescência Verde/genética , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Dados de Sequência Molecular , Filogenia , Proteínas Proto-Oncogênicas c-akt/genética , Homologia de Sequência de Aminoácidos
13.
Genesis ; 47(5): 289-99, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19298014

RESUMO

Oryzias luzonensis is closely related to the medaka, O. latipes. The sex of both species is determined by an XX-XY system. However, the testis determining gene (DMY/Dmrt1bY) found in O. latipes does not exist in O. luzonensis. Instead, a different gene is thought to act as a testis determining gene. In this study, we focused the gonadal sex differentiation process in O. luzonensis under different testis determining gene. First, we observed the gonadal development of O. luzonensis histologically. We then analyzed the expression of Sox9a2/Sox9b, Dmrt1, and Foxl2 during early development. Our results suggest that the sexual differentiation of germ cells in O. luzonensis is initiated later than in O. latipes. However, the timing of the sexual differentiation of the supporting cell linage is similar between the species.


Assuntos
Proteínas de Peixes/genética , Gônadas/metabolismo , Oryzias/genética , Diferenciação Sexual/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , DNA Complementar/química , DNA Complementar/genética , Feminino , Proteínas de Peixes/classificação , Fatores de Transcrição Forkhead/classificação , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/anatomia & histologia , Gônadas/crescimento & desenvolvimento , Hibridização In Situ , Masculino , Dados de Sequência Molecular , Oryzias/crescimento & desenvolvimento , Ovário/anatomia & histologia , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Filogenia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOX9/classificação , Fatores de Transcrição SOX9/genética , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Testículo/anatomia & histologia , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Fatores de Transcrição/classificação , Fatores de Transcrição/genética
14.
Nat Rev Mol Cell Biol ; 8(6): 440-50, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17522590

RESUMO

Members of the class O of forkhead box transcription factors (FoxO) have important roles in metabolism, cellular proliferation, stress tolerance and probably lifespan. The activity of FoxOs is tightly regulated by post-translational modifications, including phosphorylation, acetylation and ubiquitylation. Several of the enzymes that regulate the turnover of these post-translational modifications are shared between FoxO and p53. These regulatory enzymes affect FoxO and p53 function in an opposite manner. This shared yet opposing regulatory network between FoxOs and p53 may underlie a 'trade-off' between disease and lifespan.


Assuntos
Doença , Fatores de Transcrição Forkhead/metabolismo , Expectativa de Vida , Isoformas de Proteínas/metabolismo , Envelhecimento/fisiologia , Animais , Fatores de Transcrição Forkhead/classificação , Fatores de Transcrição Forkhead/genética , Glucose/metabolismo , Humanos , Modelos Biológicos , Isoformas de Proteínas/classificação , Isoformas de Proteínas/genética , Processamento de Proteína Pós-Traducional , Proteína Supressora de Tumor p53/metabolismo
15.
Mech Dev ; 123(10): 761-82, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16963235

RESUMO

Foxe3 is a winged helix/forkhead domain transcription factor necessary for mammalian and amphibian lens development. Human FOXE3 mutations cause anterior segment dysgenesis and cataracts. The zebrafish foxe3 cDNA was PCR amplified from 24 h post-fertilization (hpf) embryo cDNA. The zebrafish foxe3 gene consists of a single exon on chromosome 8 and encodes a 422 amino acid protein. This protein possesses 44% and 67% amino acid identity with the human FOXE3 and Xenopus FoxE4 proteins, respectively. A polyclonal antiserum was generated against a bacterial fusion protein containing the Foxe3 carboxyl terminus. The purified antiserum detects zebrafish Foxe3 on immunoblots, in embryo wholemounts, and frozen tissue sections. The zebrafish Foxe3 protein is first detected in the lens at 31hpf and is restricted to the nucleated cell population, including the epithelial and elongating fiber cells. Knockdown of Foxe3 protein using an antisense morpholino results in small lenses with multilayered epithelial cells and fiber cell dysmorphogenesis. The morphants posses normal retinas, although retinal cell proteins, including rhodopsin, are abnormally expressed in the morphant lens tissue. Functional interactions between foxe3 and pitx3 during lens development were assessed by RT-PCR and comparison of Foxe3 and Pitx3 protein expression in both foxe3 and pitx3 morphants. Immunoblots and immunohistochemistry reveal Pitx3 is expressed in the foxe3 morphant lens, while Pitx3 knockdown results in the elimination of Foxe3 expression. These data demonstrate that Foxe3 is necessary for lens development in zebrafish and that foxe3 lies genetically downstream of pitx3 in a zebrafish lens development pathway.


Assuntos
Proteínas do Olho/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Homeodomínio/metabolismo , Cristalino/embriologia , Morfogênese , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Sequência de Aminoácidos , Animais , Biomarcadores/metabolismo , Proteínas do Olho/classificação , Proteínas do Olho/genética , Fatores de Transcrição Forkhead/classificação , Fatores de Transcrição Forkhead/genética , Proteínas de Homeodomínio/genética , Humanos , Cristalino/anatomia & histologia , Cristalino/metabolismo , Dados de Sequência Molecular , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Filogenia , Alinhamento de Sequência , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/classificação , Proteínas de Peixe-Zebra/genética
16.
Biochem Biophys Res Commun ; 337(4): 1092-6, 2005 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-16236254

RESUMO

Sirtuin 1 (SirT1), an NAD-dependent deacetylase that is important for promoting longevity during caloric restriction, can deacetylate and enhance the function of forkhead box transcription factors, O subfamily (FoxO). We examined the effect of SirT1 on the regulation of insulin-like growth factor-binding protein 1 (IGFBP-1), a known target of FoxO proteins that is increased in fasting. Co-transfection with a SirT1 expression vector dose-dependently stimulated IGFBP-1 promoter activity and a heterologous reporter gene construct containing three FoxO-binding sites linked to a minimal promoter. This effect is mimicked by 20muM resveratrol, a potent SirT1 activator, and immunoprecipitation and Western blotting confirm that SirT1 and FoxO1 interact in cells. Interestingly, mutation of FoxO-binding sites in the IGFBP-1 promoter reduces, but does not completely disrupt, the stimulatory effect of SirT1 on promoter activity. We found that overexpression of SirT1 is accompanied by enhanced mitogen-activated protein kinase (MAPK) activation. Treatment of SirT1-cotransfected cells with PD98059, which inhibits MAPK activation, decreased IGFBP-1 promoter activity by approximately 50%, in a FoxO-binding site-independent manner, and disrupts the residual effect of SirT1. These results indicate that SirT1 stimulates IGFBP-1 promoter activity through FoxO-dependent and -independent mechanisms, and provides the first evidence that activation of MAPK contributes to effects of SirT1 on gene expression.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Sirtuínas/metabolismo , Sequência de Bases , Linhagem Celular , Fatores de Transcrição Forkhead/classificação , Fatores de Transcrição Forkhead/genética , Humanos , Sistema de Sinalização das MAP Quinases , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Sirtuínas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...