Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Aging Cell ; 20(5): e13343, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33721419

RESUMO

Although important factors governing the meiosis have been reported in the embryonic ovary, meiosis in postnatal testis remains poorly understood. Herein, we first report that SRY-box 30 (Sox30) is an age-related and essential regulator of meiosis in the postnatal testis. Sox30-null mice exhibited uniquely impaired testis, presenting the abnormal arrest of germ-cell differentiation and irregular Leydig cell proliferation. In aged Sox30-null mice, the observed testicular impairments were more severe. Furthermore, the germ-cell arrest occurred at the stage of meiotic zygotene spermatocytes, which is strongly associated with critical regulators of meiosis (such as Cyp26b1, Stra8 and Rec8) and sex differentiation (such as Rspo1, Foxl2, Sox9, Wnt4 and Ctnnb1). Mechanistically, Sox30 can activate Stra8 and Rec8, and inhibit Cyp26b1 and Ctnnb1 by direct binding to their promoters. A different Sox30 domain required for regulating the activity of these gene promoters, providing a "fail-safe" mechanism for Sox30 to facilitate germ-cell differentiation. Indeed, retinoic acid levels were reduced owing to increased degradation following the elevation of Cyp26b1 in Sox30-null testes. Re-expression of Sox30 in Sox30-null mice successfully restored germ-cell meiosis, differentiation and Leydig cell proliferation. Moreover, the restoration of actual fertility appeared to improve over time. Consistently, Rec8 and Stra8 were reactivated, and Cyp26b1 and Ctnnb1 were reinhibited in the restored testes. In summary, Sox30 is necessary, sufficient and age-associated for germ-cell meiosis and differentiation in testes by direct regulating critical regulators. This study advances our understanding of the regulation of germ-cell meiosis and differentiation in the postnatal testis.


Assuntos
Fatores de Transcrição SOX/fisiologia , Espermatozoides/citologia , Testículo/citologia , Envelhecimento , Animais , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Regulação da Expressão Gênica , Masculino , Meiose , Prófase Meiótica I , Camundongos , Regiões Promotoras Genéticas , Domínios Proteicos , Fatores de Transcrição SOX/química , Fatores de Transcrição SOX/genética , Fatores de Transcrição SOX/metabolismo , Diferenciação Sexual , Testículo/metabolismo , Tretinoína/metabolismo
2.
Semin Cancer Biol ; 67(Pt 1): 30-38, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-31539559

RESUMO

Sox proteins are a family of lineage-associated transcription factors. They regulate expression of genes involved in control of self-renewal and multipotency in both developmental and adult stem cells. Overexpression of Sox proteins is frequently observed in many different human cancers. Despite their importance as therapeutic targets, Sox proteins are difficult to 'drug' using structure-based design. However, Sox protein localisation, activity and interaction partners are regulated by a plethora of post-translational modifications (PTMs), such as: phosphorylation, acetylation, sumoylation, methylation, and ubiquitylation. Here we review the various reported post-translational modifications of Sox proteins and their potential functional importance in guiding cell fate processes. The enzymes that regulate these PTMs could be useful targets for anti-cancer drug discovery.


Assuntos
Antineoplásicos/uso terapêutico , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Processamento de Proteína Pós-Traducional , Fatores de Transcrição SOX/antagonistas & inibidores , Animais , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Fatores de Transcrição SOX/química , Fatores de Transcrição SOX/genética , Transdução de Sinais
3.
Trends Genet ; 35(9): 658-671, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31288943

RESUMO

The SRY-related (SOX) transcription factor family pivotally contributes to determining cell fate and identity in many lineages. Since the original discovery that SRY deletions cause sex reversal, mutations in half of the 20 human SOX genes have been associated with rare congenital disorders, henceforward called SOXopathies. Mutations are generally de novo, heterozygous, and inactivating, revealing gene haploinsufficiency, but other types, including duplications, have been reported too. Missense variants primarily target the HMG domain, the SOX hallmark that mediates DNA binding and bending, nuclear trafficking, and protein-protein interactions. We here review key clinical and molecular features of SOXopathies and discuss the prospect that the disease family likely involves more SOX genes and larger clinical and genetic spectrums than currently appreciated.


Assuntos
Deficiências do Desenvolvimento/etiologia , Mutação , Fatores de Transcrição SOX/genética , Deficiências do Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento , Haploinsuficiência , Humanos , Fatores de Transcrição SOX/química , Fatores de Transcrição SOX/metabolismo , Fatores de Transcrição SOXD/genética , Proteína da Região Y Determinante do Sexo/genética
4.
Curr Top Dev Biol ; 135: 35-89, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31155363

RESUMO

Primordial germ cells (PGCs) are embryonic precursors of sperm and egg that pass on genetic and epigenetic information from one generation to the next. In mammals, they are induced from a subset of cells in peri-implantation epiblast by BMP signaling from the surrounding tissues. PGCs then initiate a unique developmental program that involves comprehensive epigenetic resetting and repression of somatic genes. This is orchestrated by a set of signaling molecules and transcription factors that promote germ cell identity. Here we review significant findings on mammalian PGC biology, in particular, the genetic basis for PGC specification in mice and human, which has revealed an evolutionary divergence between the two species. We discuss the importance and potential basis for these differences and focus on several examples to illustrate the conserved and divergent roles of critical transcription factors in mouse and human germline.


Assuntos
Células Germinativas/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Fatores de Transcrição SOX/química , Fatores de Transcrição SOX/metabolismo
5.
Genes (Basel) ; 10(4)2019 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-31013663

RESUMO

Coturnix japonica (Japanese quail) has been extensively used as a model animal for biological studies. The Sox gene family, which was systematically characterized by a high-mobility group (HMG-box) in many animal species, encodes transcription factors that play central roles during multiple developmental processes. However, genome-wide investigations on the Sox gene family in birds are scarce. In the current study, we first performed a genome-wide study to explore the Sox gene family in galliform birds. Based on available genomic sequences retrieved from the NCBI database, we focused on the global identification of the Sox gene family in C. japonica and other species in Galliformes, and the evolutionary relationships of Sox genes. In our result, a total of 35 Sox genes in seven groups were identified in the C. japonica genome. Our results also revealed that dispersed gene duplications contributed the most to the expansion of the Sox gene family in Galliform birds. Evolutionary analyses indicated that Sox genes are an ancient gene family, and strong purifying selections played key roles in the evolution of CjSox genes of C. japonica. More interestingly, we observed that most Sox genes exhibited highly embryo-specific expression in both gonads. Our findings provided new insights into the molecular function and phylogeny of Sox gene family in birds.


Assuntos
Coturnix/genética , Perfilação da Expressão Gênica/veterinária , Genômica/métodos , Fatores de Transcrição SOX/genética , Animais , Proteínas Aviárias/química , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Coturnix/embriologia , Evolução Molecular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Família Multigênica , Filogenia , Fatores de Transcrição SOX/química , Fatores de Transcrição SOX/metabolismo , Análise de Sequência de RNA/veterinária , Sequenciamento Completo do Genoma
6.
Am J Hum Genet ; 104(2): 246-259, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30661772

RESUMO

SOX4, together with SOX11 and SOX12, forms group C of SRY-related (SOX) transcription factors. They play key roles, often in redundancy, in multiple developmental pathways, including neurogenesis and skeletogenesis. De novo SOX11 heterozygous mutations have been shown to cause intellectual disability, growth deficiency, and dysmorphic features compatible with mild Coffin-Siris syndrome. Using trio-based exome sequencing, we here identify de novo SOX4 heterozygous missense variants in four children who share developmental delay, intellectual disability, and mild facial and digital morphological abnormalities. SOX4 is highly expressed in areas of active neurogenesis in human fetuses, and sox4 knockdown in Xenopus embryos diminishes brain and whole-body size. The SOX4 variants cluster in the highly conserved, SOX family-specific HMG domain, but each alters a different residue. In silico tools predict that each variant affects a distinct structural feature of this DNA-binding domain, and functional assays demonstrate that these SOX4 proteins carrying these variants are unable to bind DNA in vitro and transactivate SOX reporter genes in cultured cells. These variants are not found in the gnomAD database of individuals with presumably normal development, but 12 other SOX4 HMG-domain missense variants are recorded and all demonstrate partial to full activity in the reporter assay. Taken together, these findings point to specific SOX4 HMG-domain missense variants as the cause of a characteristic human neurodevelopmental disorder associated with mild facial and digital dysmorphism.


Assuntos
Anormalidades Múltiplas/genética , Mutação de Sentido Incorreto/genética , Transtornos do Neurodesenvolvimento/genética , Fatores de Transcrição SOXC/genética , Sequência de Aminoácidos , Animais , Criança , Pré-Escolar , Síndrome de Coffin-Lowry/genética , Estudos de Coortes , Sequência Conservada , DNA/genética , DNA/metabolismo , Feminino , Domínios HMG-Box/genética , Heterozigoto , Humanos , Masculino , Fatores de Transcrição SOX/química , Fatores de Transcrição SOX/genética , Fatores de Transcrição SOXC/química , Fatores de Transcrição SOXC/metabolismo , Ativação Transcricional , Xenopus/anatomia & histologia , Xenopus/embriologia , Xenopus/genética , Proteínas de Xenopus/química , Proteínas de Xenopus/genética
7.
BMC Evol Biol ; 18(1): 205, 2018 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-30587109

RESUMO

BACKGROUND: The Sox family of transcription factors is an important part of the genetic 'toolbox' of all metazoans examined to date and is known to play important developmental roles in vertebrates and insects. However, outside the commonly studied Drosophila model little is known about the repertoire of Sox family transcription factors in other arthropod species. Here we characterise the Sox family in two chelicerate species, the spiders Parasteatoda tepidariorum and Stegodyphus mimosarum, which have experienced a whole genome duplication (WGD) in their evolutionary history. RESULTS: We find that virtually all of the duplicate Sox genes have been retained in these spiders after the WGD. Analysis of the expression of Sox genes in P. tepidariorum embryos suggests that it is likely that some of these genes have neofunctionalised after duplication. Our expression analysis also strengthens the view that an orthologue of vertebrate Group B1 genes, SoxNeuro, is implicated in the earliest events of CNS specification in both vertebrates and invertebrates. In addition, a gene in the Dichaete/Sox21b class is dynamically expressed in the spider segment addition zone, suggestive of an ancient regulatory mechanism controlling arthropod segmentation as recently suggested for flies and beetles. Together with the recent analysis of Sox gene expression in the embryos of other arthropods, our findings support the idea of conserved functions for some of these genes, including a potential role for SoxC and SoxD genes in CNS development and SoxF in limb development. CONCLUSIONS: Our study provides a new chelicerate perspective to understanding the evolution and function of Sox genes and how the retention of duplicates of such important tool-box genes after WGD has contributed to different aspects of spider embryogenesis. Future characterisation of the function of these genes in spiders will help us to better understand the evolution of the regulation of important developmental processes in arthropods and other metazoans including neurogenesis and segmentation.


Assuntos
Evolução Molecular , Fatores de Transcrição SOX/genética , Fatores de Transcrição SOX/metabolismo , Aranhas/embriologia , Aranhas/genética , Animais , Desenvolvimento Embrionário , Duplicação Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genoma , Organogênese , Filogenia , Fatores de Transcrição SOX/química
8.
J Exp Clin Cancer Res ; 37(1): 111, 2018 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-29855376

RESUMO

BACKGROUND: The expression of desmosomal genes in lung adenocarcinoma and lung squamous carcinoma is different. However, the regulatory mechanism of desmosomal gene expression in lung adenocarcinoma and lung squamous carcinoma remains unknown. METHODS: The correlation between expression of desmosomal gene expression and SOX30 expression were analyzed by bioinformatics. The expression of SOX30, DSP, JUP and DSC3 were detected in lung cancer cell lines, lung tissues of mice and patients' tissues by qPCR, WB, Immunofluorescence and Immunohistochemistry. A chromatin Immunoprecipitation assay was used to investigate the mechanisms of the SOX30 regulation on desmosomal gene expression. In vitro proliferation, migration and invasion assays, and an in vivo nude mice model were utilized to assess the important role of desmosomal genes on SOX30-induced tumor suppression. A WB assay and TOP/FOP flash reporter assay was used to investigate the downstream pathway regulated by the SOX30-desmosomal gene axis. A chemical carcinogenic model of SOX30-knockout mice was generated to confirm the role of the SOX30-desmosomal gene axis in tumorigenesis. RESULTS: The expression of desmosomal genes were upregulated by SOX30 in lung adenocarcinoma but not in lung squamous carcinoma. Further mechanism studies showed that SOX30 acts as a key transcriptional regulator of desmosomal genes by directly binding to the ACAAT motif of desmosomal genes promoter region and activating their transcription in lung adenocarcinoma. Knockdown of the expression of related desmosomal genes by miRNA significantly attenuated the inhibitory effect of SOX30 on cell proliferation, migration and invasion in vitro and on tumor growth and metastasis in vivo. In addition, knockout of SOX30 promotes lung tumor development and loss the inhibition of desmosomal genes on downstream Wnt and ERK signal in urethane-induced lung carcinogenesis in SOX30-knockout mice. CONCLUSIONS: Overall, these findings demonstrate for the first time that SOX30 acts as a master switch of desmosomal genes, inhibits lung adenocarcinoma cell proliferation, migration and invasion by activating the transcription of desmosomal genes. This study provides novel insights on the regulatory mechanism of desmosomal genes in lung adenocarcinoma.


Assuntos
Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/patologia , Desmossomos/genética , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição SOX/genética , Proteínas Supressoras de Tumor/genética , Adenocarcinoma de Pulmão/metabolismo , Animais , Sequência de Bases , Biomarcadores , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Xenoenxertos , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , Modelos Biológicos , Metástase Neoplásica , Regiões Promotoras Genéticas , Fatores de Transcrição SOX/química , Fatores de Transcrição SOX/metabolismo , Transcriptoma , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/metabolismo , Via de Sinalização Wnt
9.
Artigo em Inglês | MEDLINE | ID: mdl-29906772

RESUMO

The Sox gene family has been systematically characterized in some fish species but not in catfish Ictalurus punctatus. In this study, 25 Sox genes were identified in the channel catfish genome and classified into seven families based on their conserved domains as follows: eight genes in SoxB group (six in SoxB1 subgroup and two in SoxB2 subgroup); five genes in SoxC group; three genes in SoxD and SoxF groups; four genes in SoxE group; and one gene in SoxH and SoxK groups. The mammalian Sox groups SoxA, G, I, and J were not present in catfish. The number of introns in channel catfish Sox genes varied from zero to 13. Sox genes were distributed unevenly across 17 chromosomes. Five members of the ancestral vertebrate Sox genes (Sox1, Sox4, Sox9, Sox11 and Sox19) experienced teleost-specific whole genome duplication during evolution, and now have two copies on different chromosomes. Expression profiles analyses indicated that the accumulation of Sox genes was associated with different tissues, and the expression pattern also differed among each Sox gene group and duplicated gene. This study constitutes a comprehensive overview of the Sox gene family in channel catfish and provides new insights into the evolution of this gene family.


Assuntos
Proteínas de Peixes/genética , Ictaluridae/genética , Filogenia , Fatores de Transcrição SOX/genética , Transcriptoma , Sequência de Aminoácidos , Animais , Evolução Molecular , Proteínas de Peixes/química , Duplicação Gênica , Genoma , Fatores de Transcrição SOX/química , Alinhamento de Sequência
10.
Semin Cell Dev Biol ; 63: 2-12, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27521520

RESUMO

The Sox transcription factor family consists of 20 members in the human genome. Many of them are key determinants of cellular identities and possess the capacity to reprogram cell fates by pioneering the epigenetic remodeling of the genome. This activity is intimately tied to their ability to specifically bind and bend DNA alone or with other proteins. Here we discuss our current knowledge on how Sox transcription factors such as Sox2, Sox17, Sox18 and Sox9 'read' the genome to find and regulate their target genes and highlight the roles of partner factors including Pax6, Nanog, Oct4 and Brn2. We integrate insights from structural and biochemical studies as well as high-throughput assays to probe DNA specificity in vitro as well as in cells and tissues.


Assuntos
Genoma , Fatores de Transcrição SOX/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Modelos Moleculares , Fatores de Transcrição SOX/química , Fatores de Transcrição SOX/genética
11.
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
12.
PLoS One ; 11(6): e0157821, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27331398

RESUMO

Cephalopods provide an unprecedented opportunity for comparative studies of the developmental genetics of organ systems that are convergent with analogous vertebrate structures. The Sox-family of transcription factors is an important class of DNA-binding proteins that are known to be involved in many aspects of differentiation, but have been largely unstudied in lophotrochozoan systems. Using a degenerate primer strategy we have isolated coding sequence for three members of the Sox family of transcription factors from a cephalopod mollusk, the European cuttlefish Sepia officinalis: Sof-SoxE, Sof-SoxB1, and Sof-SoxB2. Analyses of their expression patterns during organogenesis reveals distinct spatial and temporal expression domains. Sof-SoxB1 shows early ectodermal expression throughout the developing epithelium, which is gradually restricted to presumptive sensory epithelia. Expression within the nervous system appears by mid-embryogenesis. Sof-SoxB2 expression is similar to Sof-SoxB1 within the developing epithelia in early embryogenesis, however appears in largely non-overlapping expression domains within the central nervous system and is not expressed in the maturing sensory epithelium. In contrast, Sof-SoxE is expressed throughout the presumptive mesodermal territories at the onset of organogenesis. As development proceeds, Sof-SoxE expression is elevated throughout the developing peripheral circulatory system. This expression disappears as the circulatory system matures, but expression is maintained within undifferentiated connective tissues throughout the animal, and appears within the nervous system near the end of embryogenesis. SoxB proteins are widely known for their role in neural specification in numerous phylogenetic lineages. Our data suggests that Sof-SoxB genes play similar roles in cephalopods. In contrast, Sof-SoxE appears to be involved in the early stages of vasculogenesis of the cephalopod closed circulatory system, a novel role for a member of this gene family.


Assuntos
Sequência Conservada , Fatores de Transcrição SOX/genética , Sepia/genética , Sequência de Aminoácidos , Animais , Clonagem Molecular , Desenvolvimento Embrionário/genética , Feminino , Secções Congeladas , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Filogenia , Domínios Proteicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição SOX/química , Fatores de Transcrição SOX/metabolismo , Sepia/embriologia , Alinhamento de Sequência , Fatores de Tempo
13.
Nucleic Acids Res ; 44(8): 3922-35, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-26939885

RESUMO

The transcription factor (TF) SOX18 drives lymphatic vessel development in both embryogenesis and tumour-induced neo-lymphangiogenesis. Genetic disruption of Sox18 in a mouse model protects from tumour metastasis and established the SOX18 protein as a molecular target. Here, we report the crystal structure of the SOX18 DNA binding high-mobility group (HMG) box bound to a DNA element regulating Prox1 transcription. The crystals diffracted to 1.75Å presenting the highest resolution structure of a SOX/DNA complex presently available revealing water structure, structural adjustments at the DNA contact interface and non-canonical conformations of the DNA backbone. To explore alternatives to challenging small molecule approaches for targeting the DNA-binding activity of SOX18, we designed a set of five decoys based on modified Prox1-DNA. Four decoys potently inhibited DNA binding of SOX18 in vitro and did not interact with non-SOX TFs. Serum stability, nuclease resistance and thermal denaturation assays demonstrated that a decoy circularized with a hexaethylene glycol linker and terminal phosphorothioate modifications is most stable. This SOX decoy also interfered with the expression of a luciferase reporter under control of a SOX18-dependent VCAM1 promoter in COS7 cells. Collectively, we propose SOX decoys as potential strategy for inhibiting SOX18 activity to disrupt tumour-induced neo-lymphangiogenesis.


Assuntos
DNA/química , Proteínas de Homeodomínio/genética , Fatores de Transcrição SOXF/antagonistas & inibidores , Fatores de Transcrição SOXF/química , Proteínas Supressoras de Tumor/genética , Animais , Células COS , Chlorocebus aethiops , DNA/metabolismo , Regulação da Expressão Gênica , Camundongos , Conformação de Ácido Nucleico , Oligonucleotídeos , Fatores de Transcrição SOX/química , Fatores de Transcrição SOX/metabolismo , Fatores de Transcrição SOXF/metabolismo , Transcrição Gênica
14.
Theriogenology ; 84(2): 268-76.e1, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25913275

RESUMO

Because little is known about the function of Sox2 (Sry-related box-2) in teleosts, the objective of this study was to clone and characterize Sox2 complementary DNA (cDNA) from the testis of Indian major carp, Labeo rohita (rohu). The full-length cDNA contained an open reading frame of 936 nucleotides bearing the typical structural features. Phylogenetically, Sox2 of L rohita was most closely related to freshwater counterparts than marine water. The sequence information of cDNA and genomic DNA together revealed that the Sox2 gene is encoded by an uninterrupted exon. Furthermore, comparative mRNA expression profile in various organs including proliferating spermatogonial stem cells (SSCs) suggested about the participatory role of Sox2 during fish male germ cell development and maintenance of stem cells. In support, we have also provided evidence that Sox2 protein is indeed present in rohu SSCs by Western blot analysis. The evolutionarily conserved high-mobility group box domain indicated its possible involvement in common networking pathways for stem cell maintenance and pluripotency between mammals and nonmammals. Our findings could be the first step toward the use of Sox2 as a potential biomarker for proliferating SSCs and understanding the transcriptional regulatory network involved during male germ cell development and maintenance in fish species.


Assuntos
Carpas/metabolismo , Proteínas de Peixes/genética , Expressão Gênica , Fatores de Transcrição SOX/genética , Espermatogônias/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Proliferação de Células , Clonagem Molecular , DNA Complementar/genética , Masculino , Dados de Sequência Molecular , Filogenia , RNA Mensageiro/análise , Fatores de Transcrição SOX/análise , Fatores de Transcrição SOX/química , Análise de Sequência de DNA/veterinária , Espermatogônias/química , Testículo/química , Transcriptoma
15.
Cell Tissue Res ; 359(1): 111-24, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24894327

RESUMO

Neuroepithelial precursor cells of the vertebrate central nervous system either self-renew or differentiate into neurons, oligodendrocytes or astrocytes under the influence of a gene regulatory network that consists in transcription factors, epigenetic modifiers and microRNAs. Sox transcription factors are central to this regulatory network, especially members of the SoxB, SoxC, SoxD, SoxE and SoxF groups. These Sox proteins are widely expressed in neuroepithelial precursor cells and in newly specified, differentiating and mature neurons, oligodendrocytes and astrocytes and influence their identity, survival and development. They exert their effect predominantly at the transcriptional level but also have substantial impact on expression at the epigenetic and posttranscriptional levels with some Sox proteins acting as pioneer factors, recruiting chromatin-modifying and -remodelling complexes or influencing microRNA expression. They interact with a large variety of other transcription factors and influence the expression of regulatory molecules and effector genes in a cell-type-specific and temporally controlled manner. As versatile regulators with context-dependent functions, they are not only indispensable for central nervous system development but might also be instrumental for the development of reprogramming and cell conversion strategies for replacement therapies and for assisted regeneration after injury or degeneration-induced cell loss in the central nervous system.


Assuntos
Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Fatores de Transcrição SOX/metabolismo , Células-Tronco/metabolismo , Animais , Humanos , Neurogênese , Neuroglia/citologia , Neurônios/citologia , Fatores de Transcrição SOX/química , Células-Tronco/citologia
16.
Artigo em Inglês | MEDLINE | ID: mdl-22940139

RESUMO

The Sox family of transcription factors are involved in a variety of developmental processes including sex determination and gonadal differentiation. Sox19 is a particularly interesting member of this family that has been found only in fish, though mammals have a very diverged orthologue that is designated Sox15 and assigned to a different Sox family subgroup. Here we describe the cloning and characterisation of sox19 from the European sea bass (Dicentrarchus labrax), an important aquaculture species in which sex ratios skewed in favour of males are frequently encountered. The sea bass sox19 gene contains a single intron, encodes a protein of 309 amino acids, has multiple transcription start sites and may produce a truncated splice variant. Sox19 mRNA is present in many adult tissues, with the highest expression in the brain and gonads. Interestingly, the gene is strongly upregulated in the differentiation of the ovary but not the testis, suggesting a role in ovarian differentiation.


Assuntos
Bass/crescimento & desenvolvimento , Bass/genética , Proteínas de Peixes/genética , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição SOX/genética , Diferenciação Sexual/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Feminino , Proteínas de Peixes/química , Masculino , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Fatores de Transcrição SOX/química , Caracteres Sexuais , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
17.
Nucleic Acids Res ; 40(11): 4933-41, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22344693

RESUMO

Several Sox-Oct transcription factor (TF) combinations have been shown to cooperate on diverse enhancers to determine cell fates. Here, we developed a method to quantify biochemically the Sox-Oct cooperation and assessed the pairing of the high-mobility group (HMG) domains of 11 Sox TFs with Oct4 on a series of composite DNA elements. This way, we clustered Sox proteins according to their dimerization preferences illustrating that Sox HMG domains evolved different propensities to cooperate with Oct4. Sox2, Sox14, Sox21 and Sox15 strongly cooperate on the canonical element but compete with Oct4 on a recently discovered compressed element. Sry also cooperates on the canonical element but binds additively to the compressed element. In contrast, Sox17 and Sox4 cooperate more strongly on the compressed than on the canonical element. Sox5 and Sox18 show some cooperation on both elements, whereas Sox8 and Sox9 compete on both elements. Testing rationally mutated Sox proteins combined with structural modeling highlights critical amino acids for differential Sox-Oct4 partnerships and demonstrates that the cooperativity correlates with the efficiency in producing induced pluripotent stem cells. Our results suggest selective Sox-Oct partnerships in genome regulation and provide a toolset to study protein cooperation on DNA.


Assuntos
Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOX/química , Fatores de Transcrição SOX/metabolismo , Sequência de Aminoácidos , Animais , Ligação Competitiva , DNA/metabolismo , Dimerização , Camundongos , Dados de Sequência Molecular , Mutação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Fatores de Transcrição SOX/genética , Alinhamento de Sequência
18.
Chromosome Res ; 20(1): 153-62, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22161124

RESUMO

Even though the mammalian sex-determining gene Sry has been intensively studied for the two decades since its discovery, the regions outside the conserved HMG box DNA-binding domain have received less attention due to a lack of sequence conservation and of obvious structural/functional motifs. Here, we summarize the available evidence for function beyond the HMG box, identify the known and postulated biochemical functions of the non-HMG-box domains in sex determination, and present possible explanations for the puzzling diversity of these non-HMG-box domains.


Assuntos
Domínios HMG-Box , Fatores de Transcrição SOX/genética , Cromossomos Sexuais/genética , Processos de Determinação Sexual , Animais , DNA/química , DNA/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Feminino , Variação Genética , Disgenesia Gonadal/genética , Humanos , Masculino , Mamíferos , Mutação , Ligação Proteica , Fatores de Transcrição SOX/química , Cromossomos Sexuais/química , Ativação Transcricional
19.
Int J Biochem Cell Biol ; 42(3): 449-52, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19909824

RESUMO

In mammals, the group G of the Sry-related high-mobility-group (HMG) box genes (Sox) contains only one member, Sox15. Comparative genomic analysis of the Sox genes in the B1 and G groups indicates that an ancestral gene may have originated as an intron-containing gene belonging to group B1 and evolved into zebrafish Sox19a/b, Xenopus SoxD, and mammalian Sox15. Although these genes have different names, they are orthologous. The zebrafish and Xenopus orthologues are highly expressed in the central nervous system, whereas mouse Sox15 only shows strong expression in the placenta, an organ characteristic of all mammals except monotremes. Interestingly, Sox15 appears to be a pseudogene in the marsupial opossum. Sox15-deficient mice exhibit delayed skeletal muscle regeneration, indicating that Sox15 plays a crucial role in this process. On the other hand, Xenopus SoxD induces anterior neural development. Thus, there appears to be little functional overlap between Sox15 and its orthologues, Sox19a/b and SoxD. In this review, I discuss the roles of Sox15, its functional redundancy with SoxB1 group members, and its molecular evolution.


Assuntos
Evolução Molecular , Mamíferos/genética , Fatores de Transcrição SOX/genética , Fatores de Transcrição SOX/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Desenvolvimento Muscular/genética , Regiões Promotoras Genéticas/genética , Fatores de Transcrição SOX/química
20.
Int J Biochem Cell Biol ; 42(3): 411-6, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19808100

RESUMO

The control of access of SOX proteins to their nuclear target genes is a powerful strategy to activate or repress complex genetic programs. The sub-cellular targeting sequences of SOX proteins are concentrated within the DNA binding motif, the HMG (for high mobility group) domain. Each SOX protein displays two different nuclear localization signals located at the N-terminal and C-terminal part of their highly conserved DNA binding domain. The N-terminal nuclear localization signal binds calmodulin and is potentially regulated by intracellular calcium signalling, while the C-terminal nuclear localization signal, which binds importin-beta, responds to other signalling pathways such as cyclic AMP/protein kinase A. Mutations inducing developmental disorders like sex reversal have been reported in both NLSs of SRY, interfering with its nuclear localization and suggesting that both functional nuclear localization signal are required for its nuclear activity. A nuclear export signal is also present in the HMG box of SOX proteins. Group E SOX proteins harbour a perfect consensus nuclear export signal sequence in contrast to all other SOX proteins, which display only imperfect ones. However, observations made during mouse embryonic development suggest that non-group E SOX proteins could also be regulated by a nuclear export mechanism. The presence of nuclear localization and nuclear export signal sequences confers nucleocytoplasmic shuttling properties to SOX proteins, and suggests that cellular events regulated by SOX proteins are highly dynamic.


Assuntos
Fatores de Transcrição SOX/metabolismo , Sequência de Aminoácidos , Animais , Núcleo Celular/metabolismo , Desenvolvimento Embrionário , Humanos , Dados de Sequência Molecular , Transporte Proteico , Fatores de Transcrição SOX/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...