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












Base de datos
Intervalo de año de publicación
1.
Nature ; 580(7805): 669-672, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32350470

RESUMEN

'Pioneer' transcription factors are required for stem-cell pluripotency, cell differentiation and cell reprogramming1,2. Pioneer factors can bind nucleosomal DNA to enable gene expression from regions of the genome with closed chromatin. SOX2 is a prominent pioneer factor that is essential for pluripotency and self-renewal of embryonic stem cells3. Here we report cryo-electron microscopy structures of the DNA-binding domains of SOX2 and its close homologue SOX11 bound to nucleosomes. The structures show that SOX factors can bind and locally distort DNA at superhelical location 2. The factors also facilitate detachment of terminal nucleosomal DNA from the histone octamer, which increases DNA accessibility. SOX-factor binding to the nucleosome can also lead to a repositioning of the N-terminal tail of histone H4 that includes residue lysine 16. We speculate that this repositioning is incompatible with higher-order nucleosome stacking, which involves contacts of the H4 tail with a neighbouring nucleosome. Our results indicate that pioneer transcription factors can use binding energy to initiate chromatin opening, and thereby facilitate nucleosome remodelling and subsequent transcription.


Asunto(s)
Ensamble y Desensamble de Cromatina , Microscopía por Crioelectrón , Nucleosomas/metabolismo , Factores de Transcripción SOXB1/química , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción SOXC/química , Factores de Transcripción SOXC/metabolismo , Secuencia de Bases , ADN Superhelicoidal/química , ADN Superhelicoidal/genética , ADN Superhelicoidal/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Lisina/metabolismo , Modelos Biológicos , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Nucleosomas/química , Nucleosomas/ultraestructura , Factores de Transcripción SOXB1/ultraestructura , Factores de Transcripción SOXC/ultraestructura
2.
Am J Hum Genet ; 104(2): 246-259, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30661772

RESUMEN

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.


Asunto(s)
Anomalías Múltiples/genética , Mutación Missense/genética , Trastornos del Neurodesarrollo/genética , Factores de Transcripción SOXC/genética , Secuencia de Aminoácidos , Animales , Niño , Preescolar , Síndrome de Coffin-Lowry/genética , Estudios de Cohortes , Secuencia Conservada , ADN/genética , ADN/metabolismo , Femenino , Dominios HMG-Box/genética , Heterocigoto , Humanos , Masculino , Factores de Transcripción SOX/química , Factores de Transcripción SOX/genética , Factores de Transcripción SOXC/química , Factores de Transcripción SOXC/metabolismo , Activación Transcripcional , Xenopus/anatomía & histología , Xenopus/embriología , Xenopus/genética , Proteínas de Xenopus/química , Proteínas de Xenopus/genética
3.
Artículo en Inglés | MEDLINE | ID: mdl-26827849

RESUMEN

The characterization and expression of Sox4 in large yellow croaker (Lc-Sox4) were studied in this paper. Lc-Sox4 contains a protein of 371 amino acids with a conserved high mobility group box. Quantitative real-time PCR displayed that the expression of Lc-Sox4 had tissue and gender specificity existing in brain, gonad, heart, intestine, and head kidney with male>female, in eye with female>male. During embryogenesis, Lc-Sox4 was expressed highest in one-day-post-hatching stage, next in formation-of-eye-lens stage. The expression pattern of Lc-Sox4 was different from that of Lc-Sox11a. The expression of Lc-Sox4 was significantly lower than that of Lc-Sox11a in the all tested tissues and embryonic stages except in heart, spleen, mutiple-cell, formation-of-eye-lens, and one-day-post-hatching stages (with Lc-Sox4 higher than Lc-Sox11a). There was overlapping expression between Lc-Sox4 and Lc-Sox11a in brain, gill, female eye, testis, formation-of-eye-lens stage and one-day post hatching stage. The whole mount in situ hybridization results indicated that Lc-Sox4 was expressed at all embryonic stages except 2-cell stage. The positive signals were mainly distributed in the central nervous system and notochord at one-day-post-hatching stage. In short, we first identified and analyzed the temporal and spatial expression patterns of Lc-Sox4 to elucidate its important influence on the development of nervous system, visual system and heart. We also detected the overlapping expression between Lc-Sox4 and Lc-Sox11a which may reveal the functional redundancy of them. These data would shed light on the molecular mechanism of development in large yellow croaker.


Asunto(s)
Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Regulación del Desarrollo de la Expresión Génica , Perciformes/genética , Factores de Transcripción SOXC/genética , Factores de Transcripción SOXC/metabolismo , Animales , Clonación Molecular , Evolución Molecular , Femenino , Proteínas de Peces/química , Masculino , Modelos Moleculares , Perciformes/embriología , Conformación Proteica , Transporte de Proteínas , Factores de Transcripción SOXC/química , Alineación de Secuencia
4.
Gene ; 574(2): 287-301, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26275936

RESUMEN

Sox genes play important roles in various developmental processes such as sex determination, embryogenesis, oogenesis, neurogenesis, and larval development. In order to clarify the roles of Sox genes in the developmental process of large yellow croaker, the full-length cDNA of the Sox11a gene (Lc-Sox11a) was cloned for the first time. Bioinformatics analysis indicated that Lc-Sox11a contains a protein of 366 amino acids with a Ser-rich region, a C-terminal conserved region, and a high mobility group box. The expression of Lc-Sox11a in different tissues of both sexes and in different developmental embryonic stages revealed that Lc-Sox11a were expressed with tissue and gender specificity, of which the expression level in female was ovary>brain>eye>gill; in male was brain>testis>gill. The gender differences occurred in the brain and eye with the male brain>female brain, female eye>male eye. Moreover, the expression of Lc-Sox11a in the gonad and brain at different growth stages was detected. Significant up-regulated expression of Lc-Sox11a was found in the ovary and the male brain at 1000dph (days post hatching) compared with 270dph and 635dph. However, significant down-regulated expression of Lc-Sox11a occurred in the testis with growth. Besides, the expression of Lc-Sox11a in the female brain showed a trend of first rising then falling, with the highest peak in 635dph. The results of in situ hybridization displayed that Lc-Sox11a was widely distributed only in cytoplasm of oocytes at each stage in oogenesis. In early stage of oocytes, Lc-Sox11a was expressed weakly and evenly. As the appearance of vacuoles and synthesis of yolks, positive signals of Lc-Sox11a distributed intensively in the residual cytoplasm. In spermatogenesis, Lc-Sox11a was distributed in cytoplasm of all male germ cells except spermatozoon with spermatogonium>spermatocyte>spermatid. During embryogenesis, Lc-Sox11a was expressed in most embryonic stages, the highest expression occurred in the formation-of-eye-lens stage, closely followed by the closure-of-blastopore stage, then the beginning-of-heart-pulsation stage. The results of whole mount in situ hybridization showed that the expression of Lc-Sox11a began to increase beginning with the multiple-cell stage, with the major distribution of Lc-Sox11a in the brain and eye areas in the pre-hatching stage.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Perciformes/genética , Factores de Transcripción SOXC/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Desarrollo Embrionario/genética , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Perciformes/embriología , Filogenia , Factores de Transcripción SOXC/química , Factores de Transcripción SOXC/clasificación , Análisis de Secuencia de ADN
5.
Sci Rep ; 5: 10066, 2015 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-25970172

RESUMEN

Hepatitis B virus (HBV) infection is a major cause of liver diseases. However, the mechanisms underlying HBV infection and pathogenesis remain largely unknown. The sex-determining region Y box 4 (Sox4) is a transcriptional factor, which preferentially regulates the development of various organs, tissues, and cancers. But, the role of Sox4 in viral infection and pathogenesis has not been elucidated. Here, we demonstrated that Sox4 is up-regulated by HBV, and revealed the mechanism by which HBV regulates Sox4 expression. First, HBV stimulates Sox4 expression through transcriptional factor Yin Yang 1 (YY1), which binds to Sox4 promoter to activate Sox4 transcriptional activity. Second, miR-335, miR-129-2 and miR-203 inhibit Sox4 expression by targeting its mRNA 3'UTR, while HBV suppresses the microRNAs expression, resulting in up-regulating Sox4 post-transcriptionally. Third, Sox4 protein is degraded by proteasome, while HBV surface protein (HBsAg) prevents Sox4 from degradation by directly interacting with the protein, thereby enhancing Sox4 production post-translationlly. More interestingly, HBV-activated Sox4 in turn facilitates HBV replication by direct binding to the viral genome via its HMG box. Thus, this study revealed a novel positive feedback mechanism by which Sox4 production and HBV replication are tightly correlated.


Asunto(s)
Regulación de la Expresión Génica , Virus de la Hepatitis B/fisiología , Hepatitis B/genética , Hepatitis B/virología , Factores de Transcripción SOXC/genética , Replicación Viral , Carcinoma Hepatocelular/etiología , Epigénesis Genética , Genoma Viral , Hepatitis B/complicaciones , Hepatitis B/metabolismo , Humanos , Neoplasias Hepáticas/etiología , Sistema de Señalización de MAP Quinasas , MicroARNs/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Factores de Transcripción SOXC/química , Factores de Transcripción SOXC/metabolismo , Activación Transcripcional , Proteínas Virales/metabolismo , Factor de Transcripción YY1/metabolismo
6.
Mol Cancer ; 14: 24, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25644061

RESUMEN

BACKGROUND: Tumor metastasis is responsible for 90% of cancer-related deaths. Recently, a strong link between microRNA dysregulation and human cancers has been established. However, the molecular mechanisms through which microRNAs regulate metastasis and cancer progression remain unclear. METHODS: We analyzed the reciprocal expression regulation of miR-31 and SOX4 in esophageal squamous and adenocarcinoma cell lines by qRT-PCR and Western blotting using overexpression and shRNA knock-down approaches. Furthermore, methylation studies were used to assess epigenetic regulation of expression. Functionally, we determined the cellular consequences using migration and invasion assays, as well as proliferation assays. Immunoprecipitation and ChIP were used to identify complex formation of SOX4 and co-repressor components. RESULTS: Here, we report that SOX4 promotes esophageal tumor cell proliferation and invasion by silencing miR-31 via activation and stabilization of a co-repressor complex with EZH2 and HDAC3. We demonstrate that miR-31 is significantly decreased in invasive esophageal cancer cells, while upregulation of miR-31 inhibits growth, migration and invasion of esophageal adenocarcinoma (EAC) and squamous cell carcinoma (ESCC) cell lines. miR-31, in turn, targets SOX4 for degradation by directly binding to its 3'-UTR. Additionally, miR-31 regulates EZH2 and HDAC3 indirectly. SOX4, EZH2 and HDAC3 levels inversely correlate with miR-31 expression in ESCC cell lines. Ectopic expression of miR-31 in ESCC and EAC cell lines leads to down regulation of SOX4, EZH2 and HDAC3. Conversely, pharmacologic and genetic inhibition of SOX4 and EZH2 restore miR-31 expression. We show that SOX4, EZH2 and HDAC3 form a co-repressor complex that binds to the miR-31 promoter, repressing miR-31 through an epigenetic mark by H3K27me3 and by histone acetylation. Clinically, when compared to normal adjacent tissues, esophageal tumor samples show upregulation of SOX4, EZH2, and HDAC3, and EZH2 expression is significantly increased in metastatic ESCC tissues. CONCLUSIONS: Thus, we identified a novel molecular mechanism by which the SOX4, EZH2 and miR-31 circuit promotes tumor progression and potential therapeutic targets for invasive esophageal carcinomas.


Asunto(s)
Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasas/metabolismo , MicroARNs/genética , Complejo Represivo Polycomb 2/metabolismo , Factores de Transcripción SOXC/metabolismo , Regiones no Traducidas 3' , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Regulación hacia Abajo , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Neoplasias Esofágicas/patología , Histona Desacetilasas/genética , Humanos , MicroARNs/química , Invasividad Neoplásica , Complejo Represivo Polycomb 2/genética , Unión Proteica , Interferencia de ARN , Proteínas Represoras/metabolismo , Factores de Transcripción SOXC/química , Factores de Transcripción SOXC/genética
7.
Anal Chem ; 86(10): 5116-24, 2014 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-24766394

RESUMEN

A highly sensitive platform coupling capillary ion chromatography (Cap IC) with Q Exactive mass spectrometer has been developed for metabolic profiling of head and neck squamous cell carcinoma (HNSCC) cells. The Cap IC allowed an excellent separation of anionic polar metabolites, and the sensitivities increased by up to 100-fold compared to reversed-phase liquid chromatography and hydrophilic interaction chromatography performed at either high- or capillary-flow rates. The detection limits for a panel of standard metabolites were between 0.04 to 0.5 nmol/L (0.2 to 3.4 fmol) at a signal-to-noise ratio of 3. This platform was applied to an untargeted metabolomic analysis of head and neck cancer cells and stem-like cancer cells. Differential metabolomics analysis identified significant changes in energy metabolism pathways (e.g., glycolysis and tricarboxylic acid cycle). These experiments demonstrate Cap IC/MS as a powerful metabolomics tool by providing enhanced separation and sensitivity of polar metabolites combined with high resolution and accurate mass measurement (HR/AM) capabilities to differentiate isobaric metabolites.


Asunto(s)
Aniones/química , Neoplasias de Cabeza y Cuello/química , Metabolómica/métodos , Electrocromatografía Capilar , Línea Celular Tumoral , Humanos , Espectrometría de Masas , Células Madre Neoplásicas/química , ARN Interferente Pequeño/farmacología , Factores de Transcripción SOXC/química , Factores de Transcripción SOXC/genética
8.
Biochem J ; 443(1): 39-47, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22181698

RESUMEN

It has recently been proposed that the sequence preferences of DNA-binding TFs (transcription factors) can be well described by models that include the positional interdependence of the nucleotides of the target sites. Such binding models allow for multiple motifs to be invoked, such as principal and secondary motifs differing at two or more nucleotide positions. However, the structural mechanisms underlying the accommodation of such variant motifs by TFs remain elusive. In the present study we examine the crystal structure of the HMG (high-mobility group) domain of Sox4 [Sry (sex-determining region on the Y chromosome)-related HMG box 4] bound to DNA. By comparing this structure with previously solved structures of Sox17 and Sox2, we observed subtle conformational differences at the DNA-binding interface. Furthermore, using quantitative electrophoretic mobility-shift assays we validated the positional interdependence of two nucleotides and the presence of a secondary Sox motif in the affinity landscape of Sox4. These results suggest that a concerted rearrangement of two interface amino acids enables Sox4 to accommodate primary and secondary motifs. The structural adaptations lead to altered dinucleotide preferences that mutually reinforce each other. These analyses underline the complexity of the DNA recognition by TFs and provide an experimental validation for the conceptual framework of positional interdependence and secondary binding motifs.


Asunto(s)
ADN/química , Sustancias Macromoleculares/química , Factores de Transcripción SOXC/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Secuencia Conservada , Cristalografía por Rayos X , Ensayo de Cambio de Movilidad Electroforética , Elementos de Facilitación Genéticos , Proteínas HMGB/química , Laminina/genética , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Unión Proteica , Estructura Terciaria de Proteína , Factores de Transcripción SOXF/química
9.
Cell Mol Life Sci ; 68(23): 3949-61, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21538160

RESUMEN

Tbx3, a T-box transcription factor, regulates key steps in development of the heart and other organ systems. Here, we identify Sox4 as an interacting partner of Tbx3. Pull-down and nuclear retention assays verify this interaction and in situ hybridization reveals Tbx3 and Sox4 to co-localize extensively in the embryo including the atrioventricular and outflow tract cushion mesenchyme and a small area of interventricular myocardium. Tbx3, SOX4, and SOX2 ChIP data, identify a region in intron 1 of Gja1 bound by all tree proteins and subsequent ChIP experiments verify that this sequence is bound, in vivo, in the developing heart. In a luciferase reporter assay, this element displays a synergistic antagonistic response to co-transfection of Tbx3 and Sox4 and in vivo, in zebrafish, drives expression of a reporter in the heart, confirming its function as a cardiac enhancer. Mechanistically, we postulate that Sox4 is a mediator of Tbx3 transcriptional activity.


Asunto(s)
Conexina 43/genética , Regulación de la Expresión Génica , Factores de Transcripción SOXC/metabolismo , Proteínas de Dominio T Box/metabolismo , Transcripción Genética , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Factores de Transcripción SOXC/química , Pez Cebra
10.
Carcinogenesis ; 31(7): 1298-307, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20400479

RESUMEN

BACKGROUND AND AIMS: The underlying molecular mechanisms of hepatocellular carcinoma (HCC) remain poorly understood due to its complex development process. The human T cell-specific transcription factor sex-determining region Y-related high-mobility group (HMG) box 4 (SOX4) has been linked to development and tumorigenesis. In this study, we characterized the roles of SOX4 in regulation of the p53 transcription activity and evaluated the expression patterns and prognostic value of the transcription factor SOX4 in HCC. METHODS: The expression levels of human SOX4 were examined in HCC samples obtained from 58 patients having curative partial hepatectomy. The interaction and effects of SOX4 on the p53 pathway were assessed in HCC cell lines. Luciferase reporter assay to examine p53-mediated transcription of target genes was performed. The association of SOX4 expression level with tumor recurrence and overall survival was evaluated. RESULTS: We showed that the HMG box domain of SOX4 interacted with p53, resulting in the inhibition of p53-mediated transcription by the Bax promoter. More importantly, SOX4 overexpression led to a significant repression of p53-induced Bax expression and subsequent repression of p53-mediated apoptosis induced by gamma-irradiation. In clinicopathological analysis, nuclear overexpression of SOX4 was observed in 37 out of 58 (63.8%) HCC samples, and this correlated with diminished risk of recurrence (P = 0.014) and improved overall survival time (P = 0.045) in HCC patients. CONCLUSION: These results suggest that SOX4 contributes to hepatocarcinogenesis by inhibiting p53-mediated apoptosis and that its overexpression might be a useful prognostic marker for survival after surgical resection.


Asunto(s)
Carcinoma Hepatocelular/etiología , Neoplasias Hepáticas/etiología , Factores de Transcripción SOXC/fisiología , Proteína p53 Supresora de Tumor/fisiología , Apoptosis , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/radioterapia , Células Hep G2 , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/radioterapia , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Factores de Transcripción SOXC/análisis , Factores de Transcripción SOXC/química , Activación Transcripcional
11.
Int J Biochem Cell Biol ; 42(3): 425-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19651233

RESUMEN

Sox4, Sox11 and Sox12 constitute the group C of Sry-related HMG box proteins. They are co-expressed in embryonic neuronal progenitors and in mesenchymal cells in many developing organs. More closely related to each other than to any other proteins, they nevertheless bind DNA and activate transcription in vitro with different efficiencies. Sox4-null embryos and Sox11-null newborns die from heart malformations and the latter display widespread defects, while Sox12-null mice are viable and do not show obvious malformations. Sox4 facilitates differentiation of lymphocytes, pancreatic beta cells, osteoblasts and acts in redundancy with Sox11 to promote neuronal differentiation. Sox4 and Sox11 are upregulated in many tumor types in humans, where their roles in cell survival, proliferation, and metastasis remain controversial. Together, these data hint that Sox4 and Sox11 regulate cell differentiation, proliferation and survival in multiple essential processes, and suggest that they may act in redundancy to control many more developmental, physiological and pathological processes than currently known.


Asunto(s)
Desarrollo Embrionario , Neoplasias/metabolismo , Factores de Transcripción SOXC/metabolismo , Animales , Humanos , Neoplasias/genética , Factores de Transcripción SOXC/química , Factores de Transcripción SOXC/genética
12.
Hum Mol Genet ; 18(7): 1343-52, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19153074

RESUMEN

The search for oncogenes is becoming increasingly important in cancer genetics because they are suitable targets for therapeutic intervention. To identify novel oncogenes, activated by gene amplification, we analyzed cDNA microarrays by high-resolution comparative genome hybridization and compared DNA copy number and mRNA expression levels in lung cancer cell lines. We identified several amplicons (5p13, 6p22-21, 11q13, 17q21 and 19q13) that had a concomitant increase in gene expression. These regions were also found to be amplified in lung primary tumours. We mapped the boundaries and measured expression levels of genes within the chromosome 6p amplicon. The Sry-HMG box gene SOX4 (sex-determining region Y box 4), which encodes a transcription factor involved in embryonic cell differentiation, was overexpressed by a factor of 10 in cells with amplification relative to normal cells. SOX4 expression was also stronger in a fraction of lung primary tumours and lung cancer cell lines and was associated with the presence of gene amplification. We also found variants of SOX4 in lung primary tumours and cancer cell lines, including a somatic mutation that introduced a premature stop codon (S395X) at the serine-rich C-terminal domain. Although none of the variants increased the transactivation ability of SOX4, overexpression of the wildtype and of the non-truncated variants in NIH3T3 cells significantly increased the transforming ability of the weakly oncogenic RHOA-Q63L. In conclusion, our results show that, in lung cancer, SOX4 is overexpressed due to gene amplification and provide evidence of oncogenic properties of SOX4.


Asunto(s)
Cromosomas Humanos Par 6/genética , Amplificación de Genes , Neoplasias Pulmonares/genética , Factores de Transcripción SOXC/genética , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Mutación/genética , Células 3T3 NIH , Factores de Transcripción SOXC/química , Activación Transcripcional/genética
13.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 24(5): 505-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17922414

RESUMEN

OBJECTIVE: To investigate the mutation of SOX4 gene in the different tumor tissues with pathological stages and types of non-small cell lung cancer (NSCLC), and to explore its roles in the progression of lung carcinoma. METHODS: The SOX4 gene HMG-box of lung cancer tissues and paracancerous tissues were amplified by PCR, 20 cases shown difference by single strand conformation polymorphyism analysis were sequenced. The DNA sequences were compared with normal sequences by software Clustal and DNAStar. RESULTS: In the 90 NSCLCs, 18 cases were found with mutations of SOX4 gene and were sequenced, and there were 2 mutational points. Seven were detected from squamous cell carcinoma, five from adenocarcinoma and six from adeno-squamous. Three were obtained from tissues in stage I, five in stage II, six in stage III, and four in stage IV. The mutation rate in stage II, III and IV was significantly higher than that in stage I. CONCLUSION: SOX4 gene mutation is not associated with pathology histological types of tumor, but it is significantly associated with pathological stages and the mutation rate increases gradually, which has relation with advanced pathological stages in NSCLC. The results indicate that the SOX4 gene mutations might be related in the lung carcinogenesis and tumor metastasis. The study also provides molecular data for study the links between the mutation of SOX gene and human oncogenesis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mutación , Factores de Transcripción SOXC/genética , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Secuencia de Bases , Femenino , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Polimorfismo Conformacional Retorcido-Simple , Factores de Transcripción SOXC/química , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...