Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sex Dev ; 6(5): 223-30, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22797524

RESUMEN

In an effort to identify novel candidate genes involved in testis determination, we previously used suppression subtraction hybridisation PCR on male and female whole embryonic (12.0-12.5 days post coitum) mouse gonads. One gene to emerge from our screen was Redd1. In the current study, we demonstrate by whole-mount in situ hybridisation that Redd1 is differentially expressed in the developing mouse gonad at the time of sex determination, with higher expression in testis than ovary. Furthermore, Redd1 expression was first detected as Sry expression peaks, immediately prior to morphological sex determination, suggesting a potential role for Redd1 during testis development. To determine the functional importance of this gene during testis development, we generated Redd1-deficient mice. Morphologically, Redd1-deficient mice were indistinguishable from control littermates and showed normal fertility. Our results show that Redd1 alone is not required for testis development or fertility in mice. The lack of a male reproductive phenotype in Redd1 mice may be due to functional compensation by the related gene Redd2.


Asunto(s)
Reproducción/fisiología , Testículo/embriología , Factores de Transcripción/metabolismo , Animales , Biomarcadores/metabolismo , Cruzamientos Genéticos , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Femenino , Fertilidad , Feto/embriología , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Fenotipo , Reacción en Cadena de la Polimerasa , Reproducibilidad de los Resultados , Células de Sertoli/metabolismo , Testículo/citología , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
2.
Sex Dev ; 5(1): 7-15, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21099207

RESUMEN

Mammalian sex determination is a dynamic process involving balanced gene expression leading to the development of either a testis or an ovary. Candidate sex-determining genes have been identified through microarray-based studies of gonadal gene expression; however, few methods exist for validation. This study describes a new technique for transfecting gonads using nucleofection. Fifteen micrograms of expression plasmid DNA was transfected into E11.5 gonads, cultured for 3 days and gene expression analyzed. Following optimization, we consistently achieved cell transfection efficiencies of 11% of cells using pMax-GFP plasmid. To test the applicability of nucleofection to studies of gene function, a testis-determining gene was transfected into gonads and its ability to sex-reverse was examined. When Sry was transfected into female (XX) gonads, upregulation of its target gene Sox9 was observed, as well as a downregulation of the ovarian gene Foxl2. Conversely, when shSox9 was introduced into male (XY) gonads, reduction of Sox9 and its target gene, Amh was observed, with a concomitant upregulation of Foxl2. Nucleofection-based gene delivery can recapitulate in vivo events of gonadal development that demonstrates 'proof-of-principle' of the method as a screening tool to evaluate the cellular function of potential sex-determining and gonadal differentiation genes.


Asunto(s)
Ovario/embriología , Ovario/metabolismo , Procesos de Determinación del Sexo/genética , Diferenciación Sexual/genética , Testículo/embriología , Testículo/metabolismo , Animales , Técnicas de Cultivo de Embriones , Femenino , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes sry , Proteínas Fluorescentes Verdes/genética , Masculino , Ratones , Proteínas Recombinantes de Fusión/genética , Factor de Transcripción SOX9/antagonistas & inhibidores , Factor de Transcripción SOX9/genética , Proteína de la Región Y Determinante del Sexo/genética , Transfección
3.
Sex Dev ; 4(6): 321-5, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20838034

RESUMEN

During mouse sex determination, SRY upregulates the core testis-specific enhancer of Sox9, TESCO. Mutations in human SRY are found in one third of cases with XY pure gonadal dysgenesis (XY GD; Swyer syndrome), while two thirds remain unexplained. Heterozygous SOX9 mutations can cause XY GD in association with the skeletal malformation syndrome campomelic dysplasia. We hypothesized that human TESCO mutations could cause isolated XY GD. Sixty-six XY GD cases with an intact SRY were analyzed for TESCO point mutations or deletions. No mutations were identified. We conclude that TESCO mutations are not a common cause of XY GD.


Asunto(s)
Disgenesia Gonadal 46 XY/genética , Proteínas de Homeodominio/genética , Mutación/genética , Proteína de la Región Y Determinante del Sexo/genética , Proteínas Supresoras de Tumor/genética , Animales , Proteínas del Citoesqueleto , Humanos , Proteínas con Dominio LIM , Masculino , Ratones , Proteínas de Unión al ARN
4.
Sex Dev ; 3(4): 194-204, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19752599

RESUMEN

In biological research, quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) assays are commonly employed to study mRNA abundance in cells and tissues. This type of assay usually relies on assessing transcript abundance relative to constitutively expressed endogenous reference genes. Therefore, it is important that the reference genes themselves are stably expressed in the cells or tissues analyzed, independent of factors such as age, sex, disease or experimental manipulations. Since no gene is expressed at the same level in all cells at all times, suitable reference genes must be identified for the specific cellular system or tissue being investigated. Here, we sought to identify stably expressed endogenous reference genes during embryonic gonad development in the mouse. We measured the transcript abundance of 10 frequently employed normalizing genes, of which 4 were stably expressed in fetal gonads from 11.5 to 14.5 dpc irrespective of sex. Based on our analysis, we suggest that Rn18s, Rps29, Tbp and Sdha are suitable reference genes for qRT-PCR expression studies during early gonad differentiation in the mouse.


Asunto(s)
Feto/metabolismo , Regulación del Desarrollo de la Expresión Génica , Gónadas/embriología , Gónadas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/normas , Animales , Cartilla de ADN/análisis , Electroforesis en Gel de Agar , Perfilación de la Expresión Génica , Mesonefro/embriología , Mesonefro/metabolismo , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estándares de Referencia
5.
Sex Dev ; 1(2): 114-26, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18391522

RESUMEN

The study of the mammalian sex-determining pathway has been hampered by the lack of cell culture systems to investigate the underlying molecular relationships between sex-determining genes. Recent approaches using high-throughput genome-wide studies have revealed a number of sexually dimorphic genes expressed in the developing mouse gonad. Here, we investigated a human testicular cell line in terms of its expression of known sex-determining genes and newly identified candidates. The human embryonal carcinoma cell line NT2/D1 was screened for the expression of 46 genes with known or potential roles in the sex-determining and differentiation pathway. Forty genes tested were expressed in NT2/D1 cells including the testis-determining genes SRY, SOX9, SF-1, DHH and FGF9. Genes not expressed included WT1, DAX1 and the ovary-specific genes FOXL2 and WNT4. Cell-specific markers demonstrate that NT2/D1 cells reflect a number of cell types in the gonad including Sertoli, Leydig and germ cells. Our results suggest that male pathways initiated by SRY, SOX9 and SF-1 remain intact in these cells. Lack of expression of ovary-specific genes is consistent with a commitment of these cells to the male lineage. Manipulation of gene expression in this cell line could be an important new in vitro tool for the discovery of new human sex-determining genes.


Asunto(s)
Perfilación de la Expresión Génica , Sistema Urogenital/metabolismo , Carcinoma Embrionario/genética , Carcinoma Embrionario/patología , Línea Celular Tumoral , Femenino , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células Germinativas/metabolismo , Gónadas/metabolismo , Humanos , Células Intersticiales del Testículo/metabolismo , Masculino , Ovario/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Sertoli/metabolismo , Procesos de Determinación del Sexo , Diferenciación Sexual/genética , Transducción de Señal/genética , Testículo/metabolismo , Sistema Urogenital/embriología
6.
Cytogenet Genome Res ; 101(3-4): 185-98, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14684982

RESUMEN

The decision of the bi-potential gonad to develop into either a testis or ovary is determined by the presence or absence of the Sex-determining Region gene on the Y chromosome (SRY). Since its discovery, almost 13 years ago, the molecular role that SRY plays in initiating the male sexual development cascade has proven difficult to ascertain. While biochemical studies of clinical mutants and mouse genetic models have helped in our understanding of SRY function, no direct downstream targets of SRY have yet been identified. There are, however, a number of other genes of equal importance in determining sexual phenotype, expressed before and after expression of SRY. Of these, one has proven of central importance to mammals and vertebrates, SOX9. This review describes our current knowledge of SRY and SOX9 structure and function in the light of recent key developments.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Proteínas del Grupo de Alta Movilidad/fisiología , Mamíferos/genética , Proteínas Nucleares , Procesos de Determinación del Sexo , Factores de Transcripción/fisiología , Animales , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Genes sry , Proteínas del Grupo de Alta Movilidad/química , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Masculino , Ratones , Modelos Genéticos , Mutación , Factor de Transcripción SOX9 , Proteína de la Región Y Determinante del Sexo , Factores de Transcripción/química , Factores de Transcripción/genética
7.
J Dent Res ; 82(9): 725-30, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12939358

RESUMEN

Regulatory factors governing the formation of bone in the glenoid fossa in response to functional appliance therapy have not been identified. Therefore, the purpose of this study was to investigate the temporal pattern of expression of two key chondrogenesis markers-SOX9 and its target gene, type II collagen-in the glenoid fossa by immunostaining in a 35-day-old Sprague Dawley rat model during both natural growth and forward mandibular positioning. The expression of both factors was up-regulated when the mandible was positioned forward, indicating an enhancement of chondrocyte differentiation and chondroid matrix formation. Our results indicate that chondroid bone formation in the glenoid fossa in response to forward mandibular positioning is regulated by molecular markers indicative of endochondral ossification.


Asunto(s)
Colágeno Tipo II/análisis , Proteínas del Grupo de Alta Movilidad/análisis , Mandíbula/anatomía & histología , Aparatos Ortodóncicos Funcionales , Hueso Temporal/metabolismo , Factores de Transcripción/análisis , Regulación hacia Arriba , Análisis de Varianza , Animales , Diferenciación Celular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Colágeno Tipo II/genética , Matriz Extracelular/efectos de los fármacos , Femenino , Proteínas del Grupo de Alta Movilidad/genética , Mandíbula/crecimiento & desarrollo , Modelos Animales , Osteogénesis/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factor de Transcripción SOX9 , Hueso Temporal/crecimiento & desarrollo , Factores de Transcripción/genética , Regulación hacia Arriba/genética
8.
Brain Res ; 906(1-2): 143-8, 2001 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-11430871

RESUMEN

Human mutations in the transcription factor gene, SOX9, cause campomelic dysplasia (CD), a severe dwarfism associated with brain abnormalities including dilation of lateral ventricles, hypoplasia of the corpus callosum and cerebellum defects. To improve our understanding of how SOX9 contributes to the molecular genetic pathway of brain development we sought to investigate the distribution of SOX9 protein in rat and mouse brain. The regions of SOX9 expression identified in this study correlated with the sites of reported brain abnormalities in CD patients. SOX9 immunoreactivity was observed in nuclei of scattered cells throughout the brain, in the ependymal layer and cells of the choroid plexus. In the forebrain most SOX9-immunoreactive nuclei co-localised with the glial astrocyte marker S-100. In the cerebellum, SOX9 was observed mostly in cells surrounding Purkinje cells, which were identified, by electron microscopy, as Golgi epithelial cells, also known as Bergmann glia. Using SOX9 antibody as a marker for the precursors of Bergmann glia, we traced their origin during mouse development. At embryonic day (E)14.5 and E16.5, SOX9 immunoreactivity was present mainly in the primordial choroid plexus, and ventricular zone. By E18.5, SOX9 was observed in the granular cell and Purkinje cell layers but no labelling was detectable in the external granular layer. These results suggest that SOX9 immunoreactivity is a marker for Bergmann cells during development and favour the proposed origin of the secondary glial scaffold arising from Bergmann cells derived exclusively from the ventricular zone.


Asunto(s)
Diferenciación Celular/fisiología , Cerebelo/embriología , Cerebelo/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Malformaciones del Sistema Nervioso/metabolismo , Neuroglía/metabolismo , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Cerebelo/ultraestructura , Plexo Coroideo/embriología , Plexo Coroideo/metabolismo , Plexo Coroideo/ultraestructura , Epéndimo/embriología , Epéndimo/metabolismo , Epéndimo/ultraestructura , Femenino , Feto , Inmunohistoquímica , Masculino , Ratones , Microscopía Electrónica , Malformaciones del Sistema Nervioso/patología , Malformaciones del Sistema Nervioso/fisiopatología , Neuroglía/ultraestructura , Ratas , Factor de Transcripción SOX9 , Células Madre/ultraestructura
9.
FEBS Lett ; 496(2-3): 75-80, 2001 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-11356186

RESUMEN

The campomelic dysplasia/autosomal sex reversal protein SOX9 is an important developmental transcription factor, required for correct bone and testis formation. Through in vitro and in vivo studies we have identified the heat shock protein HSP70 as an interacting partner for SOX9 in chondrocyte and testicular cell lines. HSP70 forms a ternary complex with DNA-bound SOX9. The interaction between HSP70 and SOX9 is ATP-independent and involves a highly conserved region of SOX9 hitherto of unknown function and the C-terminal region of HSP70. Our results implicate HSP70-SOX9 interactions in the assembly of multi-protein complexes during SOX9-mediated transcription.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Factores de Transcripción/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Sitios de Unión , Western Blotting , Huesos/embriología , Línea Celular , Condrocitos/metabolismo , Electroforesis en Gel de Poliacrilamida , Eliminación de Gen , Humanos , Inmunohistoquímica , Masculino , Mutagénesis , Pruebas de Precipitina , Unión Proteica , Ratas , Proteínas Recombinantes/metabolismo , Factor de Transcripción SOX9 , Procesos de Determinación del Sexo , Testículo/embriología , Transcripción Genética , Activación Transcripcional , Transfección , Células Tumorales Cultivadas
10.
Mol Genet Metab ; 72(4): 356-9, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11286511

RESUMEN

SOX13 is a member of the SOX family of transcription factors that encodes the type 1 diabetes autoantigen, ICA12. The SOX13 gene maps at chromosome 1q31.3-32.1 near a region containing a susceptibility locus for type 1 diabetes. SOX13 was assessed as a candidate susceptibility gene. Analysis of the SOX13 gene identified a number of single nucleotide polymorphisms and a polymorphic CA dinucleotide repeat. Linkage and association studies indicate that SOX13 is unlikely to make a substantial contribution to type 1 diabetes susceptibility.


Asunto(s)
Autoantígenos/genética , Diabetes Mellitus Tipo 1/genética , Proteínas del Grupo de Alta Movilidad/genética , Mapeo Cromosómico , Cromosomas Humanos Par 1/genética , ADN/metabolismo , Familia , Frecuencia de los Genes , Genotipo , Humanos , Polimorfismo de Nucleótido Simple , Factores de Transcripción SOXD
11.
J Biol Chem ; 276(30): 27864-72, 2001 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-11323423

RESUMEN

Human mutations in the transcription factor SOX9 cause campomelic dysplasia/autosomal sex reversal. Here we identify and characterize two novel heterozygous mutations, F154L and A158T, that substitute conserved "hydrophobic core" amino acids of the high mobility group domain at positions thought to stabilize SOX9 conformation. Circular dichroism studies indicated that both mutations disrupt alpha-helicity within their high mobility group domain, whereas tertiary structure is essentially maintained as judged by fluorescence spectroscopy. In cultured cells, strictly nuclear localization was observed for wild type SOX9 and the F154L mutant; however, the A158T mutant showed a 2-fold reduction in nuclear import efficiency. Importin-beta was demonstrated to be the nuclear transport receptor recognized by SOX9, with both mutant proteins binding importin-beta with wild type affinity. Whereas DNA bending was unaffected, DNA binding was drastically reduced in both mutants (to 5% of wild type activity in F154L, 17% in A158T). Despite this large effect, transcriptional activation in cultured cells was only reduced to 26% in F154L and 62% in A158T of wild type activity, suggesting that a small loss of SOX9 transactivation activity could be sufficient to disrupt proper regulation of target genes during bone and testis formation. Thus, clinically relevant mutations of SOX9 affect protein structure leading to compound effects of reduced nuclear import and reduced DNA binding, the net effect being loss of transcriptional activation.


Asunto(s)
Anomalías Múltiples/genética , Transporte Activo de Núcleo Celular/genética , Huesos/anomalías , ADN/metabolismo , Trastornos del Desarrollo Sexual , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Mutación , Mutación Puntual , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Adulto , Secuencia de Aminoácidos , Animales , Células COS , Núcleo Celular/metabolismo , Células Cultivadas , Dicroismo Circular , Electroforesis en Gel de Poliacrilamida , Femenino , Genes Dominantes , Heterocigoto , Proteínas del Grupo de Alta Movilidad/química , Humanos , Inmunohistoquímica , Recién Nacido , Carioferinas , Cariotipificación , Cinética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Nucleares/metabolismo , Fenotipo , Polimorfismo Conformacional Retorcido-Simple , Unión Proteica , Conformación Proteica , Desnaturalización Proteica , Estructura Terciaria de Proteína , Factor de Transcripción SOX9 , Análisis de Secuencia de ADN , Espectrometría de Fluorescencia , Relación Estructura-Actividad , Temperatura , Factores de Transcripción/química , Transfección , Triptófano/metabolismo
12.
Dev Biol ; 229(2): 319-26, 2001 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11150238

RESUMEN

Although sex determination starts in the gonads, this may not be the case for species with temperature sex determination (TSD). Since temperature affects the whole embryo, extragonadal thermosensitive cells may produce factors that induce gonadal sex determination as a secondary event. To establish if gonads of a species with TSD respond directly to temperature, pairs of gonads were cultured, one at female-promoting temperature (FPT) and the contralateral at male-promoting temperature (MPT). Histological and immunohistochemical detection of SOX9 revealed that the response to temperature of isolated gonads was similar to that of the gonads of whole embryos. While gonads cultured at MPT maintained SOX9 expression, it was downregulated in gonads at FPT. Downregulation of SOX9 took longer in gonads cultured at stage 23 than in gonads cultured at stage 24, suggesting that a developmental clock was already established at the onset of culture. To find out if sex commitment occurs in vitro, gonads were switched from FPT to MPT at different days. Results showed that the ovarian pathway was established after 4 days of culture. The present demonstration that gonads have an autonomous temperature detector that regulates SOX9 expression provides a useful starting point from which the molecular pathways underlying TSD can be elucidated.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas del Grupo de Alta Movilidad/genética , Ovario/embriología , Diferenciación Sexual , Testículo/embriología , Factores de Transcripción/genética , Tortugas/embriología , Secuencia de Aminoácidos , Animales , Epítopos/química , Femenino , Proteínas del Grupo de Alta Movilidad/análisis , Inmunohistoquímica , Masculino , Datos de Secuencia Molecular , Técnicas de Cultivo de Órganos , Factor de Transcripción SOX9 , Temperatura , Factores de Transcripción/análisis
14.
Diabetes ; 49(4): 555-61, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10871192

RESUMEN

The SOX (sex-determining region [SRY]-type high mobility group [HMG] box) family of transcription factors play key roles in determining cell fate during organ development. In this study, we have identified a new human SOX gene, SOX13, as encoding the type 1 diabetes autoantigen, islet cell antigen 12 (ICA12). Sequence analysis showed that SOX13 belongs to the class D subgroup of SOX transcription factors, which contain a leucine zipper motif and a region rich in glutamine. SOX13 autoantibodies occurred at a significantly higher frequency among 188 people with type 1 diabetes (18%) than among 88 with type 2 diabetes (6%) or 175 healthy control subjects (4%). Deletion mapping of the antibody epitopes showed that the autoantibodies were primarily directed against an epitope requiring the majority of the protein. SOX13 RNA was detected in most human tissues, with the highest levels in the pancreas, placenta, and kidney. Immunohistochemistry on sections of human pancreas identified SOX13 in the islets of Langerhans, where staining was mostly cytoplasmic. In mouse pancreas, Sox13 was present in the nucleus and cytoplasm of beta-cells as well as other islet cell types. Recombinant SOX13 protein bound to the SOX consensus DNA motif AACAAT, and binding was inhibited by homodimer formation. These observations-along with the known molecular interactions of the closely related protein, rainbow trout Sox23-suggest that SOX13 may be activated for nuclear import and DNA binding through heterodimer formation. In conclusion, we have identified ICA12 as the putative transcription factor SOX13 and demonstrated an increased frequency of autoantibody reactivity in sera from type 1 diabetic subjects compared with type 2 diabetic and healthy control subjects.


Asunto(s)
Autoantígenos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/inmunología , Islotes Pancreáticos/inmunología , Secuencia de Aminoácidos , Animales , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Autoantígenos/genética , ADN/metabolismo , Dimerización , Expresión Génica , Proteínas del Grupo de Alta Movilidad/química , Humanos , Leucina Zippers , Ratones , Datos de Secuencia Molecular , Especificidad de Órganos , ARN Mensajero/análisis , Proteínas Recombinantes/metabolismo , Factores de Transcripción SOXD
15.
Gene ; 250(1-2): 181-9, 2000 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-10854791

RESUMEN

SOX13 is the member of the SOX (Sry related HMG BOX) family of transcription factors which encodes the type-1 diabetes autoantigen, ICA12, and is expressed in a number of tissues including pancreatic islets and arterial walls. By fluorescence in situ hybridisation, radiation hybrid mapping and YAC analysis we determined that the human SOX13 gene maps to Chromosome 1q31.3-32.1 near the marker D1S504, a region associated with type-1 diabetes susceptibility and familial dilated cardiomyopathy. Mouse Sox13 maps to the syntenic region near the marker D1Mit57. The human SOX13 gene spans >15.5kb of genomic DNA and is composed of 14 exons with introns interrupting regions encoding the HMG DNA binding domain and the leucine zipper/glutamine-rich dimerisation domain. Comparison with the mouse Sox13 gene suggests the existence of long and short forms of the SOX13 protein which may arise by differential splicing during different stages in embryogenesis. The high sequence conservation between human SOX13 and mouse, Xenopus and trout orthologues implies a conserved function in vertebrates. SOX13 belongs to SOX Group D members which contain a leucine zipper/glutamine-rich region. Phylogenetic analyses of SOX proteins suggest that such domains were acquired after the initial divergence of groups A to G.


Asunto(s)
Autoantígenos , Genes/genética , Proteínas del Grupo de Alta Movilidad/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Mapeo Contig , ADN/química , ADN/genética , ADN/aislamiento & purificación , Exones , Humanos , Células Híbridas , Intrones , Ratones , Datos de Secuencia Molecular , Filogenia , Factores de Transcripción SOXD , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido
16.
Histochem Cell Biol ; 113(1): 31-6, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10664067

RESUMEN

We studied the location of Sox9 protein in the embryonic, juvenile, and adult rat testis by immunohistochemistry and immunoblotting. Sox9 belongs to a family of Sox proteins that are transcription factors and important in several developmental processes. In the incipient embryonic testis, Sox9 was prominently present in the gonadal blastema. With further embryonic differentiation, Sox9-positive cells arranged in the periphery of the testicular cords, showing the location of the Sertoli cells. Thereafter the immunoreaction for Sox9 gradually declined and was only weakly detectable in the 2-day-old postnatal rat testis. This situation remained for some period of time. In the 15-day-old rat testis, Sox9 protein strongly reappeared in the testicular cords. In the adult, the Sertoli cells of most regions of the seminiferous tubules were positive for Sox9. The strongest reaction for Sox9 was found in the dark zone. However, clearly negative or only weakly positive spermatogenic stages for the protein were also found, as seen for example in the pale zone. In fertile 1-year-old rats this basic situation was still detectable. Analyzed rat ovaries were all negative for Sox9, showing the sex-specific nature of Sox9. The results showed that Sox9 protein is distinctly present in the developing and mature Sertoli cells, but that its presence and amount is dependent on the age and the spermatogenetic stage within the seminiferous tubuli. The prominent presence of Sox9 in the incipient testis and at puberty suggests that this protein is needed at important phases of aggregation and reorganization of the Sertoli cells. The age and stage-specific presence of Sox9 in the testicular cords and in the seminiferous tubules of the adult suggests that Sox9 also may have a pivotal role in germ cell differentiation.


Asunto(s)
Envejecimiento/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Células de Sertoli/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/fisiología , Colágeno/análisis , Femenino , Proteínas del Grupo de Alta Movilidad/fisiología , Masculino , Ovario/embriología , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Embarazo , Ratas , Ratas Wistar , Factor de Transcripción SOX9 , Túbulos Seminíferos/embriología , Túbulos Seminíferos/crecimiento & desarrollo , Túbulos Seminíferos/metabolismo , Células de Sertoli/fisiología , Diferenciación Sexual/fisiología , Espermatogénesis/fisiología , Factores de Transcripción/fisiología
17.
J Exp Zool ; 284(6): 705-10, 1999 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-10531557

RESUMEN

In mouse and chick embryos, the SOX9 gene is down-regulated in genetic females whereas in genetic males it remains in the Sertoli cells. We studied the distribution of SOX9 protein in developing genital ridges of embryos of the sea turtle Lepidochelys olivacea incubated at male- or female-promoting temperatures, using the antibody for detection. At stages 22-24, cells in medullary cords show SOX9 positive nuclei, while coelomic epithelial cells appear negative. At stage 25 however, most medullary cells are SOX9 negative and at the female-promoting temperature, and from stage 26 onwards, SOX9 protein is not detected. At the male-promoting temperature, medullary cords remain SOX9-positive at all stages. These results suggest that SOX9 is up-regulated in Sertoli cells irrespective of primary sex-determining switch. Sex is irreversibly determined at stage 24 or 26 at the male- or female-promoting temperature, respectively (Merchant-Larios et al.,'97). The present results suggest that there is a correlation between SOX9 expression and sex determination in the olive ridley. At the male-promoting temperature, Sertoli cells expressing SOX9 become committed at stage 24 and male sex is determined, whereas at the female-promoting temperature, SOX9 is down-regulated at stage 26 and female sex is determined. J. Exp. Zool. 284:705-710, 1999.


Asunto(s)
Embrión no Mamífero/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Ovario/metabolismo , Diferenciación Sexual/fisiología , Testículo/metabolismo , Factores de Transcripción/metabolismo , Tortugas/metabolismo , Secuencia de Aminoácidos , Animales , Embrión no Mamífero/embriología , Femenino , Regulación de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/genética , Masculino , Datos de Secuencia Molecular , Ovario/embriología , Factor de Transcripción SOX9 , Procesos de Determinación del Sexo , Diferenciación Sexual/genética , Temperatura , Testículo/embriología , Factores de Transcripción/genética
18.
Nucleic Acids Res ; 27(5): 1359-64, 1999 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-9973626

RESUMEN

SOX (SRY-related HMG box) proteins are transcription factors that have critical roles in the regulation of numerous developmental processes. They share at least 50% homology in their HMG domains, which bind the DNA element AACAAT. How different SOX proteins achieve specific regulation of target genes is not known. We determined the DNA-binding specificity of SOX9 using a random oligonucleotide selection assay. The optimal SOX9 binding sequence, AGAACAATGG, contained a core DNA-binding element AACAAT, flanked by 5' AG and 3' GG nucleotides. The specific interaction between SOX9 and AGAACAATGG was confirmed by mobility shift assays, DNA competition and dissociation studies. The 5' AG and 3' GG flanking nucleotides enhance binding by SOX9 HMG domain, but not by the HMG domain of another SOX factor, SRY. For SRY, different 5' and 3' flanking nucleotides are preferred. Our studies support the notion that SOX proteins achieve DNA sequence specificity through subtle preferences for flanking nucleotides and that this is likely to be dictated by signature amino acids in their HMG domains. Furthermore, the related HMG domains of SOX9 and Sox17 have similar optimal binding sites that differ from those of SRY and Sox5, suggesting that SOX factors may co-evolve with their DNA targets to achieve specificity.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Bases , Sitios de Unión , Cartilla de ADN , Factor de Transcripción SOX9
19.
Mol Cell Biol ; 17(4): 2336-46, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9121483

RESUMEN

The identification of mutations in the SRY-related SOX9 gene in patients with campomelic dysplasia, a severe skeletal malformation syndrome, and the abundant expression of Sox9 in mouse chondroprogenitor cells and fully differentiated chondrocytes during embryonic development have suggested the hypothesis that SOX9 might play a role in chondrogenesis. Our previous experiments with the gene (Col2a1) for collagen II, an early and abundant marker of chondrocyte differentiation, identified a minimal DNA element in intron 1 which directs chondrocyte-specific expression in transgenic mice. This element is also a strong chondrocyte-specific enhancer in transient transfection experiments. We show here that Col2a1 expression is closely correlated with high levels of SOX9 RNA and protein in chondrocytes. Our experiments indicate that the minimal Col2a1 enhancer is a direct target for Sox9. Indeed, SOX9 binds to a sequence of the minimal Col2a1 enhancer that is essential for activity in chondrocytes, and SOX9 acts as a potent activator of this enhancer in cotransfection experiments in nonchondrocytic cells. Mutations in the enhancer that prevent binding of SOX9 abolish enhancer activity in chondrocytes and suppress enhancer activation by SOX9 in nonchondrocytic cells. Other SOX family members are ineffective. Expression of a truncated SOX9 protein lacking the transactivation domain but retaining DNA-binding activity interferes with enhancer activation by full-length SOX9 in fibroblasts and inhibits enhancer activity in chondrocytes. Our results strongly suggest a model whereby SOX9 is involved in the control of the cell-specific activation of COL2A1 in chondrocytes, an essential component of the differentiation program of these cells. We speculate that in campomelic dysplasia a decrease in SOX9 activity would inhibit production of collagen II, and eventually other cartilage matrix proteins, leading to major skeletal anomalies.


Asunto(s)
Cartílago/metabolismo , Elementos de Facilitación Genéticos , Proteínas del Grupo de Alta Movilidad/genética , Procolágeno/genética , Factores de Transcripción/genética , Células 3T3 , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Enfermedades del Desarrollo Óseo/genética , Cartílago/citología , Diferenciación Celular/genética , Sondas de ADN/genética , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Ratones , Datos de Secuencia Molecular , Factor de Transcripción SOX9 , Transactivadores/genética , Transfección
20.
Microbiol Immunol ; 41(6): 509-12, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9251063

RESUMEN

Analysis of the NS and M genes of the archetype H6N5 influenza virus strain A/shearwater/Australia/1/72 shows it to be a typical example of the avian host reservoir, containing Old World/Eurasian internal proteins with divergent surface proteins, which is a potential source of new pandemic strains.


Asunto(s)
Genes Virales , Virus de la Influenza A/genética , Proteínas de la Matriz Viral/genética , Proteínas no Estructurales Virales/genética , Secuencia de Aminoácidos , Secuencia de Bases , ADN Complementario/genética , Brotes de Enfermedades , Reservorios de Enfermedades , Evolución Molecular , Biblioteca de Genes , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...