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










Base de datos
Intervalo de año de publicación
1.
J Bacteriol ; 198(3): 427-35, 2016 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-26553849

RESUMEN

UNLABELLED: Recent breakthroughs in next-generation sequencing technologies have led to the identification of small noncoding RNAs (sRNAs) as a new important class of regulatory molecules. In prokaryotes, sRNAs are often bound to the chaperone protein Hfq, which allows them to interact with their partner mRNA(s). We screened the genome of the zoonotic and human pathogen Brucella suis 1330 for the presence of this class of RNAs. We designed a coimmunoprecipitation strategy that relies on the use of Hfq as a bait to enrich the sample with sRNAs and eventually their target mRNAs. By deep sequencing analysis of the Hfq-bound transcripts, we identified a number of mRNAs and 33 sRNA candidates associated with Hfq. The expression of 10 sRNAs in the early stationary growth phase was experimentally confirmed by Northern blotting and/or reverse transcriptase PCR. IMPORTANCE: Brucella organisms are facultative intracellular pathogens that use stealth strategies to avoid host defenses. Adaptation to the host environment requires tight control of gene expression. Recently, small noncoding RNAs (sRNAs) and the sRNA chaperone Hfq have been shown to play a role in the fine-tuning of gene expression. Here we have used RNA sequencing to identify RNAs associated with the B. suis Hfq protein. We have identified a novel list of 33 sRNAs and 62 Hfq-associated mRNAs for future studies aiming to understand the intracellular lifestyle of this pathogen.


Asunto(s)
Brucella suis/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Proteína de Factor 1 del Huésped/metabolismo , ARN Bacteriano/metabolismo , Transcriptoma , Animales , Brucella suis/genética , Línea Celular , Regulación Bacteriana de la Expresión Génica/fisiología , Estudio de Asociación del Genoma Completo , Proteína de Factor 1 del Huésped/genética , Ratones , Chaperonas Moleculares , ARN Bacteriano/genética , ARN no Traducido
2.
Proc Natl Acad Sci U S A ; 104(41): 16335-40, 2007 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-17913892

RESUMEN

Anorexia nervosa is a growing concern in mental health, often inducing death. The potential neuronal deficits that may underlie abnormal inhibitions of food intake, however, remain largely unexplored. We hypothesized that anorexia may involve altered signaling events within the nucleus accumbens (NAc), a brain structure involved in reward. We show here that direct stimulation of serotonin (5-hydroxytryptamine, 5-HT) 4 receptors (5-HT(4)R) in the NAc reduces the physiological drive to eat and increases CART (cocaine- and amphetamine-regulated transcript) mRNA levels in fed and food-deprived mice. It further shows that injecting 5-HT(4)R antagonist or siRNA-mediated 5-HT(4)R knockdown into the NAc induced hyperphagia only in fed mice. This hyperphagia was not associated with changes in CART mRNA expression in the NAc in fed and food-deprived mice. Results include that 5-HT(4)R control CART mRNA expression into the NAc via a cAMP/PKA signaling pathway. Considering that CART may interfere with food- and drug-related rewards, we tested whether the appetite suppressant properties of 3,4-N-methylenedioxymethamphetamine (MDMA, ecstasy) involve the 5-HT(4)R. Using 5-HT(4)R knockout mice, we demonstrate that 5-HT(4)R are required for the anorectic effect of MDMA as well as for the MDMA-induced enhancement of CART mRNA expression in the NAc. Directly injecting CART peptide or CART siRNA into the NAc reduces or increases food consumption, respectively. Finally, stimulating 5-HT(4)R- and MDMA-induced anorexia were both reduced by injecting CART siRNA into the NAc. Collectively, these results demonstrate that 5-HT(4)R-mediated up-regulation of CART in the NAc triggers the appetite-suppressant effects of ecstasy.


Asunto(s)
Anorexia Nerviosa/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Núcleo Accumbens/metabolismo , Receptores de Serotonina 5-HT4/metabolismo , Animales , Anorexia Nerviosa/etiología , Anorexia Nerviosa/genética , Secuencia de Bases , Ingestión de Alimentos , Masculino , Ratones , Ratones Noqueados , N-Metil-3,4-metilenodioxianfetamina/farmacología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Núcleo Accumbens/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Receptores de Serotonina 5-HT4/deficiencia , Receptores de Serotonina 5-HT4/genética , Transducción de Señal
3.
Cancer Lett ; 255(2): 182-93, 2007 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-17532558

RESUMEN

New therapeutic strategies for ovarian cancer include the identification of involved signaling pathways that could potentially serve as a source of biomarkers for early stages of the disease. In this study, we show that the embryonic male prostaglandin D synthase (Pgds)/SOX9 pathway is expressed at both the RNA and protein levels in different types of human ovarian tumors, pointing to Pgds and SOX9 as possible diagnostic markers for ovarian carcinomas. Using ovarian cancer cell lines, we found, first, that components of the Pgds/SOX9 pathway are expressed in these cells, and second, that treatment of these cells with prostaglandin D2 (PGD2) can inhibit their growth via its DP1 receptor and induce apoptosis. Finally, using siRNA and overexpression strategies, we demonstrate that SOX9 expression is induced by PDG2 and is responsible for PDG2-mediated growth inhibition. Accordingly, as stimulating the PGD2/DP1 signal transduction pathway upregulates SOX9 expression, either activators of this pathway or DP1 agonists may be useful as new therapeutic agents.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Neoplasias Ováricas/metabolismo , Factores de Transcripción/metabolismo , Apoptosis , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Carcinoma/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Proteínas del Grupo de Alta Movilidad/análisis , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/farmacología , Lipocalinas , Masculino , Neoplasias Ováricas/patología , ARN Interferente Pequeño/farmacología , Receptores de Prostaglandina/agonistas , Factor de Transcripción SOX9 , Transducción de Señal , Factores de Transcripción/análisis , Factores de Transcripción/genética , Regulación hacia Arriba
4.
Gene Expr Patterns ; 6(7): 695-702, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16488195

RESUMEN

SOX9 is an essential activating transcription factor that plays a critical role in Sertoli cell differentiation and subsequent testis cord formation. Cytoplasmic SOX9 is present in both sexes during early gonadal embryogenesis. While in males the protein is later translocated into the nucleus of pre-Sertoli cells, its expression is rapidly turned off in females. In mammalian male gonads, SOX9 activates the expression of anti-Müllerian hormone (AMH), a male hormone that initiates Müllerian ducts regression and that is also expressed in postnatal ovarian follicles. Here, we confirm that the SOX9 protein is not present in the immature ovary but also show that SOX9 is transiently expressed in the mature ovary depending on the follicular cycle. Indeed, SOX9 protein was found in the nuclear compartment of the inner cells of the theca interna cell layer which surrounds the pre-antral/antral follicles. In contrast, no expression was detected in the AMH expressing granulosa cells. While these findings exclude the possibility that SOX9 regulates AMH expression in the ovary, they show that SOX9 could nevertheless play a role in the developing follicle.


Asunto(s)
Proteínas del Grupo de Alta Movilidad/análisis , Proteínas del Grupo de Alta Movilidad/genética , Folículo Ovárico/química , Folículo Ovárico/fisiología , Ovario/química , Factores de Transcripción/análisis , Factores de Transcripción/genética , Animales , Femenino , Regulación de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/inmunología , Ratones , Ovario/fisiología , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción SOX9 , Células Tecales/química , Factores de Transcripción/inmunología
5.
J Biol Chem ; 280(46): 38625-30, 2005 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-16166090

RESUMEN

In mammals, male sex determination is controlled by the SRY protein, which drives differentiation of the bipotential embryonic gonads into testes by activating the Sertoli cell differentiation program. The morphological effects of SRY are well documented; however, its molecular mechanism of action remains unknown. Moreover, SRY proteins display high sequence variability among mammalian species, which makes protein motifs difficult to delineate. We previously isolated SIP-1/NHERF2 as a human SRY-interacting protein. SIP-1/NHERF2, a PDZ protein, interacts with the C-terminal extremity of the human SRY protein. Here we showed that the interaction of SIP-1/NHERF2 and SRY via the SIP-1/NHERF2 PDZ1 domain is conserved in mice. However, the interaction occurs via a domain that is internal to the mouse SRY protein and involves a different recognition mechanism than human SRY. Furthermore, we show that mouse and human SRY induce nuclear accumulation of the SIP-1/NHERF2 protein in cultured cells. Finally, a transgenic mouse line expressing green fluorescent protein under the control of the mouse Sry promoter allowed us to show that SRY and SIP-1/NHERF2 are co-expressed in the nucleus of pre-Sertoli cells during testis determination. Taken together, our results suggested that the function of SIP-1/NHERF2 as an SRY cofactor during testis determination is conserved between human and mouse.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Proteínas del Tejido Nervioso/fisiología , Proteína de la Región Y Determinante del Sexo/metabolismo , Secuencias de Aminoácidos , Animales , Diferenciación Celular , Línea Celular , Secuencia Conservada , Proteínas del Citoesqueleto/metabolismo , Femenino , Glutatión Transferasa/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunoprecipitación , Masculino , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Células 3T3 NIH , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , Proteínas de Unión al ARN , Proteínas Recombinantes/química , Células de Sertoli/citología , Intercambiadores de Sodio-Hidrógeno , Especificidad de la Especie , Fracciones Subcelulares , Testículo/metabolismo , Transfección
6.
Exp Cell Res ; 309(2): 468-75, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16087173

RESUMEN

SOX9 is a sex-determining factor which induces Sertoli cell differentiation and subsequent testis cord formation. It is expressed both in male and female undifferentiated gonads in the cytoplasmic compartment of pre-Sertoli cells. At the time of sexual differentiation, SOX9 moves into the nucleus of male pre-Sertoli cells whereas in female, it remains in the cytoplasm and then its expression decreases. To study the cytoplasmic localization of SOX9, we have analyzed its interaction with the cytoskeleton components. By treatment of NT2/D1 and transfected NIH3T3 cell lines and embryonic gonads with nocodazole, a drug depolymerizing the microtubules, we show that cytoplasmic retention of SOX9 requires the integrity of the microtubule network. Using biochemical experiments, we demonstrated that SOX9 is able to interact with microtubules in vitro and in vivo. On the other hand, we observed a complete male-specific reorganization of the microtubule network in epithelial Sertoli cells of the male embryonic gonad at the time of sexual differentiation and testis cord formation.


Asunto(s)
Proteínas del Grupo de Alta Movilidad/metabolismo , Microtúbulos/metabolismo , Procesos de Determinación del Sexo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/fisiología , Línea Celular Tumoral , Femenino , Humanos , Masculino , Ratones , Células 3T3 NIH , Técnicas de Cultivo de Órganos , Ovario/citología , Ovario/embriología , Ovario/metabolismo , Factor de Transcripción SOX9 , Células de Sertoli/metabolismo , Testículo/citología , Testículo/embriología , Testículo/metabolismo , Tubulina (Proteína)/metabolismo
7.
EMBO J ; 24(10): 1798-809, 2005 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-15889150

RESUMEN

During mammalian gonadal development, nuclear import/export of the transcription factor SOX9 is a critical step of the Sry-initiated testis-determining cascade. In this study, we identify a molecular mechanism contributing to the SOX9 nuclear translocation in NT2/D1 cells, which is mediated by the prostaglandin D2 (PGD2) signalling pathway via stimulation of its adenylcyclase-coupled DP1 receptor. We find that activation of cAMP-dependent protein kinase A (PKA) induces phosphorylation of SOX9 on its two S64 and S181 PKA sites, and its nuclear localization by enhancing SOX9 binding to the nucleocytoplasmic transport protein importin beta. Moreover, in embryonic gonads, we detect a male-specific prostaglandin D synthase expression and an active PGD2 signal at the time and place of SOX9 expression. We thus propose a new step in the sex-determining cascade where PGD2 acts as an autocrine factor inducing SOX9 nuclear translocation and subsequent Sertoli cell differentiation.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Prostaglandina D2/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Femenino , Masculino , Ratones , Técnicas de Cultivo de Órganos , Ovario/embriología , Ovario/enzimología , Ovario/metabolismo , Fosforilación , Transporte de Proteínas/fisiología , Factor de Transcripción SOX9 , Testículo/embriología , Testículo/enzimología , Testículo/metabolismo
8.
Cancer Res ; 65(6): 2193-8, 2005 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15781631

RESUMEN

The human carcinoembryonic antigen (CEA) is overexpressed in many types of human cancers and is commonly used as a clinical marker. In colon cancer, this overexpression protects cells against apoptosis and contributes to carcinogenesis. Therefore, CEA-expressing cells as well as CEA expression itself constitute potential therapeutic targets. In this report, we show that the transcription factor SOX9 down-regulates CEA gene expression and, as a probable consequence, induces apoptosis in the human colon carcinoma cell line HT29Cl.16E.


Asunto(s)
Antígeno Carcinoembrionario/genética , Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Proteínas del Grupo de Alta Movilidad/fisiología , Factores de Transcripción/fisiología , Apoptosis/fisiología , Antígeno Carcinoembrionario/biosíntesis , Diferenciación Celular/fisiología , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Regulación hacia Abajo/fisiología , Células HT29 , Proteínas del Grupo de Alta Movilidad/biosíntesis , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Regiones Promotoras Genéticas , Factor de Transcripción SOX9 , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
9.
EMBO J ; 23(16): 3336-45, 2004 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-15297880

RESUMEN

SRY, a Y chromosome-encoded DNA-binding protein, is required for testis organogenesis in mammals. Expression of the SRY gene in the genital ridge is followed by diverse early cell events leading to Sertoli cell determination/differentiation and subsequent sex cord formation. Little is known about SRY regulation and its mode of action during testis development, and direct gene targets for SRY are still lacking. In this study, we demonstrate that interaction of the human SRY with histone acetyltransferase p300 induces the acetylation of SRY both in vitro and in vivo at a single conserved lysine residue. We show that acetylation participates in the nuclear localisation of SRY by increasing SRY interaction with importin beta, while specific deacetylation by HDAC3 induces a cytoplasmic delocalisation of SRY. Finally, by analysing p300 and HDAC3 expression profiles during both human or mouse gonadal development, we suggest that acetylation and deacetylation of SRY may be important mechanisms for regulating SRY activity during mammalian sex determination.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Acetilación , Acetiltransferasas/metabolismo , Transporte Activo de Núcleo Celular , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular , ADN/metabolismo , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Gónadas/embriología , Gónadas/metabolismo , Histona Acetiltransferasas , Histona Desacetilasas/metabolismo , Humanos , Lisina/genética , Lisina/metabolismo , Masculino , Ratones , Proteínas Nucleares/genética , Unión Proteica , Proteína de la Región Y Determinante del Sexo , Factores de Transcripción/genética , Factores de Transcripción p300-CBP
10.
Development ; 131(15): 3795-804, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15240557

RESUMEN

Gastrointestinal (GI) development is highly conserved across vertebrates. Although several transcription factors and morphogenic proteins are involved in the molecular controls of GI development, the interplay between these factors is not fully understood. We report herein the expression pattern of Sox9 during GI development, and provide evidence that it functions, in part, to define the pyloric sphincter epithelium. SOX9 is expressed in the endoderm of the GI tract (with the exclusion of the gizzard) and its derivate organs, the lung and pancreas. Moreover, SOX9 is also expressed at the mesoderm of the pyloric sphincter, a structure that demarcates the gizzard from the duodenum. Using retroviral misexpression technique, we show that Sox9 expression in the pyloric sphincter is under the control of the BMP signaling pathway, known to play a key role in the development of this structure. By misexpressing SOX9 in the mesoderm of the gizzard, we show that SOX9 is able to transdifferentiate the adjacent gizzard epithelium into pyloric sphincter-like epithelium through the control of mesodermal-epithelial signals mediated in part by Gremlin (a modulator of the BMP pathway). Our results suggest that SOX9 is necessary and sufficient to specify the pyloric sphincter epithelial properties.


Asunto(s)
Epitelio/embriología , Regulación del Desarrollo de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/metabolismo , Mesodermo/fisiología , Músculo Liso/embriología , Transducción de Señal/fisiología , Estómago/embriología , Factores de Transcripción/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Embrión de Pollo , Epitelio/anatomía & histología , Epitelio/metabolismo , Mucosa Gástrica/metabolismo , Molleja de las Aves/citología , Molleja de las Aves/embriología , Molleja de las Aves/metabolismo , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mesodermo/citología , Morfogénesis , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Píloro/anatomía & histología , Píloro/embriología , Píloro/metabolismo , Factor de Transcripción SOX9 , Estómago/anatomía & histología , Factores de Transcripción/genética
11.
J Cell Biol ; 166(1): 37-47, 2004 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-15240568

RESUMEN

TCF and SOX proteins belong to the high mobility group box transcription factor family. Whereas TCFs, the transcriptional effectors of the Wnt pathway, have been widely implicated in the development, homeostasis and disease of the intestine epithelium, little is known about the function of the SOX proteins in this tissue. Here, we identified SOX9 in a SOX expression screening in the mouse fetal intestine. We report that the SOX9 protein is expressed in the intestinal epithelium in a pattern characteristic of Wnt targets. We provide in vitro and in vivo evidence that a bipartite beta-catenin/TCF4 transcription factor, the effector of the Wnt signaling pathway, is required for SOX9 expression in epithelial cells. Finally, in colon epithelium-derived cells, SOX9 transcriptionally represses the CDX2 and MUC2 genes, normally expressed in the mature villus cells of the intestinal epithelium, and may therefore contribute to the Wnt-dependent maintenance of a progenitor cell phenotype.


Asunto(s)
Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas de Homeodominio/metabolismo , Mucinas/metabolismo , Factores de Transcripción/metabolismo , Animales , Northern Blotting , Western Blotting , Factor de Transcripción CDX2 , Carcinoma/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Neoplasias del Colon/metabolismo , Proteínas del Citoesqueleto/metabolismo , ADN/metabolismo , Epitelio/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Ratones , Microscopía Fluorescente , Mucina 2 , Trasplante de Neoplasias , Fenotipo , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción SOX9 , Transducción de Señal , Células Madre/metabolismo , Transactivadores/metabolismo , Transcripción Genética , Transfección , beta Catenina
12.
Int J Dev Biol ; 47(6): 451-8, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-14584782

RESUMEN

We have performed a morphological, hormonal and molecular study of the development of the sex ducts in the mole Talpa occidentalis. Females develop bilateral ovotestes with a functional ovarian portion and disgenic testicular tissue. The Müllerian ducts develop normally in females and their regression is very fast in males, suggesting a powerful action of the anti-Müllerian hormone in the mole. RT-PCR demonstrated that the gene governing this hormone begins to be expressed in males coinciding with testis differentiation, and expression continues until shortly after birth. Immunohistochemical studies showed that expression occurs in the Sertoli cells of testes. No expression was detected in females. Wolffian duct development was normal in males and degenerate in prenatal females, but developmental recovery after birth gave rise to the formation of rudimentary epididymides. This event coincides in time with increasing serum testosterone levels and Leydig cell differentiation in the female gonad, thus suggesting that testosterone produced by the ovotestes is responsible for masculinisation of female moles. During postnatal development, serum testosterone concentrations decreased in males but increased in females, thus approaching the levels that adult males and females have during the non-breeding season.


Asunto(s)
Genitales Femeninos/anomalías , Glicoproteínas/metabolismo , Topos/embriología , Hormonas Testiculares/metabolismo , Testosterona/metabolismo , Animales , Hormona Antimülleriana , Secuencia de Bases , Bovinos , Femenino , Genitales Femeninos/embriología , Genitales Femeninos/metabolismo , Glicoproteínas/genética , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Conejos , Hormonas Testiculares/genética , Testosterona/sangre
13.
Mol Reprod Dev ; 66(3): 211-7, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14502599

RESUMEN

The Sox9 gene encodes a transcription factor that is critical for testis determination and chondrogenesis in vertebrates. Mutations in human SOX9 cause campomelic dysplasia, a dominant skeletal dysmorphology syndrome often associated with male to female sex reversal. Here we show that the Sox9a gene was duplicated during evolution of the rice field eel, Monopterus albus, a freshwater fish which undergoes natural sex reversal from female to male during its life, and has a haploid genome size (0.6-0.8 pg) that is among the smallest of the vertebrates. The duplicated copies of the gene (named Sox9al and Sox9a2) fit within the Sox9 clade of vertebrates, especially in the Sox9a subfamily, not in the Sox9b subfamily. They have similar structures as revealed by both genomic and cDNA analysis. Furthermore, both Sox9al and Sox9a2 are expressed in testis, ovary, and ovotestis; and specifically in the outer layer (mainly gonocytes) of gonadal epithelium with bipotential capacity to form testis or ovary, suggesting that they have similar roles in gonadal differentiation during sex reversal in this species. The closely related gene structure and expression patterns of the two sox9a genes in the rice field eel also suggest that they arose in recent gene duplication events during evolution of this fish lineage.


Asunto(s)
Anguilas/crecimiento & desarrollo , Gónadas/crecimiento & desarrollo , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Diferenciación Sexual/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , Anguilas/anatomía & histología , Anguilas/genética , Femenino , Duplicación de Gen , Gónadas/citología , Gónadas/metabolismo , Proteínas del Grupo de Alta Movilidad/clasificación , Humanos , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Familia de Multigenes , Filogenia , Estructura Terciaria de Proteína , Factor de Transcripción SOX9 , Alineación de Secuencia , Factores de Transcripción/clasificación
15.
Proc Natl Acad Sci U S A ; 99(17): 11199-204, 2002 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-12169669

RESUMEN

In mammals, male sex determination starts when the Y chromosome Sry gene is expressed within the undetermined male gonad. One of the earliest effect of Sry expression is to induce up-regulation of Sox9 gene expression in the developing gonad. SOX9, like SRY, contains a high mobility group domain and is sufficient to induce testis differentiation in transgenic XX mice. Before sexual differentiation, SOX9 protein is initially found in the cytoplasm of undifferentiated gonads from both sexes. At the time of testis differentiation and anti-Müllerian hormone expression, it becomes localized to the nuclear compartment in males whereas it is down-regulated in females. In this report, we used NIH 3T3 cells as a model to examine the regulation of SOX9 nucleo-cytoplasmic shuttling. SOX9-transfected cells expressed nuclear and cytoplasmic SOX9 whereas transfected cells treated with the nuclear export inhibitor leptomycin B, displayed an exclusive nuclear localization of SOX9. By using SOX9 deletion constructs in green fluorescent protein fusion proteins, we identified a functional nuclear export signal sequence between amino acids 134 and 147 of SOX9 high mobility group box. More strikingly, we show that inhibiting nuclear export with leptomycin B in mouse XX gonads cultured in vitro induced a sex reversal phenotype characterized by nuclear SOX9 and anti-Müllerian hormone expression. These results indicate that SOX9 nuclear export signal is essential for SOX9 sex-specific subcellular localization and could be part of a regulatory switch repressing (in females) or triggering (in males) male-specific sexual differentiation.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas del Grupo de Alta Movilidad/genética , Proteínas Nucleares , Diferenciación Sexual , Factores de Transcripción/genética , Células 3T3 , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Línea Celular , Chlorocebus aethiops , Citoplasma/metabolismo , Cartilla de ADN , Proteínas de Unión al ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes , Proteínas del Grupo de Alta Movilidad/química , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Técnicas de Cultivo de Órganos , Reacción en Cadena de la Polimerasa , Transporte de Proteínas , Factor de Transcripción SOX9 , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Proteína de la Región Y Determinante del Sexo , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Transfección , Cromosoma Y
16.
Nucleic Acids Res ; 30(14): 3245-52, 2002 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12136106

RESUMEN

SOX9 transcription factor is involved in chondrocyte differentiation and male sex determination. Heterozygous defects in the human SOX9 gene cause campomelic dysplasia. The mechanisms behind SOX9 function are not understood despite the description of different target genes. This study therefore sets out to identify SOX9-associated proteins to unravel how SOX9 interacts with the cellular transcription machinery. We report the ability of SOX9 to interact with TRAP230, a component of the thyroid hormone receptor-associated protein (TRAP) complex. Both in vitro and in vivo assays have confirmed that the detected interaction is specific and occurs endogenously in cells. Using co-transfection experiments, we have also shown that the TRAP230 interacting domain can act in a dominant-negative manner regarding SOX9 activity. Our results add SOX9 to the list of activators that communicate with the general transcription machinery through the TRAP complex and suggest a basis for the collaboration of SOX9 with different coactivators that could contact the same coactivator/integrator complex.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Unión Competitiva , Células COS , Proteínas Portadoras/química , Proteínas Portadoras/genética , Condrocitos/citología , Condrocitos/metabolismo , Expresión Génica , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Unión Proteica , Receptores de Hormona Tiroidea/metabolismo , Factor de Transcripción SOX9 , Saccharomyces cerevisiae/genética , Factores de Transcripción/genética , Activación Transcripcional/genética , Células Tumorales Cultivadas , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...