Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Bacteriol ; 198(3): 427-35, 2016 02 01.
Article in English | MEDLINE | ID: mdl-26553849

ABSTRACT

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.


Subject(s)
Brucella suis/metabolism , High-Throughput Nucleotide Sequencing/methods , Host Factor 1 Protein/metabolism , RNA, Bacterial/metabolism , Transcriptome , Animals , Brucella suis/genetics , Cell Line , Gene Expression Regulation, Bacterial/physiology , Genome-Wide Association Study , Host Factor 1 Protein/genetics , Mice , Molecular Chaperones , RNA, Bacterial/genetics , RNA, Untranslated
2.
Proc Natl Acad Sci U S A ; 104(41): 16335-40, 2007 Oct 09.
Article in English | MEDLINE | ID: mdl-17913892

ABSTRACT

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.


Subject(s)
Anorexia Nervosa/metabolism , Nerve Tissue Proteins/metabolism , Nucleus Accumbens/metabolism , Receptors, Serotonin, 5-HT4/metabolism , Animals , Anorexia Nervosa/etiology , Anorexia Nervosa/genetics , Base Sequence , Eating , Male , Mice , Mice, Knockout , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nucleus Accumbens/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Receptors, Serotonin, 5-HT4/deficiency , Receptors, Serotonin, 5-HT4/genetics , Signal Transduction
3.
Cancer Lett ; 255(2): 182-93, 2007 Oct 08.
Article in English | MEDLINE | ID: mdl-17532558

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma/metabolism , High Mobility Group Proteins/metabolism , Intramolecular Oxidoreductases/metabolism , Ovarian Neoplasms/metabolism , Transcription Factors/metabolism , Apoptosis , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , High Mobility Group Proteins/analysis , High Mobility Group Proteins/genetics , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/pharmacology , Lipocalins , Male , Ovarian Neoplasms/pathology , RNA, Small Interfering/pharmacology , Receptors, Prostaglandin/agonists , SOX9 Transcription Factor , Signal Transduction , Transcription Factors/analysis , Transcription Factors/genetics , Up-Regulation
4.
Gene Expr Patterns ; 6(7): 695-702, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16488195

ABSTRACT

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.


Subject(s)
High Mobility Group Proteins/analysis , High Mobility Group Proteins/genetics , Ovarian Follicle/chemistry , Ovarian Follicle/physiology , Ovary/chemistry , Transcription Factors/analysis , Transcription Factors/genetics , Animals , Female , Gene Expression Regulation , High Mobility Group Proteins/immunology , Mice , Ovary/physiology , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor , Theca Cells/chemistry , Transcription Factors/immunology
5.
J Biol Chem ; 280(46): 38625-30, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16166090

ABSTRACT

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.


Subject(s)
Cytoskeletal Proteins/physiology , Nerve Tissue Proteins/physiology , Sex-Determining Region Y Protein/metabolism , Amino Acid Motifs , Animals , Cell Differentiation , Cell Line , Conserved Sequence , Cytoskeletal Proteins/metabolism , Female , Glutathione Transferase/metabolism , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , NIH 3T3 Cells , Nerve Tissue Proteins/metabolism , Phosphoproteins , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , RNA-Binding Proteins , Recombinant Proteins/chemistry , Sertoli Cells/cytology , Sodium-Hydrogen Exchangers , Species Specificity , Subcellular Fractions , Testis/metabolism , Transfection
6.
Exp Cell Res ; 309(2): 468-75, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16087173

ABSTRACT

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.


Subject(s)
High Mobility Group Proteins/metabolism , Microtubules/metabolism , Sex Determination Processes , Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Cell Line, Tumor , Female , Humans , Male , Mice , NIH 3T3 Cells , Organ Culture Techniques , Ovary/cytology , Ovary/embryology , Ovary/metabolism , SOX9 Transcription Factor , Sertoli Cells/metabolism , Testis/cytology , Testis/embryology , Testis/metabolism , Tubulin/metabolism
7.
EMBO J ; 24(10): 1798-809, 2005 May 18.
Article in English | MEDLINE | ID: mdl-15889150

ABSTRACT

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.


Subject(s)
Cell Nucleus/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , High Mobility Group Proteins/metabolism , Prostaglandin D2/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Female , Male , Mice , Organ Culture Techniques , Ovary/embryology , Ovary/enzymology , Ovary/metabolism , Phosphorylation , Protein Transport/physiology , SOX9 Transcription Factor , Testis/embryology , Testis/enzymology , Testis/metabolism
8.
Cancer Res ; 65(6): 2193-8, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15781631

ABSTRACT

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.


Subject(s)
Carcinoembryonic Antigen/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic/physiology , High Mobility Group Proteins/physiology , Transcription Factors/physiology , Apoptosis/physiology , Carcinoembryonic Antigen/biosynthesis , Cell Differentiation/physiology , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Down-Regulation/physiology , HT29 Cells , High Mobility Group Proteins/biosynthesis , High Mobility Group Proteins/genetics , Humans , Promoter Regions, Genetic , SOX9 Transcription Factor , Transcription Factors/biosynthesis , Transcription Factors/genetics
9.
EMBO J ; 23(16): 3336-45, 2004 Aug 18.
Article in English | MEDLINE | ID: mdl-15297880

ABSTRACT

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.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Acetylation , Acetyltransferases/metabolism , Active Transport, Cell Nucleus , Animals , Cell Cycle Proteins/metabolism , Cell Line , DNA/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation , Gonads/embryology , Gonads/metabolism , Histone Acetyltransferases , Histone Deacetylases/metabolism , Humans , Lysine/genetics , Lysine/metabolism , Male , Mice , Nuclear Proteins/genetics , Protein Binding , Sex-Determining Region Y Protein , Transcription Factors/genetics , p300-CBP Transcription Factors
10.
Development ; 131(15): 3795-804, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15240557

ABSTRACT

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.


Subject(s)
Epithelium/embryology , Gene Expression Regulation, Developmental , High Mobility Group Proteins/metabolism , Mesoderm/physiology , Muscle, Smooth/embryology , Signal Transduction/physiology , Stomach/embryology , Transcription Factors/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Chick Embryo , Epithelium/anatomy & histology , Epithelium/metabolism , Gastric Mucosa/metabolism , Gizzard, Avian/cytology , Gizzard, Avian/embryology , Gizzard, Avian/metabolism , High Mobility Group Proteins/genetics , Humans , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/metabolism , Mesoderm/cytology , Morphogenesis , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Pylorus/anatomy & histology , Pylorus/embryology , Pylorus/metabolism , SOX9 Transcription Factor , Stomach/anatomy & histology , Transcription Factors/genetics
11.
J Cell Biol ; 166(1): 37-47, 2004 Jul 05.
Article in English | MEDLINE | ID: mdl-15240568

ABSTRACT

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.


Subject(s)
High Mobility Group Proteins/metabolism , Homeodomain Proteins/metabolism , Mucins/metabolism , Transcription Factors/metabolism , Animals , Blotting, Northern , Blotting, Western , CDX2 Transcription Factor , Carcinoma/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Nucleus/metabolism , Colonic Neoplasms/metabolism , Cytoskeletal Proteins/metabolism , DNA/metabolism , Epithelium/metabolism , Gene Expression Regulation, Neoplastic , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Intestinal Mucosa/metabolism , Mice , Microscopy, Fluorescence , Mucin-2 , Neoplasm Transplantation , Phenotype , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor , Signal Transduction , Stem Cells/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Transfection , beta Catenin
12.
Int J Dev Biol ; 47(6): 451-8, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14584782

ABSTRACT

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.


Subject(s)
Genitalia, Female/abnormalities , Glycoproteins/metabolism , Moles/embryology , Testicular Hormones/metabolism , Testosterone/metabolism , Animals , Anti-Mullerian Hormone , Base Sequence , Cattle , Female , Genitalia, Female/embryology , Genitalia, Female/metabolism , Glycoproteins/genetics , Humans , Male , Mice , Molecular Sequence Data , Rabbits , Testicular Hormones/genetics , Testosterone/blood
13.
Mol Reprod Dev ; 66(3): 211-7, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14502599

ABSTRACT

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.


Subject(s)
Eels/growth & development , Gonads/growth & development , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sex Differentiation/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Eels/anatomy & histology , Eels/genetics , Female , Gene Duplication , Gonads/cytology , Gonads/metabolism , High Mobility Group Proteins/classification , Humans , Male , Models, Molecular , Molecular Sequence Data , Multigene Family , Phylogeny , Protein Structure, Tertiary , SOX9 Transcription Factor , Sequence Alignment , Transcription Factors/classification
15.
Proc Natl Acad Sci U S A ; 99(17): 11199-204, 2002 Aug 20.
Article in English | MEDLINE | ID: mdl-12169669

ABSTRACT

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.


Subject(s)
Cell Nucleus/metabolism , High Mobility Group Proteins/genetics , Nuclear Proteins , Sex Differentiation , Transcription Factors/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cell Line , Chlorocebus aethiops , Cytoplasm/metabolism , DNA Primers , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Green Fluorescent Proteins , High Mobility Group Proteins/chemistry , High Mobility Group Proteins/metabolism , Luminescent Proteins/genetics , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Organ Culture Techniques , Polymerase Chain Reaction , Protein Transport , SOX9 Transcription Factor , Sequence Alignment , Sequence Homology, Amino Acid , Sex-Determining Region Y Protein , Transcription Factors/chemistry , Transcription Factors/metabolism , Transfection , Y Chromosome
16.
Nucleic Acids Res ; 30(14): 3245-52, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12136106

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , High Mobility Group Proteins/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Binding, Competitive , COS Cells , Carrier Proteins/chemistry , Carrier Proteins/genetics , Chondrocytes/cytology , Chondrocytes/metabolism , Gene Expression , High Mobility Group Proteins/genetics , Humans , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Receptors, Thyroid Hormone/metabolism , SOX9 Transcription Factor , Saccharomyces cerevisiae/genetics , Transcription Factors/genetics , Transcriptional Activation/genetics , Tumor Cells, Cultured , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...