Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
BMC Genomics ; 17(1): 776, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27716130

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are short, non-coding RNAs that regulate gene expression mainly through translational repression of target mRNA molecules. More than 2700 human miRNAs have been identified and some are known to be associated with disease phenotypes and to display tissue-specific patterns of expression. METHODS: We used high-throughput small RNA sequencing to discover novel miRNAs in 93 human post-mortem prefrontal cortex samples from individuals with Huntington's disease (n = 28) or Parkinson's disease (n = 29) and controls without neurological impairment (n = 36). A custom miRNA identification analysis pipeline was built, which utilizes miRDeep* miRNA identification and result filtering based on false positive rate estimates. RESULTS: Ninety-nine novel miRNA candidates with a false positive rate of less than 5 % were identified. Thirty-four of the candidate miRNAs show sequence similarity with known mature miRNA sequences and may be novel members of known miRNA families, while the remaining 65 may constitute previously undiscovered families of miRNAs. Nineteen of the 99 candidate miRNAs were replicated using independent, publicly-available human brain RNA-sequencing samples, and seven were experimentally validated using qPCR. CONCLUSIONS: We have used small RNA sequencing to identify 99 putative novel miRNAs that are present in human brain samples.


Subject(s)
Brain/metabolism , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , MicroRNAs/genetics , Autopsy , Brain/pathology , Gene Expression Regulation , Humans , Huntington Disease/genetics , Parkinson Disease/genetics
2.
PLoS Genet ; 8(4): e1002569, 2012.
Article in English | MEDLINE | ID: mdl-22496664

ABSTRACT

Sex reversal can occur in XY humans with only a single functional WT1 or SF1 allele or a duplication of the chromosome region containing WNT4. In contrast, XY mice with only a single functional Wt1, Sf1, or Wnt4 allele, or mice that over-express Wnt4 from a transgene, reportedly are not sex-reversed. Because genetic background plays a critical role in testis differentiation, particularly in C57BL/6J (B6) mice, we tested the hypothesis that Wt1, Sf1, and Wnt4 are dosage sensitive in B6 XY mice. We found that reduced Wt1 or Sf1 dosage in B6 XY(B6) mice impaired testis differentiation, but no ovarian tissue developed. If, however, a Y(AKR) chromosome replaced the Y(B6) chromosome, these otherwise genetically identical B6 XY mice developed ovarian tissue. In contrast, reduced Wnt4 dosage increased the amount of testicular tissue present in Sf1+/- B6 XY(AKR), Wt1+/- B6 XY(AKR), B6 XY(POS), and B6 XY(AKR) fetuses. We propose that Wt1(B6) and Sf1(B6) are hypomorphic alleles of testis-determining pathway genes and that Wnt4(B6) is a hypermorphic allele of an ovary-determining pathway gene. The latter hypothesis is supported by the finding that expression of Wnt4 and four other genes in the ovary-determining pathway are elevated in normal B6 XX E12.5 ovaries. We propose that B6 mice are sensitive to XY sex reversal, at least in part, because they carry Wt1(B6) and/or Sf1(B6) alleles that compromise testis differentiation and a Wnt4(B6) allele that promotes ovary differentiation and thereby antagonizes testis differentiation. Addition of a "weak" Sry allele, such as the one on the Y(POS) chromosome, to the sensitized B6 background results in inappropriate development of ovarian tissue. We conclude that Wt1, Sf1, and Wnt4 are dosage-sensitive in mice, this dosage-sensitivity is genetic background-dependant, and the mouse strains described here are good models for the investigation of human dosage-sensitive XY sex reversal.


Subject(s)
Ovary/metabolism , Sex Determination Processes , Steroidogenic Factor 1/metabolism , Testis/metabolism , WT1 Proteins/metabolism , Wnt4 Protein/metabolism , Alleles , Animals , Female , Gene Dosage , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Ovary/growth & development , SOXB1 Transcription Factors/genetics , Steroidogenic Factor 1/genetics , Testis/growth & development , WT1 Proteins/genetics , Wnt4 Protein/genetics , X Chromosome/genetics , Y Chromosome/genetics
3.
Dev Dyn ; 238(11): 2877-90, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19842175

ABSTRACT

SMOC1 and SMOC2 are matricellular proteins thought to influence growth factor signaling, migration, proliferation, and angiogenesis. We examined the expression and regulation of Smoc1 and Smoc2 in fetal gonad/mesonephros complexes to discover possible roles for these genes in gonad and mesonephros development. Smoc1 was upregulated at approximately E10.75 in a center-to-poles wave in pre-Sertoli and pre-granulosa cells and its expression was greatly reduced in Wt1, Sf1, and Fog2 mutants. After E13.5, Smoc1 was downregulated in an anterior-to-posterior wave in granulosa cells but persisted in Sertoli cells, suggesting a sexually dimorphic requirement in supporting cell lineage differentiation. Smoc2 was expressed in Leydig cells, mesonephroi, and Wnt4 mutant ovaries, but not wildtype ovaries. Using organ culture, we determined that Smoc2 expression was dependent on Hedgehog signaling in testes, mesonephroi, and kidneys. Overall, these results demonstrate that SMOC1 and SMOC2 may mediate intercellular signaling and cell type-specific differentiation during gonad and reproductive tract development.


Subject(s)
Calcium-Binding Proteins/metabolism , Hedgehog Proteins/metabolism , Osteonectin/metabolism , Ovary/embryology , Testis/embryology , Animals , Cell Differentiation/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Down-Regulation/physiology , Female , Kidney/embryology , Kidney/metabolism , Leydig Cells/metabolism , Male , Mesonephros/cytology , Mesonephros/embryology , Mesonephros/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovary/metabolism , RNA Splicing Factors , Sertoli Cells/metabolism , Signal Transduction/physiology , Testis/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tretinoin/metabolism , Up-Regulation/genetics , Up-Regulation/physiology , WT1 Proteins/genetics , WT1 Proteins/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt4 Protein
4.
Dev Dyn ; 238(4): 812-25, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19301398

ABSTRACT

Mammalian gonad differentiation involves sexually dimorphic cell-fate decisions within the bipotential gonadal primordia. Testis differentiation is initiated by a center-to-poles wave of Sry expression that induces supporting cell precursors (SCPs) to become Sertoli rather than granulosa cells. The initiation of ovary differentiation is less well understood. We identified two novel SCP markers, 1700106J16Rik and Sprr2d, whose expression is ovary-biased during early gonad development, and altered in Wnt4, Sf1, Wt1, and Fog2 mutant gonads. In XX and XY gonads, both genes were up-regulated at approximately E11 in a center-to-poles wave, and then rapidly down-regulated in XY gonads in a center-to-poles wave, which is reminiscent of Sry expression in XY gonads. Our data suggest that 1700106J16Rik and Sprr2d may have important roles in early gonad development, and are consistent with the hypothesis that ovarian SCP differentiation occurs in a center-to-poles wave with similar timing to that of testicular SCP differentiation.


Subject(s)
Cornified Envelope Proline-Rich Proteins/metabolism , Gene Expression Regulation, Developmental , Ovary/embryology , Ovary/metabolism , Sex-Determining Region Y Protein/metabolism , Animals , Biomarkers/metabolism , Body Patterning , Cornified Envelope Proline-Rich Proteins/genetics , Down-Regulation , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Male , Mice , Sex-Determining Region Y Protein/genetics , Signal Transduction , Time Factors , Tretinoin/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt4 Protein
5.
Gene ; 433(1-2): 72-80, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19146932

ABSTRACT

SPARC-Related Modular Calcium Binding Protein-2 (Smoc-2) is a broadly-expressed matricellular protein which contributes to mitogenesis via activation of Integrin-Linked Kinase (ILK). Here we show that expression of Smoc2 is repressed in cultured cells following treatment with Aryl-hydrocarbon receptor (Ahr) ligands including the ubiquitous environmental pollutants Benzo[a]pyrene (B[a]P) and 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD). The Smoc2 promoter contains two consensus putative Ahr-binding sites and Smoc2 promoter-driven reporter genes are repressed in response to B[a]P in an Ahr-dependent manner in cultured cells. Using organ culture experiments we show that TCDD also represses Smoc2 mRNA expression in testes from Ahr(+/+) but not Ahr(-/-) mice. Therefore, exposure to Ahr ligands is likely to affect Smoc2 expression in vivo. Taken together our results indicate that Smoc2 is a novel transcriptional target of activated Ahr. Perturbation of Smoc2 expression may mediate the adverse developmental effects of environmental aryl-hydrocarbon exposure.


Subject(s)
Calcium-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , 3T3 Cells , Animals , Base Sequence , Benzo(a)pyrene/pharmacology , DNA Primers , Gene Expression Regulation, Developmental/drug effects , In Situ Hybridization , Mice , Promoter Regions, Genetic
6.
Cell Metab ; 8(4): 310-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18840361

ABSTRACT

Caveolae are specialized invaginations of the plasma membrane found in numerous cell types. They have been implicated as playing a role in a variety of physiological processes and are typically characterized by their association with the caveolin family of proteins. We show here by means of targeted gene disruption in mice that a distinct caveolae-associated protein, Cavin/PTRF, is an essential component of caveolae. Animals lacking Cavin have no morphologically detectable caveolae in any cell type examined and have markedly diminished protein expression of all three caveolin isoforms while retaining normal or above normal caveolin mRNA expression. Cavin-knockout mice are viable and of normal weight but have higher circulating triglyceride levels, significantly reduced adipose tissue mass, glucose intolerance, and hyperinsulinemia--characteristics that constitute a lipodystrophic phenotype. Our results underscore the multiorgan role of caveolae in metabolic regulation and the obligate presence of Cavin for caveolae formation.


Subject(s)
Caveolae/metabolism , Dyslipidemias/metabolism , Glucose Intolerance/metabolism , Membrane Proteins/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Body Composition , Caveolae/ultrastructure , Cell Membrane/metabolism , Female , Insulin/metabolism , Liver/cytology , Liver/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , RNA-Binding Proteins , Signal Transduction/physiology , Tissue Distribution
7.
Genesis ; 46(1): 8-18, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18196599

ABSTRACT

Heparan sulfate (HS) proteoglycans modulate the biological activity of a number of growth factors in development, homeostasis, and cancer. Specific modifications of HS chains by HS biosynthetic enzymes have been implicated in growth factor signaling in multiple aspects of organogenesis. Although the role of HS 6-O-sulfotransferases has been described in processes such as trachea formation in Drosophila and vasculogenesis in zebrafish, little is known about how HS 6-O-sulfotransferases (Hs6st1-3 in mice) influence mouse development. To address this issue, we generated a conditionally mutant Hs6st1 mouse line and then generated mice with systemic inactivation of Hs6st1. Hs6st1-null pups were viable and grossly normal at birth. The lack of obvious abnormalities in lung, liver, and kidney, which express high levels of Hs6st1 during development, suggests that at least during embryonic life, the loss of Hs6st1 function may be compensated for by mechanisms involving other HS modifying enzymes. During early adulthood, however, Hs6st1-null mice failed to thrive and exhibited growth retardation, body weight loss, enlargement of airspaces in the lung and, in some cases, lethality. Our results suggest a potentially critical role for HS 6-O sulfation by Hs6st1 in postnatal processes.


Subject(s)
Gene Expression Regulation, Developmental , Sulfotransferases/genetics , Sulfotransferases/physiology , Alleles , Animals , Crosses, Genetic , Female , Genetic Techniques , Genotype , Heparitin Sulfate/metabolism , Lung/pathology , Male , Mice , Mice, Mutant Strains , Mutation , Signal Transduction
8.
Proc Natl Acad Sci U S A ; 104(38): 14994-9, 2007 Sep 18.
Article in English | MEDLINE | ID: mdl-17848526

ABSTRACT

Previous reports suggested that humans and mice differ in their sensitivity to the genetic dosage of transcription factors that play a role in early testicular development. This difference implies that testis determination might be somewhat different in these two species. We report that the Fog2 and Gata4 transcription factors are haploinsufficient for testis determination in mice. Whether gonadal sex reversal occurs depends on genetic background (i.e., modifier genes). For example, C57BL/6J (B6) XY mice develop testes if they are heterozygous for a mutant Fog2 (Fog2-) or Gata4 (Gata4(ki)) allele. However, if the B6 Y chromosome (Y(B6)) is replaced by the AKR Y chromosome (Y(AKR)), B6 Fog2-/+ XY(AKR) mice develop ovaries, and B6 Gata4(ki)/+ XY(AKR) mice develop ovaries and ovotestes (gonads containing both ovarian and testicular tissue). Furthermore, DBA/2J (D2) Fog2-/+ XY(AKR) mice and (B6 x D2)F1 hybrid Gata4(ki)/+ XY(AKR) mice develop testes. Sry is expressed in the mutant XY gonads, indicating that the lack of Sry expression is not the cause of ovarian tissue development in B6 Fog2-/+ or Gata4(ki)/+ XY(AKR) mice. However, up-regulation of Sox9 expression, which is critical for normal testicular development, does not occur in mutant XY gonads that develop as ovaries. We conclude that under certain genetic conditions, Sox9 up-regulation depends on the proper dosage of Fog2 and Gata4. We propose that in humans the FOG2 and/or GATA4 genes might be haploinsufficient for normal testis determination and thus could be the cause of some previously unassigned cases of XY gonadal sex reversal.


Subject(s)
DNA-Binding Proteins/genetics , Fetal Development , GATA4 Transcription Factor/genetics , Gene Dosage , Testis/embryology , Transcription Factors/genetics , Animals , DNA-Binding Proteins/metabolism , Disorders of Sex Development , Female , GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Heterozygote , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , SOX9 Transcription Factor , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Testis/growth & development , Transcription Factors/metabolism
9.
Curr Biol ; 16(4): 415-20, 2006 Feb 21.
Article in English | MEDLINE | ID: mdl-16488877

ABSTRACT

The central dogma of mammalian brain sexual differentiation has contended that sex steroids of gonadal origin organize the neural circuits of the developing brain. Recent evidence has begun to challenge this idea and has suggested that, independent of the masculinizing effects of gonadal secretions, XY and XX brain cells have different patterns of gene expression that influence their differentiation and function. We have previously shown that specific differences in gene expression exist between male and female developing brains and that these differences precede the influences of gonadal hormones. Here we demonstrate that the Y chromosome-linked, male-determining gene Sry is specifically expressed in the substantia nigra of the adult male rodent in tyrosine hydroxylase-expressing neurons. Furthermore, using antisense oligodeoxynucleotides, we show that Sry downregulation in the substantia nigra causes a statistically significant decrease in tyrosine hydroxylase expression with no overall effect on neuronal numbers and that this decrease leads to motor deficits in male rats. Our studies suggest that Sry directly affects the biochemical properties of the dopaminergic neurons of the nigrostriatal system and the specific motor behaviors they control. These results demonstrate a direct male-specific effect on the brain by a gene encoded only in the male genome, without any mediation by gonadal hormones.


Subject(s)
Genes, sry/physiology , Sex Characteristics , Substantia Nigra/metabolism , Animals , Dopamine/metabolism , Down-Regulation , Female , Gene Expression , Male , Mice , Motor Activity , Neostriatum/metabolism , Neurons/metabolism , Rats , Tyrosine 3-Monooxygenase/metabolism
10.
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
11.
Development ; 132(13): 3045-54, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15944188

ABSTRACT

The nuclear receptor transcription factor Dax1 is hypothesized to play a role in testicular development, although the mechanism of its action is unknown. Here, we present evidence that Dax1 plays an early essential role in fetal testis development. We hypothesize that upregulation of Sox9 expression in precursor somatic cells, a process required for their differentiation as Sertoli cells, depends on the coordinated expression of Dax1, Sry and another gene, Tda1. Our conclusion and model are based on the following experimental findings: (1) presence of a mutant Dax1 allele (Dax1-) results in complete gonadal sex reversal in C57BL/6JEi (B6) XY mice, whereas testes develop in DBA/2J (D2) and (B6xD2)F1 XY mice; (2) B6-DAX1 sex reversal is inherited as a complex trait that includes the chromosome 4 gene Tda1; (3) B6 Dax1-/Y fetal gonads initiate development as ovaries, even though Sry expression is activated at the correct time and at appropriate levels; (4) upregulation of Sox9 does not occur in B6 Dax1-/Y fetal gonads in spite of apparently normal Sry expression; and (5) overexpression of Sry in B6 Dax1-/Y fetal gonads upregulates Sox9 and corrects testis development.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/genetics , High Mobility Group Proteins/metabolism , Sertoli Cells/metabolism , Sex Determination Processes , Transcription Factors/metabolism , Up-Regulation/physiology , Animals , DAX-1 Orphan Nuclear Receptor , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Female , Male , Mice , Mice, Congenic , Mice, Inbred AKR , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Ovary/embryology , Ovary/metabolism , SOX9 Transcription Factor , Sertoli Cells/cytology , Sex-Determining Region Y Protein , Testis/embryology , Testis/metabolism , Transcription Factors/genetics , Transcription Factors/physiology , X Chromosome , Y Chromosome
12.
Genetics ; 164(1): 277-88, 2003 May.
Article in English | MEDLINE | ID: mdl-12750339

ABSTRACT

Transfer of certain Mus domesticus-derived Y chromosomes (Sry(DOM) alleles, e.g., Sry(POS) and Sry(AKR)) onto the C57BL/6J (B6) mouse strain causes abnormal gonad development due to an aberrant interaction between the Sry(DOM) allele and the B6-derived autosomal (tda) genes. For example, B6 XY(POS) fetuses develop ovaries and ovotestes and B6 XY(AKR) fetuses have delayed testis cord development. To test whether abnormal testis development is caused by insufficient Sry(DOM) expression, two approaches were used. First, gonad development and relative Sry expression levels were examined in fetal gonads from two strains of B6 mice that contained a single M. domesticus-derived and a single M. musculus-derived Sry allele (B6-Y(POS,RIII) and B6-Y(AKR,RIII)). In both cases, presence of the M. musculus Sry(RIII) allele corrected abnormal testis development. On the B6 background, Sry(POS) was expressed at about half the level of Sry(RIII) whereas Sry(AKR) and Sry(RIII) were equally expressed. On an F(1) hybrid background, both Sry(POS) and Sry(RIII) expression increased, but Sry(POS) expression increased to a greater extent. Second, sexual development and Sry expression levels were determined in XX mice carrying a transgene expressing Sry(POS) controlled by POS-derived or MUS-derived regulatory regions. In both cases one B6 transgenic line was recovered in which XX transgenic mice developed only testicular tissue but cord development was delayed despite normal Sry transcriptional initiation and overexpression. For three transgenes where B6 XX transgenic mice developed as females, hermaphrodites, or males, the percentage of XX transgenic males increased on an F(1) background. For the one transgene examined, Sry expression increased on an F(1) background. These results support a model in which delayed testis development is caused by the presence of particular DOM SRY protein isoforms and this, combined with insufficient Sry expression, causes sex reversal. These results also indicate that at least one tda gene regulates Sry expression, possibly by directly binding to Sry regulatory regions.


Subject(s)
DNA-Binding Proteins/genetics , Nuclear Proteins , Sex Determination Processes , Testis/metabolism , Transcription Factors , Animals , DNA-Binding Proteins/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Sex-Determining Region Y Protein , X Chromosome , Y Chromosome
13.
Development ; 129(19): 4627-34, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12223418

ABSTRACT

In mammals, Sry expression in the bipotential, undifferentiated gonad directs the support cell precursors to differentiate as Sertoli cells, thus initiating the testis differentiation pathway. In the absence of Sry, or if Sry is expressed at insufficient levels, the support cell precursors differentiate as granulosa cells, thus initiating the ovarian pathway. The molecular mechanisms upstream and downstream of Sry are not well understood. We demonstrate that the transcription factor GATA4 and its co-factor FOG2 are required for gonadal differentiation. Mouse fetuses homozygous for a null allele of Fog2 or homozygous for a targeted mutation in Gata4 (Gata4(ki)) that abrogates the interaction of GATA4 with FOG co-factors exhibit abnormalities in gonadogenesis. We found that Sry transcript levels were significantly reduced in XY Fog2(-/-) gonads at E11.5, which is the time when Sry expression normally reaches its peak. In addition, three genes crucial for normal Sertoli cell function (Sox9, Mis and Dhh) and three Leydig cell steroid biosynthetic enzymes (p450scc, 3betaHSD and p450c17) were not expressed in XY Fog2(-/-) and Gata(ki/ki) gonads, whereas Wnt4, a gene required for normal ovarian development, was expressed ectopically. By contrast, Wt1 and Sf1, which are expressed prior to Sry and necessary for gonad development in both sexes, were expressed normally in both types of mutant XY gonads. These results indicate that GATA4 and FOG2 and their physical interaction are required for normal gonadal development.


Subject(s)
DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Nuclear Proteins , Ovary/embryology , Sex Determination Processes , Testis/embryology , Transcription Factors/metabolism , Zinc Fingers , Animals , Biomarkers , Cell Differentiation , Cholesterol Side-Chain Cleavage Enzyme/genetics , DNA-Binding Proteins/genetics , Female , Fushi Tarazu Transcription Factors , GATA4 Transcription Factor , Gonads , High Mobility Group Proteins/genetics , Homeodomain Proteins , Male , Mice , Mice, Knockout , Mice, Transgenic , Multienzyme Complexes/genetics , Ovary/abnormalities , Progesterone Reductase/genetics , Receptors, Cytoplasmic and Nuclear , SOX9 Transcription Factor , Sertoli Cells/cytology , Sex-Determining Region Y Protein , Steroid 17-alpha-Hydroxylase/genetics , Steroid Isomerases/genetics , Steroidogenic Factor 1 , Testis/abnormalities , Transcription Factors/genetics , WT1 Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...