Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
PLoS Genet ; 8(4): e1002569, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22496664

RESUMO

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.


Assuntos
Ovário/metabolismo , Processos de Determinação Sexual , Fator Esteroidogênico 1/metabolismo , Testículo/metabolismo , Proteínas WT1/metabolismo , Proteína Wnt4/metabolismo , Alelos , Animais , Feminino , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Ovário/crescimento & desenvolvimento , Fatores de Transcrição SOXB1/genética , Fator Esteroidogênico 1/genética , Testículo/crescimento & desenvolvimento , Proteínas WT1/genética , Proteína Wnt4/genética , Cromossomo X/genética , Cromossomo Y/genética
2.
Nat Genet ; 33(2): 145-53, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12548288

RESUMO

Hermansky-Pudlak syndrome (HPS) is a genetically heterogeneous disease involving abnormalities of melanosomes, platelet dense granules and lysosomes. Here we have used positional candidate and transgenic rescue approaches to identify the genes mutated in ruby-eye 2 and ruby-eye mice (ru2 and ru, respectively), two 'mimic' mouse models of HPS. We also show that these genes are orthologs of the genes mutated in individuals with HPS types 5 and 6, respectively, and that their protein products directly interact. Both genes are previously unknown and are found only in higher eukaryotes, and together represent a new class of genes that have evolved in higher organisms to govern the synthesis of highly specialized lysosome-related organelles.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular , Proteínas de Drosophila , Síndrome de Hermanski-Pudlak/genética , Proteínas de Insetos/genética , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras , Mutação/genética , Proteínas/genética , Complexo 3 de Proteínas Adaptadoras , Subunidades beta do Complexo de Proteínas Adaptadoras , Adulto , Sequência de Aminoácidos , Animais , Células COS , Pré-Escolar , Chlorocebus aethiops , Cromossomos Artificiais Bacterianos/genética , Cromossomos Artificiais de Bacteriófago P1/genética , Modelos Animais de Doenças , Feminino , Síndrome de Hermanski-Pudlak/metabolismo , Síndrome de Hermanski-Pudlak/patologia , Humanos , Masculino , Melanossomas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Dados de Sequência Molecular , Oligopeptídeos , Peptídeos/imunologia , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , Proteínas Proto-Oncogênicas c-myc/imunologia , Saccharomyces cerevisiae/metabolismo , Homologia de Sequência de Aminoácidos , Transfecção , Técnicas do Sistema de Duplo-Híbrido
3.
J Mol Med (Berl) ; 101(9): 1141-1151, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37584738

RESUMO

Autosomal-recessive polycystic kidney disease (ARPKD; MIM #263200) is a severe, hereditary, hepato-renal fibrocystic disorder that causes early childhood morbidity and mortality. Mutations in the polycystic kidney and hepatic disease 1 (PKHD1) gene, which encodes the protein fibrocystin/polyductin complex (FPC), cause all typical forms of ARPKD. Several mouse lines carrying diverse, genetically engineered disruptions in the orthologous Pkhd1 gene have been generated, but none expresses the classic ARPKD renal phenotype. In the current study, we characterized a spontaneous mouse Pkhd1 mutation that is transmitted as a recessive trait and causes cysticliver (cyli), similar to the hepato-biliary disease in ARPKD, but which is exacerbated by age, sex, and parity. We mapped the mutation to Chromosome 1 and determined that an insertion/deletion mutation causes a frameshift within Pkhd1 exon 48, which is predicted to result in a premature termination codon (UGA). Pkhd1cyli/cyli (cyli) mice exhibit a severe liver pathology but lack renal disease. Further analysis revealed that several alternatively spliced Pkhd1 mRNA, all containing exon 48, were expressed in cyli kidneys, but in lower abundance than in wild-type kidneys, suggesting that these transcripts escaped from nonsense-mediated decay (NMD). We identified an AAAAAT motif in exon 48 upstream of the cyli mutation which could enable ribosomal frameshifting, thus potentially allowing production of sufficient amounts of FPC for renoprotection. This mechanism, expressed in a species-specific fashion, may help explain the disparities in the renal phenotype observed between Pkhd1 mutant mice and patients with PKHD1-related disease. KEY MESSAGES: The Pkhd1cyli/cyli mouse expresses cystic liver disease, but no kidney phenotype. Pkhd1 mRNA expression is decreased in cyli liver and kidneys compared to wild-type. Ribosomal frameshifting may be responsible for Pkhd1 mRNA escape from NMD. Pkhd1 mRNA escape from NMD could contribute to the absent kidney phenotype.


Assuntos
Hepatopatias , Rim Policístico Autossômico Recessivo , Pré-Escolar , Camundongos , Humanos , Animais , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/patologia , Rim/metabolismo , Mutação , Fatores de Transcrição/genética , RNA Mensageiro/genética , Receptores de Superfície Celular/genética
4.
Biol Reprod ; 82(2): 380-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19864314

RESUMO

Mammalian gonadal sex-determining (GSD) genes are expressed in a unique population of somatic cells that differentiate into granulosa cells in XX gonads or Sertoli cells in XY gonads. The ability to efficiently isolate these somatic support cells (SSCs) during the earliest stages of gonad development would facilitate identifying 1) new candidate GSD genes that may be involved in cases of unexplained abnormal gonad development and 2) genes involved in the earliest stages of granulosa and Sertoli cell differentiation. We report the development of a unique mouse carrying two transgenes that allow XX and XY mice to be distinguished as early as Embryonic Day 11.5 (E11.5) and allow SSCs to be isolated from undifferentiated (E11.5) and early differentiated (E12.5) fetal gonads. The Mouse Genome 430v2.0 GeneChip (Affymetrix) was used to identify transcripts exhibiting a sexual dimorphic expression pattern in XX and XY isolated SSCs. The analysis revealed previously unidentified sexually dimorphic transcripts, including low-level expressed genes such as Sry, a gene not identified in other microarray studies. Multigene real-time PCR analysis of 57 genes verified that 53 were expressed in fetal gonads in a sexually dimorphic pattern, and whole-mount in situ hybridization analysis verified 4930563E18Rik, Pld1, and Sprr2d are expressed in XX gonads, and Fbln2, Ppargc1a, and Scrn1 are expressed in XY gonads. Taken together, the data provide a comprehensive resource for the spatial-temporal expression pattern of genes that are part of the genetic network underlying the early stages of mammalian fetal gonadal development, including the development of granulosa and Sertoli cells.


Assuntos
Diferenciação Celular/genética , Perfilação da Expressão Gênica/veterinária , Gônadas/embriologia , Células da Granulosa/citologia , Células de Sertoli/citologia , Processos de Determinação Sexual , Animais , Feminino , Genes sry/genética , Proteínas de Fluorescência Verde/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Ovário/embriologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Caracteres Sexuais , Testículo/embriologia
5.
Genetics ; 179(1): 419-27, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18458101

RESUMO

C57BL/6J (B6) mice containing the Mus domesticus poschiavinus Y chromosome, YPOS, develop ovarian tissue, whereas testicular tissue develops in DBA/2J or 129S1/SvImJ (129) mice containing the YPOS chromosome. To identify genes involved in sex determination, we used a congenic strain approach to determine which chromosomal regions from 129Sl/SvImJ provide protection against sex reversal in XYPOS mice of the C57BL/6J.129-YPOS strain. Genome scans using microsatellite and SNP markers identified a chromosome 11 region of 129 origin in C57BL/6J.129-YPOS mice. To determine if this region influenced testis development in XYPOS mice, two strains of C57BL/6J-YPOS mice were produced and used in genetic experiments. XYPOS adults homozygous for the 129 region had a lower incidence of sex reversal than XYPOS adults homozygous for the B6 region. In addition, many homozygous 129 XYPOS fetuses developed normal-appearing testes, an occurrence never observed in XYPOS mice of the C57BL/6J-YPOS strain. Finally, the amount of testicular tissue observed in ovotestes of heterozygous 129/B6 XYPOS fetuses was greater than the amount observed in ovotestes of homozygous B6 XYPOS fetuses. We conclude that a chromosome 11 locus derived from 129Sl/SvImJ essentially protects against sex reversal in XYPOS mice. A number of genes located in this chromosome 11 region are discussed as potential candidates.


Assuntos
Cromossomos de Mamíferos/genética , Transtornos do Desenvolvimento Sexual , Processos de Determinação Sexual , Testículo/metabolismo , Cromossomo Y/genética , Animais , Biologia Computacional , Primers do DNA/genética , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Repetições de Microssatélites/genética , Polimorfismo de Nucleotídeo Único , Testículo/crescimento & desenvolvimento
6.
Gene Expr Patterns ; 7(1-2): 113-23, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16839824

RESUMO

Gonadal sex determining (GSD) genes that initiate fetal ovarian and testicular development and differentiation are expressed in the cells of the urogenital ridge that differentiate as somatic support cells (SSCs), i.e., granulosa cells of the ovary and Sertoli cells of the testis. To identify potential new mammalian GSD genes, we used the Mouse Genome 430v2.0 GeneChip to analyze gene expression differences between XX and XY SSCs cells isolated from the gonads of embryonic day (E) 13 C57BL/6J fetuses carrying an EGFP reporter transgene expressed specifically in SSCs. In addition, genome wide expression differences between XX and XY E13 whole gonads were examined. Analysis revealed that XX and XY E13 SSCs differentially express 647 transcripts (False Discovery Rate cutoff 1%), including transcripts not previously reported to exhibit a sexually dimorphic expression pattern in this unique cell population. Enrichment for genes controlling cell proliferation was noted in XY SSCs, whereas enrichment for genes controlling cell morphology and metabolic status was identified in XX SSCs. Among the newly identified differentially expressed transcripts are potential GSD genes involved in unexplained human sex reversal cases.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Animais , Diferenciação Celular , Transtornos do Desenvolvimento Sexual , Feminino , Perfilação da Expressão Gênica , Disgenesia Gonadal/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Ovário/citologia , Ovário/embriologia , Ovário/metabolismo , Gravidez , Caracteres Sexuais , Processos de Determinação Sexual , Testículo/citologia , Testículo/embriologia , Testículo/metabolismo
7.
Cancer Res ; 63(23): 8197-202, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14678975

RESUMO

SWR/Bm (SWR) female mice spontaneously develop early-onset ovarian granulosa cell (GC) tumors that can progress to metastatic carcinoma and thus provide a model system for human, juvenile-type GC tumors. In SWR mice, GC tumor susceptibility is an inherited, polygenic trait that appears at a low frequency. A dramatic increase in tumor frequency occurs when the autosomal SWR genetic complement is combined with the X-linked Gct4 allele of the mouse strain SJL/Bm (SJL). The modifier effect of the SJL Gct4 allele (Gct4(J)) also shows a strong parent-of-origin effect, occurring only when the Gct4(J) allele is paternally inherited. To genetically localize Gct4, we generated seven congenic mouse strains (SWR.SJL-X1 through -X7) that contained a defined segment of the SJL X chromosome (Chr) on the SWR autosomal strain background and mapped Gct4 to a 3 cM region. To better define the location of Gct4, we created an additional congenic strain (SWR.CAST-X) that contains most of the genetically polymorphic Chr X from the strain CAST/Ei. From crosses of the SWR.CAST-X and SWR.SJL-X congenic strains, we derived males carrying unique combinations of SJL-X and CAST-X segments. Progeny testing subsequently revealed a second SJL-derived, GC tumor frequency modifier gene, Gct6, located 6.5 cM distal to Gct4 on Chr X. In summary, we have mapped two modifier genes on the mouse Chr X that cause high-frequency, juvenile-type GC tumor development in female mice. The identity of these genes will provide a solid foundation for determination of tumor susceptibility genes in human cases of juvenile-type GC tumors.


Assuntos
Doenças Genéticas Ligadas ao Cromossomo X/genética , Tumor de Células da Granulosa/genética , Oncogenes/genética , Neoplasias Ovarianas/genética , Cromossomo X/genética , Animais , Mapeamento Cromossômico/métodos , Feminino , Predisposição Genética para Doença , Endogamia , Masculino , Camundongos , Recombinação Genética
8.
Genetics ; 164(1): 277-88, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12750339

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas Nucleares , Processos de Determinação Sexual , Testículo/metabolismo , Fatores de Transcrição , Animais , Proteínas de Ligação a DNA/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Proteína da Região Y Determinante do Sexo , Cromossomo X , Cromossomo Y
9.
Gene Expr Patterns ; 5(1): 141-9, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15533830

RESUMO

New techniques are being applied to identify all the genes involved in mammalian gonad development and differentiation. As this list of genes increases, understanding the potential interactions between these genes will become increasingly difficult. We used a real time reverse transcription PCR (real time RTPCR) protocol to examine and compare the relative expression levels of 55 genes in individual mouse fetal gonads. Real time PCR analysis demonstrated that except for Sry, no differences in relative gene expression were detectable between XX and XY gonad/mesonephroi complexes at embryonic day (E)11.5. Following Sry peak expression at E11.5, a number of genes were expressed at significantly higher relative levels in E12-14 XY than XX gonads. Of six genes expressed at higher levels in E12.5-14 XX than XY gonads, three, Bmp2, Emx2, and Fgfr2, had not been reported previously. Our results caution that differential localization patterns observed with whole mount in situ hybridization techniques may not accurately reflect changes in transcript levels. We conclude that real time PCR is an efficient and powerful tool for studying multiple gene expression patterns during gonad development and differentiation, and can provide insight into gene interactions.


Assuntos
Expressão Gênica/fisiologia , Ovário/embriologia , Testículo/embriologia , Animais , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Ovário/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/metabolismo , Cromossomo X , Cromossomo Y
10.
Mech Dev ; 126(5-6): 324-36, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19269320

RESUMO

Ovotestis development in B6-XY(POS) mice provides a rare opportunity to study the interaction of the testis- and ovary-determining pathways in the same tissue. We studied expression of several markers of mouse fetal testis (SRY, SOX9) or ovary (FOXL2, Rspo1) development in B6-XY(POS) ovotestes by immunofluorescence, using normal testes and ovaries as controls. In ovotestes, SOX9 was expressed only in the central region where SRY is expressed earliest, resulting in testis cord formation. Surprisingly, FOXL2-expressing cells also were found in this region, but individual cells expressed either FOXL2 or SOX9, not both. At the poles, even though SOX9 was not up-regulated, SRY expression was down-regulated normally as in XY testes, and FOXL2 was expressed from an early stage, demonstrating ovarian differentiation in these areas. Our data (1) show that SRY must act within a specific developmental window to activate Sox9; (2) challenge the established view that SOX9 is responsible for down-regulating Sry expression; (3) disprove the concept that testicular and ovarian cells occupy discrete domains in ovotestes; and (4) suggest that FOXL2 is actively suppressed in Sertoli cell precursors by the action of SOX9. Together these findings provide important new insights into the molecular regulation of testis and ovary development.


Assuntos
Ovário/embriologia , Processos de Determinação Sexual , Testículo/embriologia , Animais , Biomarcadores/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/genética , Feminino , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Ovário/citologia , Ovário/metabolismo , Fatores de Processamento de RNA , Fatores de Transcrição SOX9/metabolismo , Proteína da Região Y Determinante do Sexo/genética , Proteína da Região Y Determinante do Sexo/metabolismo , Testículo/citologia , Testículo/metabolismo , Fatores de Tempo , Fatores de Transcrição/metabolismo
11.
Chromosome Res ; 16(4): 623-36, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18483871

RESUMO

In both humans and mice there are numerous reports of Y chromosome abnormalities that interfere with sex determination. Recent studies in the mouse of one such mutation have identified Y chromosome nondisjunction during preimplantation development as the cause of abnormal testis determination that results in a high frequency of true hermaphroditism. We report here that the mouse Y chromosome from the A/HeJ inbred strain induces similar aberrations in sex determination. Our analyses provide evidence, however, that the mechanism underlying these aberrations is not Y chromosome nondisjunction. On the basis of our findings, we postulate that a mutation at or near the centromere affects both the segregation and sex-determining properties of the A/HeJ Y chromosome. This Y chromosome adds to the growing list of Y chromosome aberrations in humans and mice. In both species, the centromere of the Y is structurally and morphologically distinct from the centromeres of all other chromosomes. We conclude that these centromeric features make the human and mouse Y chromosomes extremely sensitive to minor structural alterations, and that our studies provide yet another example of a good Y chromosome gone 'bad.'


Assuntos
Cromossomos de Mamíferos/genética , Camundongos/genética , Cromossomo Y/genética , Animais , Southern Blotting , Centrômero/metabolismo , Segregação de Cromossomos , Transtornos do Desenvolvimento Sexual , Feminino , Feto/metabolismo , Fibroblastos/citologia , Genoma/genética , Fígado/citologia , Masculino , Meiose , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mitose , Não Disjunção Genética , Ploidias , Diferenciação Sexual , Testículo/citologia
12.
Proc Natl Acad Sci U S A ; 104(38): 14994-9, 2007 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-17848526

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/genética , Desenvolvimento Fetal , Fator de Transcrição GATA4/genética , Dosagem de Genes , Testículo/embriologia , Fatores de Transcrição/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Transtornos do Desenvolvimento Sexual , Feminino , Fator de Transcrição GATA4/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Heterozigoto , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Fatores de Transcrição SOX9 , Proteína da Região Y Determinante do Sexo/genética , Proteína da Região Y Determinante do Sexo/metabolismo , Testículo/crescimento & desenvolvimento , Fatores de Transcrição/metabolismo
13.
Development ; 132(13): 3045-54, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15944188

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Células de Sertoli/metabolismo , Processos de Determinação Sexual , Fatores de Transcrição/metabolismo , Regulação para Cima/fisiologia , Animais , Receptor Nuclear Órfão DAX-1 , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Feminino , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiologia , Ovário/embriologia , Ovário/metabolismo , Fatores de Transcrição SOX9 , Células de Sertoli/citologia , Proteína da Região Y Determinante do Sexo , Testículo/embriologia , Testículo/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Cromossomo X , Cromossomo Y
14.
Evolution ; 46(4): 1146-1163, 1992 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28564395

RESUMO

We compared the patterns of movement of sex chromosomal and autosomal loci along a 160 km transect across a zone of hybridization between M. domesticus and M. musculus in southern Germany and western Austria using seven genetic markers. These included one Y-specific DNA sequence (YB10), two X-specific loci (DXWas68 and DXWas31), and four autosomal isozyme loci (Es-10, Es-1, Mpi-1, and Np-1). Random effects logistic regression analysis enabled us to examine the relationship between M. domesticus allele frequency and geographic distance from the western edge of the hybrid zone and allowed statistical evaluation of differences in cline midpoint and width among loci. More limited movement was observed for all three sex chromosomal markers across the zone compared with three of the four autosomal markers. If differential movement reflects fitness differences of specific alleles (or alleles at closely linked loci) on a hybrid background, then alleles that move to a limited extent across a hybrid zone may contribute to hybrid breakdown between two species. The limited flow of both X- and Y-specific alleles suggest that sex chromosomes have played an important role in Mus speciation.

15.
Development ; 129(19): 4627-34, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12223418

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares , Ovário/embriologia , Processos de Determinação Sexual , Testículo/embriologia , Fatores de Transcrição/metabolismo , Dedos de Zinco , Animais , Biomarcadores , Diferenciação Celular , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Proteínas de Ligação a DNA/genética , Feminino , Fatores de Transcrição Fushi Tarazu , Fator de Transcrição GATA4 , Gônadas , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Homeodomínio , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Complexos Multienzimáticos/genética , Ovário/anormalidades , Progesterona Redutase/genética , Receptores Citoplasmáticos e Nucleares , Fatores de Transcrição SOX9 , Células de Sertoli/citologia , Proteína da Região Y Determinante do Sexo , Esteroide 17-alfa-Hidroxilase/genética , Esteroide Isomerases/genética , Fator Esteroidogênico 1 , Testículo/anormalidades , Fatores de Transcrição/genética , Proteínas WT1/genética
16.
Mamm Genome ; 14(12): 817-29, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14724736

RESUMO

Chromosome deletion complexes in model organisms serve as valuable genetic tools for the functional and physical annotation of complex genomes. Among their many roles, deletions can serve as mapping tools for simple or quantitative trait loci (QTLs), genetic reagents for regional mutagenesis experiments, and, in the case of mice, models of human contiguous gene deletion syndromes. Deletions also are uniquely suited for identifying regions of the genome containing haploinsufficient or imprinted loci. Here we describe the creation of new deletions at the proximal end of mouse Chromosome (Chr) 17 by using the technique of ES cell irradiation and the extensive molecular characterization of these and previously isolated deletions that, in total, cover much of the mouse t complex. The deletions are arranged in five overlapping complexes that collectively span about 25 Mbp. Furthermore, we have integrated each of the deletion complexes with physical data from public and private mouse genome sequences, and our own genetic data, to resolve some discrepancies. These deletions will be useful for characterizing several phenomena related to the t complex and t haplotypes, including transmission ratio distortion, male infertility, and the collection of t haplotype embryonic lethal mutations. The deletions will also be useful for mapping other loci of interest on proximal Chr 17, including T-associated sex reversal ( Tas) and head-tilt ( het). The new deletions have thus far been used to localize the recently identified t haplolethal ( Thl1) locus to an approximately 1.3-Mbp interval.


Assuntos
Deleção Cromossômica , Mapeamento Cromossômico , Cromossomos de Mamíferos/genética , Raios gama , Camundongos/genética , Células-Tronco/efeitos da radiação , Animais , Sequência de Bases , Southern Blotting , Cruzamentos Genéticos , Primers do DNA , Genes Reporter/genética , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microinjeções , Repetições de Microssatélites/genética , Dados de Sequência Molecular , Plasmídeos/genética , Simplexvirus/genética , Timidina Quinase/genética
17.
Mamm Genome ; 15(3): 151-61, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15029877

RESUMO

X-linked hypophosphatemic rickets (XLH) in humans is caused by mutation in the PHEX gene. Previously, three mutations in the mouse Phex gene have been reported: Phex(Hyp), Gy, and Phex(Ska1). Here we report analysis of two new spontaneous mutation in the mouse Phex gene, Phex(Hyp-2J) and Phex(Hyp-Duk). Phex(Hyp-2J) and Phex(Hyp-Duk) involve intragenic deletions of at least 7.3 kb containing exon 15, and 30 kb containing exons 13 and 14, respectively. Both mutations cause similar phenotypes in males, including shortened hind legs and tail, a shortened square trunk, hypophosphatemia, hypocalcemia, and rachitic bone disease. In addition, mice carrying the Phex(Hyp-Duk) mutation exhibit background-dependent variable expression of deafness, circling behavior, and cranial dysmorphology, demonstrating the influence of modifying genes on Phex-related phenotypes. Cochlear cross-sections from Phex(Hyp-2J)/Y and Phex(Hyp-Duk)/Y males reveal a thickening of the temporal bones surrounding the cochlea with the presence of a precipitate in the scala tympani. Evidence of the degeneration of the organ of Corti and spiral ganglion also are present in the hearing-impaired Phex(Hyp-Duk)/Y mice, but not in the normal-hearing Phex(Hyp-2J)/Y mice. Analysis of the phenotypes noted in Phex(Hyp-Duk)/Y and Phex(Hyp-2J)/Y males, together with those noted in Phex(Ska1)/Y and Phex(Hyp)/Y males, now allow XLH-related phenotypes to be separated from non-XLH-related phenotypes, such as those noted in Gy/Y males. Also, identification of the genetic modifiers of hearing and craniofacial dysmorphology in Phex(Hyp-Duk)/Y mice could provide insight into the phenotypic variation of XLH in humans.


Assuntos
Sequência de Bases/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Hipofosfatemia/genética , Camundongos/genética , Fenótipo , Proteínas/genética , Deleção de Sequência/genética , Absorciometria de Fóton , Animais , Southern Blotting , Composição Corporal , Pesos e Medidas Corporais , Densidade Óssea , Cóclea/anormalidades , Primers do DNA , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico , Técnicas Histológicas , Masculino , Camundongos/anormalidades , Camundongos Endogâmicos C57BL , Endopeptidase Neutra Reguladora de Fosfato PHEX , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Crânio/anormalidades
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA