Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Reproduction ; 163(6): 333-340, 2022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-35315790

RESUMEN

Sex determination in mammals is controlled by the dominance of either pro-testis (SRY-SOX9-FGF9) or pro-ovary (RSPO1-WNT4-FOXL2) genetic pathways during early gonad development in XY and XX embryos, respectively. We have previously shown that early, robust expression of mouse Sry is dependent on the nuclear protein GADD45g. In the absence of GADD45g, XY gonadal sex reversal occurs, associated with a major reduction of Sry levels at 11.5 dpc. Here, we probe the relationship between Gadd45g and Sry further, using gain- and loss-of-function genetics. First, we show that transgenic Gadd45g overexpression can elevate Sry expression levels at 11.5 dpc in the B6.YPOS model of sex reversal, resulting in phenotypic rescue. We then show that the zygosity of pro-ovarian Rspo1 is critical for the degree of gonadal sex reversal observed in both B6.YPOS and Gadd45g-deficient XY gonads, in contrast to that of Foxl2. Phenotypic rescue of sex reversal is observed in XY gonads lacking both Gadd45g and Rspo1, but this is not associated with rescue of Sry expression levels at 11.5 dpc. Instead, Sox9 levels are rescued by around 12.5 dpc. We conclude that Gadd45g is absolutely required for timely expression of Sry in XY gonads, independently of RSPO1-mediated WNT signalling, and discuss these data in light of our understanding of antagonistic interactions between the pro-testis and pro-ovary pathways.


Asunto(s)
Gónadas , Factor de Transcripción SOX9 , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Gónadas/metabolismo , Masculino , Mamíferos/genética , Ratones , Ovario/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Procesos de Determinación del Sexo , Diferenciación Sexual , Proteína de la Región Y Determinante del Sexo/genética , Proteína de la Región Y Determinante del Sexo/metabolismo , Testículo/metabolismo , Trombospondinas/genética , Trombospondinas/metabolismo , Vía de Señalización Wnt
2.
Eur J Endocrinol ; 186(1): 65-72, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34714774

RESUMEN

CONTEXT: Homozygous and heterozygous variants in PPP2R3C are associated with syndromic 46,XY complete gonadal dysgenesis (Myo-Ectodermo-Gonadal Dysgenesis (MEGD) syndrome), and impaired spermatogenesis, respectively. This study expands the role of PPP2R3C in the aetiology of gonadal dysgenesis (GD). METHOD: We sequenced the PPP2R3C gene in four new patients from three unrelated families. The clinical, laboratory, and molecular characteristics were investigated. We have also determined the requirement for Ppp2r3c in mice (C57BL6/N) using CRISPR/Cas9 genome editing. RESULTS: A homozygous c.578T>C (p.L193S) PPP2R3C variant was identified in one 46,XX girl with primary gonadal insufficiency, two girls with 46,XY complete GD, and one undervirilised boy with 46,XY partial GD. The patients with complete GD had low gonadal and adrenal androgens, low anti-Müllerian hormone, and high follicle-stimulating hormone and luteinizing hormone concentrations. All patients manifested characteristic features of MEGD syndrome. Heterozygous Ppp2r3c knockout mice appeared overtly normal and fertile. Inspection of homozygous embryos at 14.5, 9.5, and 8.5 days post coitum(dpc) revealed evidence of dead embryos. We conclude that loss of function of Ppp2r3c is not compatible with viability in mice and results in embryonic death from 7.5 dpc or earlier. CONCLUSION: Our data indicate the essential roles for PPP2R3C in mouse and human development. Germline homozygous variants in human PPP2R3C are associated with distinctive syndromic GD of varying severity in both 46,XY and 46,XX individuals.


Asunto(s)
Disgenesia Gonadal 46 XX/genética , Disgenesia Gonadal 46 XY/genética , Proteína Fosfatasa 2/genética , Sustitución de Aminoácidos , Animales , Niño , Consanguinidad , Embrión de Mamíferos , Femenino , Disgenesia Gonadal 46 XX/patología , Disgenesia Gonadal 46 XY/patología , Homocigoto , Humanos , Leucina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense , Linaje , Embarazo , Serina/genética
3.
Nat Commun ; 12(1): 3804, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34155196

RESUMEN

In mammalian genomes, differentially methylated regions (DMRs) and histone marks including trimethylation of histone 3 lysine 27 (H3K27me3) at imprinted genes are asymmetrically inherited to control parentally-biased gene expression. However, neither parent-of-origin-specific transcription nor imprints have been comprehensively mapped at the blastocyst stage of preimplantation development. Here, we address this by integrating transcriptomic and epigenomic approaches in mouse preimplantation embryos. We find that seventy-one genes exhibit previously unreported parent-of-origin-specific expression in blastocysts (nBiX: novel blastocyst-imprinted expressed). Uniparental expression of nBiX genes disappears soon after implantation. Micro-whole-genome bisulfite sequencing (µWGBS) of individual uniparental blastocysts detects 859 DMRs. We further find that 16% of nBiX genes are associated with a DMR, whereas most are associated with parentally-biased H3K27me3, suggesting a role for Polycomb-mediated imprinting in blastocysts. nBiX genes are clustered: five clusters contained at least one published imprinted gene, and five clusters exclusively contained nBiX genes. These data suggest that early development undergoes a complex program of stage-specific imprinting involving different tiers of regulation.


Asunto(s)
Blastocisto/metabolismo , Impresión Genómica/genética , Histonas/metabolismo , Alelos , Animales , Blastocisto/citología , Metilación de ADN , Desarrollo Embrionario/genética , Femenino , Expresión Génica , Células Germinativas/metabolismo , Estratos Germinativos/metabolismo , Haploidia , Masculino , Metilación , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Familia de Multigenes , Sitio de Iniciación de la Transcripción
4.
Genet Med ; 22(1): 150-159, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31337883

RESUMEN

PURPOSE: XY individuals with disorders/differences of sex development (DSD) are characterized by reduced androgenization caused, in some children, by gonadal dysgenesis or testis regression during fetal development. The genetic etiology for most patients with 46,XY gonadal dysgenesis and for all patients with testicular regression syndrome (TRS) is unknown. METHODS: We performed exome and/or Sanger sequencing in 145 individuals with 46,XY DSD of unknown etiology including gonadal dysgenesis and TRS. RESULTS: Thirteen children carried heterozygous missense pathogenic variants involving the RNA helicase DHX37, which is essential for ribosome biogenesis. Enrichment of rare/novel DHX37 missense variants in 46,XY DSD is highly significant compared with controls (P value = 5.8 × 10-10). Five variants are de novo (P value = 1.5 × 10-5). Twelve variants are clustered in two highly conserved functional domains and were specifically associated with gonadal dysgenesis and TRS. Consistent with a role in early testis development, DHX37 is expressed specifically in somatic cells of the developing human and mouse testis. CONCLUSION: DHX37 pathogenic variants are a new cause of an autosomal dominant form of 46,XY DSD, including gonadal dysgenesis and TRS, showing that these conditions are part of a clinical spectrum. This raises the possibility that some forms of DSD may be a ribosomopathy.


Asunto(s)
Disgenesia Gonadal 46 XY/genética , Mutación Missense , ARN Helicasas/genética , Análisis de Secuencia de ADN/métodos , Testículo/crecimiento & desarrollo , Adolescente , Animales , Preescolar , Femenino , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Recién Nacido , Masculino , Ratones , Mutagénesis Sitio-Dirigida , Tasa de Mutación , Dominios Proteicos , ARN Helicasas/química , Testículo/metabolismo , Adulto Joven
5.
Genetics ; 214(2): 467-477, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31836612

RESUMEN

XY C57BL/6J (B6) mice harboring a Mus musculus domesticus-type Y chromosome (Y POS ), known as B6.Y POS mice, commonly undergo gonadal sex reversal and develop as phenotypic females. In a minority of cases, B6.Y POS males are identified and a proportion of these are fertile. This phenotypic variability on a congenic B6 background has puzzled geneticists for decades. Recently, a B6.Y POS colony was shown to carry a non-B6-derived region of chromosome 11 that protected against B6.Y POS sex reversal. Here. we show that a B6.Y POS colony bred and archived at the MRC Harwell Institute lacks the chromosome 11 modifier but instead harbors an ∼37 Mb region containing non-B6-derived segments on chromosome 13. This region, which we call Mod13, protects against B6.Y POS sex reversal in a proportion of heterozygous animals through its positive and negative effects on gene expression during primary sex determination. We discuss Mod13's influence on the testis determination process and its possible origin in light of sequence similarities to that region in other mouse genomes. Our data reveal that the B6.Y POS sex reversal phenomenon is genetically complex and the explanation of observed phenotypic variability is likely dependent on the breeding history of any local colony.


Asunto(s)
Disgenesia Gonadal 46 XY/genética , Procesos de Determinación del Sexo/genética , Cromosoma Y/genética , Animales , Cromosomas Humanos Par 13/genética , Cromosomas Humanos Par 13/metabolismo , Proteínas de Unión al ADN/genética , Trastornos del Desarrollo Sexual/genética , Trastornos del Desarrollo Sexual/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Genoma , Disgenesia Gonadal 46 XY/metabolismo , Gónadas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Ovario/metabolismo , Testículo/metabolismo , Factores de Transcripción/genética
6.
Sci Rep ; 9(1): 17195, 2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31748609

RESUMEN

Adamts16 encodes a disintegrin-like and metalloproteinase with thrombospondin motifs, 16, a member of a family of multi-domain, zinc-binding proteinases. ADAMTS-16 is implicated in a number of pathological conditions, including hypertension, cancer and osteoarthritis. A large number of observations, including a recent report of human ADAMTS16 variants in cases of 46,XY disorders/differences of sex development (DSD), also implicate this gene in human testis determination. We used CRISPR/Cas9 genome editing to generate a loss-of-function allele in the mouse in order to examine whether ADAMTS-16 functions in mouse testis determination or testicular function. Male mice lacking Adamts16 on the C57BL/6N background undergo normal testis determination in the fetal period. However, adult homozygotes have an average testis weight that is around 10% lower than age-matched controls. Cohorts of mutant males tested at 3-months and 6-months of age were fertile. We conclude that ADAMTS-16 is not required for testis determination or male fertility in mice. We discuss these phenotypic data and their significance for our understanding of ADAMTS-16 function.


Asunto(s)
Proteínas ADAMTS/fisiología , Infertilidad Masculina/prevención & control , Testículo/anatomía & histología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Testículo/embriología
7.
Sci Rep ; 8(1): 17318, 2018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30470800

RESUMEN

Bacterial artificial chromosomes (BACs) offer a means of manipulating gene expression and tagging gene products in the mammalian genome without the need to alter endogenous gene structure and risk deleterious phenotypic consequences. However, for a BAC clone to be useful for such purposes it must be shown to contain all the regulatory elements required for normal gene expression and allow phenotypic rescue in the absence of an endogenous gene. Here, we report identification of a functional BAC containing Gadd45g, a gene implicated in DNA repair, DNA demethylation and testis determination in mice and exhibiting a broad pattern of embryonic expression. Mouse fetuses lacking the endogenous Gadd45g gene undergo normal testis development in the presence of the Gadd45g BAC transgene. Moreover, a survey of embryonic Gadd45g expression from the BAC reveals that all reported sites of expression are maintained. This functional BAC can now be used for subsequent manipulation of the Gadd45g gene with the confidence that regulatory elements required for embryonic expression, including testis determination, are present. We describe the generation and characterisation of a Gadd45g-mCherry fluorescent reporter exhibiting strong expression in developing gonads and neural tissue, recapitulating endogenous gene expression, as evidence of this.


Asunto(s)
Cromosomas Artificiales Bacterianos , Regulación del Desarrollo de la Expresión Génica , Ingeniería Genética , Péptidos y Proteínas de Señalización Intracelular/genética , Secuencias Reguladoras de Ácidos Nucleicos , Testículo/crecimiento & desarrollo , Transgenes , Animales , Masculino , Ratones , Ratones Transgénicos , Testículo/metabolismo , Proteinas GADD45
8.
Proc Natl Acad Sci U S A ; 115(21): 5474-5479, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29735715

RESUMEN

Mammalian sex determination is controlled by the antagonistic interactions of two genetic pathways: The SRY-SOX9-FGF9 network promotes testis determination partly by opposing proovarian pathways, while RSPO1/WNT-ß-catenin/FOXL2 signals control ovary development by inhibiting SRY-SOX9-FGF9. The molecular basis of this mutual antagonism is unclear. Here we show that ZNRF3, a WNT signaling antagonist and direct target of RSPO1-mediated inhibition, is required for sex determination in mice. XY mice lacking ZNRF3 exhibit complete or partial gonadal sex reversal, or related defects. These abnormalities are associated with ectopic WNT/ß-catenin activity and reduced Sox9 expression during fetal sex determination. Using exome sequencing of individuals with 46,XY disorders of sex development, we identified three human ZNRF3 variants in very rare cases of XY female presentation. We tested two missense variants and show that these disrupt ZNRF3 activity in both human cell lines and zebrafish embryo assays. Our data identify a testis-determining function for ZNRF3 and indicate a mechanism of direct molecular interaction between two mutually antagonistic organogenetic pathways.


Asunto(s)
Trastornos del Desarrollo Sexual/genética , Diferenciación Sexual , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/fisiología , Proteínas Wnt/antagonistas & inhibidores , beta Catenina/antagonistas & inhibidores , Adolescente , Adulto , Animales , Células Cultivadas , Trastornos del Desarrollo Sexual/patología , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Gónadas/metabolismo , Gónadas/patología , Humanos , Masculino , Ratones , Mutación Missense , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Testículo/metabolismo , Testículo/patología , Trombospondinas/genética , Trombospondinas/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Adulto Joven , Pez Cebra , beta Catenina/genética , beta Catenina/metabolismo
9.
Biol Reprod ; 94(5): 103, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27009039

RESUMEN

Testis determination in mammals is initiated by expression of SRY in somatic cells of the embryonic gonad. Genetic analyses in the mouse have revealed a requirement for mitogen-activated protein kinase (MAPK) signaling in testis determination: targeted loss of the kinases MAP3K4 and p38 MAPK causes complete XY embryonic gonadal sex reversal. These kinases occupy positions at the top and bottom level, respectively, in the canonical three-tier MAPK-signaling cascade: MAP3K, MAP2K, MAPK. To date, no role in sex determination has been attributed to a MAP2K, although such a function is predicted to exist. Here, we report roles for the kinases MAP2K3 and MAP2K6 in testis determination. C57BL/6J (B6) embryos lacking MAP2K3 exhibited no significant abnormalities of testis development, whilst those lacking MAP2K6 exhibited a minor delay in testis determination. Compound mutants lacking three out of four functional alleles at the two loci also exhibited delayed testis determination and transient ovotestis formation as a consequence, suggestive of partially redundant roles for these kinases in testis determination. Early lethality of double-knockout embryos precludes analysis of sexual development. To reveal their roles in testis determination more clearly, we generated Map2k mutant B6 embryos using a weaker Sry allele (Sry(AKR)). Loss of Map2k3 on this highly sensitized background exacerbates ovotestis development, whilst loss of Map2k6 results in complete XY gonadal sex reversal associated with reduction of Sry expression at 11.25 days postcoitum. Our data suggest that MAP2K6 functions in mouse testis determination, via positive effects on Sry, and also indicate a minor role for MAP2K3.


Asunto(s)
MAP Quinasa Quinasa 3/fisiología , MAP Quinasa Quinasa 6/fisiología , Procesos de Determinación del Sexo/genética , Proteína de la Región Y Determinante del Sexo/metabolismo , Testículo/embriología , Animales , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , MAP Quinasa Quinasa 3/genética , MAP Quinasa Quinasa 6/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Diferenciación Sexual/genética , Proteína de la Región Y Determinante del Sexo/genética , Testículo/metabolismo
10.
PLoS One ; 9(6): e100447, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24956260

RESUMEN

The secreted molecule fibroblast growth factor 9 (FGF9) plays a critical role in testis determination in the mouse. In embryonic gonadal somatic cells it is required for maintenance of SOX9 expression, a key determinant of Sertoli cell fate. Conditional gene targeting studies have identified FGFR2 as the main gonadal receptor for FGF9 during sex determination. However, such studies can be complicated by inefficient and variable deletion of floxed alleles, depending on the choice of Cre deleter strain. Here, we report a novel, constitutive allele of Fgfr2, hobbyhorse (hob), which was identified in an ENU-based forward genetic screen for novel testis-determining loci. Fgr2hob is caused by a C to T mutation in the invariant exon 7, resulting in a polypeptide with a mis-sense mutation at position 263 (Pro263Ser) in the third extracellular immunoglobulin-like domain of FGFR2. Mutant homozygous embryos show severe limb and lung defects and, when on the sensitised C57BL/6J (B6) genetic background, undergo complete XY gonadal sex reversal associated with failure to maintain expression of Sox9. Genetic crosses employing a null mutant of Fgfr2 suggest that Fgr2hob is a hypomorphic allele, affecting both the FGFR2b and FGFR2c splice isoforms of the receptor. We exploited the consistent phenotype of this constitutive mutant by analysing MAPK signalling at the sex-determining stage of gonad development, but no significant abnormalities in mutant embryos were detected.


Asunto(s)
Extremidades/patología , Disgenesia Gonadal/genética , Gónadas/anomalías , Enfermedades Pulmonares/patología , Mutación/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Secuencia de Aminoácidos , Animales , Western Blotting , Células Cultivadas , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Femenino , Gónadas/metabolismo , Gónadas/patología , Homocigoto , Humanos , Técnicas para Inmunoenzimas , Enfermedades Pulmonares/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Fenotipo , Homología de Secuencia de Aminoácido
11.
Hum Mol Genet ; 23(11): 3035-44, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24452333

RESUMEN

Disorders of sex development in the human population range in severity from mild genital defects to gonadal sex reversal. XY female development has been associated with heterozygous mutations in several genes, including SOX9, WT1 and MAP3K1. In contrast, XY sex reversal in mice usually requires complete absence of testis-determining gene products. One exception to this involves T-associated sex reversal (Tas), a phenomenon characterized by the formation of ovotestes or ovaries in XY mice hemizygous for the hairpin-tail (T(hp)) or T-Orleans (T(Orl)) deletions on proximal mouse chromosome 17. We recently reported that mice heterozygous for a null allele of Map3k4, which resides in the T(hp) deletion, exhibit XY ovotestis development and occasional gonadal sex reversal on the sensitized C57BL/6J-Y(AKR) (B6-Y(AKR)) genetic background, reminiscent of the Tas phenotype. However, these experiments did not exclude the possibility that loss of other loci in the T(hp) deletion, or other effects of the deletion itself, might contribute to Tas. Here, we show that disruption to Sry expression underlies XY gonadal defects in B6-Y(AKR) embryos harbouring the T(hp) deletion and that a functional Map3k4 bacterial artificial chromosome rescues these abnormalities by re-establishing a normal Sry expression profile. These data demonstrate that Map3k4 haploinsufficiency is the cause of T-associated sex reversal and that levels of this signalling molecule are a major determinant of the expression profile of Sry.


Asunto(s)
Trastornos del Desarrollo Sexual/enzimología , MAP Quinasa Quinasa Quinasa 4/metabolismo , Procesos de Determinación del Sexo , Animales , Trastornos del Desarrollo Sexual/genética , Femenino , Humanos , MAP Quinasa Quinasa Quinasa 4/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovario/metabolismo , Testículo/metabolismo
12.
Hum Mol Genet ; 22(13): 2553-65, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23459932

RESUMEN

Otitis media with effusion (OME) is the most common cause of hearing loss in children and tympanostomy to alleviate the condition remains the commonest surgical intervention in children in the developed world. Chronic and recurrent forms of OM are known to have a very significant genetic component, however, until recently little was known of the underlying genes involved. The identification of mouse models of chronic OM has indicated a role of transforming growth factor beta (TGFß) signalling and its impact on responses to hypoxia in the inflamed middle ear. We have, therefore, investigated the role of TGFß signalling and identified and characterized a new model of chronic OM carrying a mutation in the gene for transforming growth interacting factor 1 (Tgif1). Tgif1 homozygous mutant mice have significantly raised auditory thresholds due to a conductive deafness arising from a chronic effusion starting at around 3 weeks of age. The OM is accompanied by a significant thickening of the middle ear mucosa lining, expansion of mucin-secreting goblet cell populations and raised levels of vascular endothelial growth factor, TNF-α and IL-1ß in ear fluids. We also identified downstream effects on TGFß signalling in middle ear epithelia at the time of development of chronic OM. Both phosphorylated SMAD2 and p21 levels were lowered in the homozygous mutant, demonstrating a suppression of the TGFß pathway. The identification and characterization of the Tgif mutant supports the role of TGFß signalling in the development of chronic OM and provides an important candidate gene for genetic studies in the human population.


Asunto(s)
Proteínas de Homeodominio/genética , Otitis Media/genética , Otitis Media/metabolismo , Proteínas Represoras/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Anomalías Craneofaciales/genética , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Oído Medio/metabolismo , Oído Medio/patología , Células Epiteliales/metabolismo , Femenino , Genotipo , Células Ciliadas Auditivas/patología , Células Ciliadas Auditivas/ultraestructura , Pérdida Auditiva/genética , Homocigoto , Masculino , Ratones , Ratones Noqueados , Mutación , Otitis Media/patología , Fenotipo , Placenta/metabolismo , Embarazo
13.
Dev Cell ; 23(5): 1020-31, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23102580

RESUMEN

Loss of the kinase MAP3K4 causes mouse embryonic gonadal sex reversal due to reduced expression of the testis-determining gene, Sry. However, because of widespread expression of MAP3K4, the cellular basis of this misregulation was unclear. Here, we show that mice lacking Gadd45γ also exhibit XY gonadal sex reversal caused by disruption to Sry expression. Gadd45γ is expressed in a dynamic fashion in somatic cells of the developing gonads from 10.5 days postcoitum (dpc) to 12.5 dpc. Gadd45γ and Map3k4 genetically interact during sex determination, and transgenic overexpression of Map3k4 rescues gonadal defects in Gadd45γ-deficient embryos. Sex reversal in both mutants is associated with reduced phosphorylation of p38 MAPK and GATA4. In addition, embryos lacking both p38α and p38ß also exhibit XY gonadal sex reversal. Taken together, our data suggest a requirement for GADD45γ in promoting MAP3K4-mediated activation of p38 MAPK signaling in embryonic gonadal somatic cells for testis determination in the mouse.


Asunto(s)
Proteínas Portadoras/metabolismo , MAP Quinasa Quinasa Quinasa 4/metabolismo , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Proteína de la Región Y Determinante del Sexo/genética , Testículo/embriología , Testículo/metabolismo , Animales , Proteínas Portadoras/genética , Metilación de ADN , Femenino , Factor de Transcripción GATA4/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes sry , Disgenesia Gonadal 46 XY/embriología , Disgenesia Gonadal 46 XY/genética , Disgenesia Gonadal 46 XY/metabolismo , Péptidos y Proteínas de Señalización Intracelular , MAP Quinasa Quinasa Quinasa 4/deficiencia , MAP Quinasa Quinasa Quinasa 4/genética , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 11 Activada por Mitógenos/deficiencia , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/genética , Modelos Biológicos , Procesos de Determinación del Sexo/genética , Procesos de Determinación del Sexo/fisiología
14.
Wiley Interdiscip Rev Dev Biol ; 1(4): 559-77, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23801533

RESUMEN

The mammalian gonad is adapted for the production of germ cells and is an endocrine gland that controls sexual maturation and fertility. Gonadal sex reversal, namely, the development of ovaries in an XY individual or testes in an XX, has fascinated biologists for decades. The phenomenon suggests the existence of genetic suppressors of the male and female developmental pathways and molecular genetic studies, particularly in the mouse, have revealed controlled antagonism at the core of mammalian sex determination. Both testis and ovary determination represent design solutions to a number of problems: how to generate cells with the right properties to populate the organ primordium; how to produce distinct organs from an initially bipotential primordium; how to pattern an organ when the expression of key cell fate determinants is initiated only in a discrete region of the primordium and extends to other regions asynchronously; how to coordinate the interaction between distinct cell types in time and space and stabilize the resulting morphology; and how to maintain the differentiated state of the organ throughout the adult period. Some of these, and related problems, are common to organogenesis in general; some are distinctive to gonad development. In this review, we discuss recent studies of the molecular and cellular events underlying testis and ovary development, with an emphasis on the phenomenon of gonadal sex reversal and its causes in mice and humans. Finally, we discuss sex-determining loci and disorders of sex development in humans and the future of research in this important area.


Asunto(s)
Diferenciación Celular , Trastornos del Desarrollo Sexual/metabolismo , Procesos de Determinación del Sexo/genética , Animales , Linaje de la Célula , Trastornos del Desarrollo Sexual/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Gónadas/citología , Gónadas/embriología , Gónadas/crecimiento & desarrollo , Humanos , Masculino , Ratones
15.
PLoS One ; 6(5): e19572, 2011 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-21559298

RESUMEN

In mammals, the Y chromosome is a dominant male determinant, causing the bipotential gonad to develop as a testis. Recently, cases of familial and spontaneous 46,XY disorders of sex development (DSD) have been attributed to mutations in the human gene encoding mitogen-activated protein kinase kinase kinase 1, MAP3K1, a component of the mitogen-activated protein kinase (MAPK) signal transduction pathway. In individuals harbouring heterozygous mutations in MAP3K1, dysregulation of MAPK signalling was observed in lymphoblastoid cell lines, suggesting a causal role for these mutations in disrupting XY sexual development. Mice lacking the cognate gene, Map3k1, are viable and exhibit the eyes open at birth (EOB) phenotype on a mixed genetic background, but on the C57BL/6J genetic background most mice die at around 14.5 dpc due to a failure of erythropoiesis in the fetal liver. However, no systematic examination of sexual development in Map3k1-deficient mice has been described, an omission that is especially relevant in the case of C57BL/6J, a genetic background that is sensitized to disruptions to testis determination. Here, we report that on a mixed genetic background mice lacking Map3k1 are fertile and exhibit no overt abnormalities of testis development. On C57BL/6J, significant non-viability is observed with very few animals surviving to adulthood. However, an examination of development in Map3k1-deficient XY embryos on this genetic background revealed no significant defects in testis determination, although minor abnormalities were observed, including an increase in gonadal length. Based on these observations, we conclude that MAP3K1 is not required for mouse testis determination. We discuss the significance of these data for the functional interpretation of sex-reversing MAP3K1 mutations in humans.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Quinasa 1 de Quinasa de Quinasa MAP/fisiología , Testículo/anomalías , Animales , Trastornos del Desarrollo Sexual/genética , Femenino , Genotipo , Homocigoto , Quinasa 1 de Quinasa de Quinasa MAP/genética , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Mutación , Fenotipo , Diferenciación Sexual/genética , Testículo/embriología , Factores de Tiempo
16.
PLoS Biol ; 7(9): e1000196, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19753101

RESUMEN

Sex determination in mammals is controlled by the presence or absence of the Y-linked gene SRY. In the developing male (XY) gonad, sex-determining region of the Y (SRY) protein acts to up-regulate expression of the related gene, SOX9, a transcriptional regulator that in turn initiates a downstream pathway of testis development, whilst also suppressing ovary development. Despite the requirement for a number of transcription factors and secreted signalling molecules in sex determination, intracellular signalling components functioning in this process have not been defined. Here we report a role for the phylogenetically ancient mitogen-activated protein kinase (MAPK) signalling pathway in mouse sex determination. Using a forward genetic screen, we identified the recessive boygirl (byg) mutation. On the C57BL/6J background, embryos homozygous for byg exhibit consistent XY gonadal sex reversal. The byg mutation is an A to T transversion causing a premature stop codon in the gene encoding MAP3K4 (also known as MEKK4), a mitogen-activated protein kinase kinase kinase. Analysis of XY byg/byg gonads at 11.5 d post coitum reveals a growth deficit and a failure to support mesonephric cell migration, both early cellular processes normally associated with testis development. Expression analysis of mutant XY gonads at the same stage also reveals a dramatic reduction in Sox9 and, crucially, Sry at the transcript and protein levels. Moreover, we describe experiments showing the presence of activated MKK4, a direct target of MAP3K4, and activated p38 in the coelomic region of the XY gonad at 11.5 d post coitum, establishing a link between MAPK signalling in proliferating gonadal somatic cells and regulation of Sry expression. Finally, we provide evidence that haploinsufficiency for Map3k4 accounts for T-associated sex reversal (Tas). These data demonstrate that MAP3K4-dependent signalling events are required for normal expression of Sry during testis development, and create a novel entry point into the molecular and cellular mechanisms underlying sex determination in mice and disorders of sexual development in humans.


Asunto(s)
MAP Quinasa Quinasa Quinasa 4/deficiencia , Sistema de Señalización de MAP Quinasas , Procesos de Determinación del Sexo , Animales , Trastornos del Desarrollo Sexual , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , MAP Quinasa Quinasa Quinasa 4/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ovario/citología , Ovario/embriología , Mutación Puntual , Proteína de la Región Y Determinante del Sexo/genética , Proteína de la Región Y Determinante del Sexo/metabolismo , Testículo/citología , Testículo/embriología
17.
Dev Biol ; 326(2): 273-84, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19100252

RESUMEN

Secreted frizzled-related proteins (Sfrps) are antagonists of WNT signalling implicated in a variety of biological processes. However, there are no reports of a direct role for Sfrps in embryonic organogenesis in mammals. Using in vivo loss-of-function studies we report here for the first time a redundant role for Sfrp1 and Sfrp2 in embryonic sexual development of the mouse. At 16.5 dpc, male embryos lacking both genes exhibit multiple defects in gonad morphology, reproductive tract maturation and gonad positioning. Abnormal positioning of the testis appears to be due to failed gubernaculum development and an unusually close association between the cranial end of the reproductive tract and the kidney. The testes of double homozygotes are smaller than controls, contain fewer cords from the earliest stages, but still express Insl3, which encodes the hormone required for gubernacular masculinisation. Lgr8, which encodes the Insl3 receptor, is also expressed in the mutant gubernaculum, suggesting that Sfrp1/Sfrp2 signalling is not required for expression of the ligand or receptor that controls transabdominal testicular descent. Similarities between the abnormalities of embryonic sexual development in Sfrp1(-/-)Sfrp2(-/-) embryos with those exhibited by the Looptail and Wnt5a mutants suggest that disrupted non-canonical Wnt signalling may cause these defects.


Asunto(s)
Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Genitales Masculinos/anatomía & histología , Genitales Masculinos/embriología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Embrión de Mamíferos/anomalías , Femenino , Genitales Masculinos/anomalías , Genitales Masculinos/crecimiento & desarrollo , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Morfogénesis , Embarazo , Testosterona/metabolismo
18.
PLoS One ; 3(12): e4091, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19116663

RESUMEN

The mammalian gonad arises as a bipotential primordium from which a testis or ovary develops depending on the chromosomal sex of the individual. We have previously used DNA microarrays to screen for novel genes controlling the developmental fate of the indifferent embryonic mouse gonad. Maestro (Mro), which encodes a HEAT-repeat protein, was originally identified as a gene exhibiting sexually dimorphic expression during mouse gonad development. Wholemount in situ hybridisation analysis revealed Mro to be expressed in the embryonic male gonad from approximately 11.5 days post coitum, prior to overt sexual differentiation. No significant expression was detected in female gonads at the same developmental stage. In order to address its physiological function, we have generated mice lacking Maestro using gene targeting. Male and female mice homozygous for a Mro null allele are viable and fertile. We examined gonad development in homozygous male embryos in detail and observed no differences when compared to wild-type controls. Immunohistochemical analysis of homozygous mutant testes of adult mice revealed no overt abnormalities. Expression profiling using DNA microarrays also indicated no significant differences between homozygote embryonic male gonads and controls. We conclude that Maestro is dispensable for normal male sexual development and fertility in laboratory mice; however, the Mro locus itself does have utility as a site for insertion of transgenes for future studies in the fields of sexual development and Sertoli cell function.


Asunto(s)
Embrión de Mamíferos/metabolismo , Fertilidad/genética , Diferenciación Sexual/genética , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Gónadas/embriología , Gónadas/metabolismo , Homocigoto , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Mutación , Células de Sertoli/metabolismo , Testículo/embriología , Testículo/metabolismo
19.
Genesis ; 40(2): 109-117, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15384171

RESUMEN

Dominantly acting mutations that produce visible phenotypes are frequently recovered, either during routine maintenance of colonies or from mutagenesis experiments. We have studied 12 dominant mouse mutations that cause a tail dysmorphology, a coat spotting phenotype, or a combination of these. The majority of these mutations act in a semidominant manner with the homozygous state associated with embryonic lethality and a visible phenotype at or before midgestation. The homozygous phenotypes include axis truncation and neural crest cell defects, as may be expected from the heterozygous phenotypes. The majority of mutations, however, also produced other phenotypes that include neural tube closure defects and aberrant heart looping. In one coat spotting mutant the homozygous condition is lethal before neural crest cell production commences. The mutated genes often function in processes additional to those alluded to by the heterozygous phenotype.


Asunto(s)
Desarrollo Embrionario/genética , Genes Dominantes , Mutación , Alquilantes/farmacología , Animales , Animales Congénicos , Biomarcadores , Mapeo Cromosómico , Etilnitrosourea/farmacología , Femenino , Genes Letales , Marcadores Genéticos , Genoma , Color del Cabello/genética , Haplotipos , Homocigoto , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Mutantes , Mutágenos/farmacología , Polimorfismo Genético , Cola (estructura animal)/anomalías
20.
Dev Biol ; 268(1): 7-23, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15031101

RESUMEN

Although the gross embryology of inner ear development has been documented for several different vertebrate species at a descriptive level, our understanding of the molecular mechanisms involved remains rudimentary. Therefore, we have used cDNA subtraction and normalization procedures to define genes upregulated in the 13.5dpc mouse inner ear, a developmental stage where inner ear morphogenesis and tissue remodeling is active and differentiation of future hair cells is being initiated. We recovered 33 different genes from this subtraction and using gene-specific primers have confirmed the transcriptional upregulation of 26 of these in the 13.5dpc inner ear. Northern analyses were used to investigate splicing differences between the inner ear and the whole embryo at 13.5dpc. Spatial localization of expression was determined through whole-ear in situ hybridization analysis, and selected genes were analyzed in more detail through in situ hybridization of tissue sections. These data illustrate that the genes isolated in this study are expressed in the developing otic capsule and/or neuroepithelium. Furthermore, the expression patterns also reveal molecular heterogeneity in the developing capsule and indicate that for some genes, the chondrogenic otic capsule is composed of distinct domains of gene expression.


Asunto(s)
ADN Complementario/genética , Oído Interno/metabolismo , Perfilación de la Expresión Génica , Animales , Oído Interno/embriología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , ARN Mensajero/genética , Técnica de Sustracción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA