Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34301885

RESUMEN

Germ cells form the basis for sexual reproduction by producing gametes. In ovaries, primordial germ cells exit the cell cycle and the pluripotency-associated state, differentiate into oogonia, and initiate meiosis. Despite the importance of germ cell differentiation for sexual reproduction, signaling pathways regulating their fate remain largely unknown. Here, we show in mouse embryonic ovaries that germ cell-intrinsic ß-catenin activity maintains pluripotency and that its repression is essential to allow differentiation and meiosis entry in a timely manner. Accordingly, in ß-catenin loss-of-function and gain-of-function mouse models, the germ cells precociously enter meiosis or remain in the pluripotent state, respectively. We further show that interaction of ß-catenin and the pluripotent-associated factor POU5F1 in the nucleus is associated with germ cell pluripotency. The exit of this complex from the nucleus correlates with germ cell differentiation, a process promoted by the up-regulation of Znrf3, a negative regulator of WNT/ß-catenin signaling. Together, these data identify the molecular basis of the transition from primordial germ cells to oogonia and demonstrate that ß-catenin is a central gatekeeper in ovarian differentiation and gametogenesis.


Asunto(s)
Diferenciación Celular , Células Germinativas/citología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Femenino , Células Germinativas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/metabolismo , Proteínas Wnt/genética , beta Catenina/genética
2.
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
3.
Reproduction ; 162(1): F69-F78, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33878027

RESUMEN

The birth of Dolly the sheep in 1996 elicited a tsunami of commentaries, both in the popular media and academic journals, including responses to the prospect of human reproductive cloning. Much of the anxiety expressed over this imagined consequence of Dolly's genesis revealed fundamental concerns about us losing our commitments to certain ethical goods, such as human dignity, or even 'what it means to be human'. Over the last 25 years, the focus of much of the ethical debate over human biotechnology has slowly shifted towards other genetic technologies that aim to influence inheritance, such as mitochondrial replacement techniques (MRT) and heritable genome editing. Genome editing, in particular, is a technology with multiple fields of application, actual and potential, in research and innovation. This review suggests that many of the fundamental concerns about the possibility of human reproductive cloning that were precipitated by Dolly persist today in the arguments of those who oppose MRT and any use of heritable human genome editing (HHGE). Whilst it is not accepted here that an understanding of human nature and dignity alone can demonstrate the ethical unacceptability of such assisted reproductive technologies, there are themes of justice, which extend into our relationships with animals, that demand continued wide-ranging examination and public dialogue. While Dolly has cast a long shadow over such discussions, this review suggests that the general existential angst over human uses of biotechnology that she came to symbolise is neither compulsory nor a reliable guide for how to think about biotechnologies today.


Asunto(s)
Animales Modificados Genéticamente/genética , Clonación de Organismos/veterinaria , Edición Génica , Genoma , Ganado/genética , Mitocondrias/genética , Técnicas de Transferencia Nuclear/ética , Animales , Animales Modificados Genéticamente/crecimiento & desarrollo , Aniversarios y Eventos Especiales , Núcleo Celular/genética , Ganado/crecimiento & desarrollo , Técnicas de Transferencia Nuclear/veterinaria
4.
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
5.
Hum Mol Genet ; 27(1): 190-198, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29145650

RESUMEN

CREB-binding protein (CBP, CREBBP, KAT3A) and its closely related paralogue p300 (EP300, KAT3B), together termed p300/CBP, are histone/lysine acetyl-transferases that control gene expression by modifying chromatin-associated proteins. Here, we report roles for both of these chromatin-modifying enzymes in mouse sex determination, the process by which the embryonic gonad develops into a testis or an ovary. By targeting gene ablation to embryonic gonadal somatic cells using an inducible Cre line, we show that gonads lacking either gene exhibit major abnormalities of XY gonad development at 14.5 dpc, including partial sex reversal. Embryos lacking three out of four functional copies of p300/Cbp exhibit complete XY gonadal sex reversal and have greatly reduced expression of the key testis-determining genes Sry and Sox9. An analysis of histone acetylation at the Sry promoter in mutant gonads at 11.5 dpc shows a reduction in levels of the positive histone mark H3K27Ac. Our data suggest a role for CBP/p300 in testis determination mediated by control of histone acetylation at the Sry locus and reveal a novel element in the epigenetic control of Sry and mammalian sex determination. They also suggest possible novel causes of human disorders of sex development (DSD).


Asunto(s)
Proteína de Unión a CREB/deficiencia , Trastornos del Desarrollo Sexual/metabolismo , Proteína p300 Asociada a E1A/deficiencia , Histonas/metabolismo , Procesos de Determinación del Sexo/fisiología , Proteína de la Región Y Determinante del Sexo/genética , Testículo/embriología , Acetilación , Animales , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Trastornos del Desarrollo Sexual/genética , Proteína p300 Asociada a E1A/genética , Proteína p300 Asociada a E1A/metabolismo , Femenino , Masculino , Ratones , Ovario/embriología , Ovario/metabolismo , Regiones Promotoras Genéticas , Proteína de la Región Y Determinante del Sexo/metabolismo , Testículo/metabolismo
6.
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
7.
Mamm Genome ; 30(1-2): 1-4, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30600355

RESUMEN

Genome editing is revolutionising our ability to modify genomes with exquisite precision for medical and agricultural applications, and in basic research. The first International Summit on Human Genome Editing, organised jointly by the US National Academies of Sciences and Medicine, the Chinese Academy of Sciences and the UK Royal Society, was held in Washington DC at the end of 2015. Its aim was to explore scientific, legal and ethical perspectives on the prospective use of human genome editing as a therapeutic intervention in disease (so-called somatic genome editing) and as a possible intervention in human reproduction (so-called germ-line genome editing). Following that Summit, the Organising Committee had, in a press release, come to the conclusion that: "It would be irresponsible to proceed with any clinical use of germ line editing unless and until (i) the relevant safety and efficacy issues have been resolved, based on appropriate understanding and balancing of risks, potential benefits and alternatives, and (ii) there is broad societal consensus about the appropriateness of the proposed application" ( http://www8.nationalacademies.org/onpinews/newsitem.aspx?RecordID=12032015a ). A report from the US National Academies subsequently reiterated and developed the approach.


Asunto(s)
Edición Génica , Genoma Humano , Animales , Humanos , Investigación
8.
Br Med Bull ; 127(1): 23-31, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30202929

RESUMEN

Introduction or background: Genome editing facilitates alterations to DNA, large or subtle, in a precise fashion. In its most popular form it uses the programmable endonuclease system, CRISPR/Cas9. Edits can be made to any genome, including the human genome. This raises the possibility of genome editing in human embryos in both a research and reproductive context. Sources of data: All reports of genome editing in human embryos are included here, along with key papers examining the science and ethics of human genome editing. Areas of agreement: As a basic research tool, genome editing promises to accelerate our understanding of genome biology. It also shows great promise as a means of combatting disease through so-called somatic genome editing. Areas of controversy: Genome editing could be used to prevent human disease transmission in a reproductive context. Such germ line interventions are opposed by some, for a number of reasons. Some of these reasons are discussed and a comparison is made with preimplantation genetic diagnosis (PGD). Growing points: It is important that scientists, clinicians, bioethicists and other stakeholders engage widely with all those with an interest in genome editing. Areas timely for developing research: In addition to offering new insights into human biology, basic (fundamental) research will deliver expertise allowing ever more precise and controllable genome editing methodologies and allied technologies. A range of clear and accessible ethical frameworks must be developed and scrutinized as part of a wider societal debate about possible applications of genome editing. In the UK, human reproductive genome editing can only take place if a change to primary legislation occurs. Inclusive discussions and assessments, involving difficult scientific and ethical concepts, must form part of any democratic decision.


Asunto(s)
Investigaciones con Embriones/ética , Edición Génica/ética , Medicina Reproductiva/ética , Sistemas CRISPR-Cas , Desarrollo Embrionario/genética , Humanos
9.
Mamm Genome ; 28(7-8): 388-393, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28653171

RESUMEN

Genome editing is facilitating the manipulation of genomes on an unprecedented scale. It promises to revolutionize our ability to study gene function and generate models of human genetic disease in a range of organisms, most notably in mammals such as the mouse. Is this new technology likely to be disruptive to our research practices in any way? Will it alter the ways in which we implement the ethical imperatives of the 3Rs? In short, what ethical questions are raised by genome editing of mammals in a biomedical research context?


Asunto(s)
Ética en Investigación , Edición Génica/ética , Genoma , Mamíferos/genética , Animales , Ingeniería Genética/ética , Ingeniería Genética/métodos , Humanos , Ratones
10.
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
11.
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
12.
Dev Biol ; 373(2): 267-80, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23195221

RESUMEN

During lung development, proper epithelial cell arrangements are critical for the formation of an arborized network of tubes. Each tube requires a lumen, the diameter of which must be tightly regulated to enable optimal lung function. Lung branching and lumen morphogenesis require close epithelial cell-cell contacts that are maintained as a result of adherens junctions, tight junctions and by intact apical-basal (A/B) polarity. However, the molecular mechanisms that maintain epithelial cohesion and lumen diameter in the mammalian lung are unknown. Here we show that Scribble, a protein implicated in planar cell polarity (PCP) signalling, is necessary for normal lung morphogenesis. Lungs of the Scrib mouse mutant Circletail (Crc) are abnormally shaped with fewer airways, and these airways often lack a visible, 'open' lumen. Mechanistically we show that Scrib genetically interacts with the core PCP gene Vangl2 in the developing lung and that the distribution of PCP pathway proteins and Rho mediated cytoskeletal modification is perturbed in Scrib(Crc/Crc) lungs. However A/B polarity, which is disrupted in Drosophila Scrib mutants, is largely unaffected. Notably, we find that Scrib mediates functions not attributed to other PCP proteins in the lung. Specifically, Scrib localises to both adherens and tight junctions of lung epithelia and knockdown of Scrib in lung explants and organotypic cultures leads to reduced cohesion of lung epithelial cells. Live imaging of Scrib knockdown lungs shows that Scrib does not affect bud bifurcation, as previously shown for the PCP protein Celsr1, but is required to maintain epithelial cohesion. To understand the mechanism leading to reduced cell-cell association, we show that Scrib associates with ß-catenin in embryonic lung and the sub-cellular distribution of adherens and tight junction proteins is perturbed in mutant lung epithelia. Our data reveal that Scrib is required for normal lung epithelial organisation and lumen morphogenesis by maintaining cell-cell contacts. Thus we reveal novel and important roles for Scrib in lung development operating via the PCP pathway, and in regulating junctional complexes and cell cohesion.


Asunto(s)
Comunicación Celular , Células Epiteliales/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Pulmón/citología , Pulmón/embriología , Mamíferos/embriología , Morfogénesis , Uniones Adherentes/efectos de los fármacos , Uniones Adherentes/metabolismo , Animales , Comunicación Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Polaridad Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Epitelio/efectos de los fármacos , Epitelio/embriología , Epitelio/metabolismo , Técnicas de Silenciamiento del Gen , Imagenología Tridimensional , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Modelos Biológicos , Morfogénesis/efectos de los fármacos , Morfolinos/farmacología , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-2/metabolismo , beta Catenina/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
13.
Development ; 138(6): 1131-42, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21307093

RESUMEN

In mammals, left-right (L-R) asymmetry is established by posteriorly oriented cilia driving a leftwards laminar flow in the embryonic node, thereby activating asymmetric gene expression. The two-cilia hypothesis argues that immotile cilia detect and respond to this flow through a Pkd2-mediated mechanism; a putative sensory partner protein has, however, remained unidentified. We have identified the Pkd1-related locus Pkd1l1 as a crucial component of L-R patterning in mouse. Systematic comparison of Pkd1l1 and Pkd2 point mutants reveals strong phenocopying, evidenced by both morphological and molecular markers of sidedness; both mutants fail to activate asymmetric gene expression at the node or in the lateral plate and exhibit right isomerism of the lungs. Node and cilia morphology were normal in mutants and cilia demonstrated typical motility, consistent with Pkd1l1 and Pkd2 activity downstream of nodal flow. Cell biological analysis reveals that Pkd1l1 and Pkd2 localise to the cilium and biochemical experiments demonstrate that they can physically interact. Together with co-expression in the node, these data argue that Pkd1l1 is the elusive Pkd2 binding partner required for L-R patterning and support the two-cilia hypothesis.


Asunto(s)
Tipificación del Cuerpo/genética , Proteínas de la Membrana/fisiología , Canales Catiónicos TRPP/metabolismo , Secuencia de Aminoácidos , Animales , Tipificación del Cuerpo/fisiología , Células Cultivadas , Cilios/genética , Cilios/metabolismo , Cilios/fisiología , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple/genética , Polimorfismo de Nucleótido Simple/fisiología , Unión Proteica/genética , Unión Proteica/fisiología , Homología de Secuencia de Aminoácido , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/fisiología
14.
Endocrine ; 84(2): 345-349, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38400880

RESUMEN

PURPOSE: Disorders/differences of sex development (DSD) result from variants in many different human genes but, frequently, have no detectable molecular cause. METHODS: Detailed clinical and genetic phenotyping was conducted on a family with three children. A Sec31a animal model and functional studies were used to investigate the significance of the findings. RESULTS: By trio whole-exome DNA sequencing we detected a heterozygous de novo nonsense SEC31A variant, in three children of healthy non-consanguineous parents. The children had different combinations of disorders that included complete gonadal dysgenesis and multiple pituitary hormone deficiency. SEC31A encodes a component of the COPII coat protein complex, necessary for intracellular anterograde vesicle-mediated transport between the endoplasmic reticulum (ER) and Golgi. CRISPR-Cas9 targeted knockout of the orthologous Sec31a gene region resulted in early embryonic lethality in homozygous mice. mRNA expression of ER-stress genes ATF4 and CHOP was increased in the children, suggesting defective protein transport. The pLI score of the gene, from gnomAD data, is 0.02. CONCLUSIONS: SEC31A might underlie a previously unrecognised clinical syndrome comprising gonadal dysgenesis, multiple pituitary hormone deficiencies, dysmorphic features and developmental delay. However, a variant that remains undetected, in a different gene, may alternatively be causal in this family.


Asunto(s)
Disgenesia Gonadal , Hipopituitarismo , Animales , Niño , Preescolar , Femenino , Humanos , Masculino , Ratones , Disgenesia Gonadal/genética , Hipopituitarismo/genética , Hipopituitarismo/metabolismo , Ratones Noqueados , Linaje , Hormonas Hipofisarias/deficiencia , Hormonas Hipofisarias/genética , Proteínas de Transporte Vesicular/genética
15.
Am J Hum Genet ; 87(6): 898-904, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-21129722

RESUMEN

Investigations of humans with disorders of sex development (DSDs) resulted in the discovery of many of the now-known mammalian sex-determining genes, including SRY, RSPO1, SOX9, NR5A1, WT1, NR0B1, and WNT4. Here, the locus for an autosomal sex-determining gene was mapped via linkage analysis in two families with 46,XY DSD to the long arm of chromosome 5 with a combined, multipoint parametric LOD score of 6.21. A splice-acceptor mutation (c.634-8T>A) in MAP3K1 segregated with the phenotype in the first family and disrupted RNA splicing. Mutations were demonstrated in the second family (p.Gly616Arg) and in two of 11 sporadic cases (p.Leu189Pro, p.Leu189Arg)-18% prevalence in this cohort of sporadic cases. In cultured primary lymphoblastoid cells from family 1 and the two sporadic cases, these mutations altered the phosphorylation of the downstream targets, p38 and ERK1/2, and enhanced binding of RHOA to the MAP3K1 complex. Map3k1 within the syntenic region was expressed in the embryonic mouse gonad prior to, and after, sex determination. Thus, mutations in MAP3K1 that result in 46,XY DSD with partial or complete gonadal dysgenesis implicate this pathway in normal human sex determination.


Asunto(s)
Trastorno del Desarrollo Sexual 46,XY/genética , Quinasa 1 de Quinasa de Quinasa MAP/genética , Mutación , Transducción de Señal , Testículo/embriología , Secuencia de Aminoácidos , Animales , Femenino , Humanos , Quinasa 1 de Quinasa de Quinasa MAP/química , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Masculino , Linaje , Fosforilación , Homología de Secuencia de Aminoácido
16.
Br Med Bull ; 127(1): 145, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30032261
17.
Nature ; 489(7417): 502, 2012 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-23018955
18.
Hum Mol Genet ; 19(11): 2251-67, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20223754

RESUMEN

The lungs are generated by branching morphogenesis as a result of reciprocal signalling interactions between the epithelium and mesenchyme during development. Mutations that disrupt formation of either the correct number or shape of epithelial branches affect lung function. This, in turn, can lead to congenital abnormalities such as cystadenomatoid malformations, pulmonary hypertension or lung hypoplasia. Defects in lung architecture are also associated with adult lung disease, particularly in cases of idiopathic lung fibrosis. Identifying the signalling pathways which drive epithelial tube formation will likely shed light on both congenital and adult lung disease. Here we show that mutations in the planar cell polarity (PCP) genes Celsr1 and Vangl2 lead to disrupted lung development and defects in lung architecture. Lungs from Celsr1(Crsh) and Vangl2(Lp) mouse mutants are small and misshapen with fewer branches, and by late gestation exhibit thickened interstitial mesenchyme and defective saccular formation. We observe a recapitulation of these branching defects following inhibition of Rho kinase, an important downstream effector of the PCP signalling pathway. Moreover, epithelial integrity is disrupted, cytoskeletal remodelling perturbed and mutant endoderm does not branch normally in response to the chemoattractant FGF10. We further show that Celsr1 and Vangl2 proteins are present in restricted spatial domains within lung epithelium. Our data show that the PCP genes Celsr1 and Vangl2 are required for foetal lung development thereby revealing a novel signalling pathway critical for this process that will enhance our understanding of congenital and adult lung diseases and may in future lead to novel therapeutic strategies.


Asunto(s)
Pulmón/embriología , Morfogénesis/genética , Morfogénesis/fisiología , Proteínas del Tejido Nervioso/genética , Receptores Acoplados a Proteínas G/genética , Mucosa Respiratoria/metabolismo , Transducción de Señal/genética , Animales , Polaridad Celular/genética , Polaridad Celular/fisiología , Immunoblotting , Inmunohistoquímica , Ratones , Modelos Biológicos , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/fisiología , Oligonucleótidos/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Mucosa Respiratoria/embriología
19.
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
20.
Eur J Endocrinol ; 186(5): K25-K31, 2022 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-35235537

RESUMEN

Background: The human INHA gene encodes the inhibin subunit alpha protein, which is common to both inhibin A and B. The functional importance of inhibins in male sex development, sexual function, and reproduction remain largely unknown. Objective: We report for the first time two male siblings with homozygous INHAmutations. Methods: The medical files were examined for clinical, biochemical, and imaging data. Genetic analysis was performed using next-generation and Sanger sequencing methods. Results: Two brothers complained of gynecomastia, testicular pain, and had a history of hypospadias. Biochemistry revealed low serum testosterone, high gonadotropin and anti-Mullerian hormone, and very low/undetectable inhibin concentrations, where available. Both patients had azoospermia in the spermiogram. We have identified a homozygous 2 bp deletion (c.208_209delAG, R70Gfs*3) variant, which leads to a truncated INHA protein in both patients, and confirmed heterozygosity in the parents. The external genital development, pubertal onset and progression, reproductive functions, serum gonadotropins, and sex hormones of mother and father, who were heterozygous carriers of the identified mutation, were normal. Conclusion: Homozygosity for INHA mutations causes decreased prenatal and postnatal testosterone production and infertility in males, while the heterozygous female and male carriers of INHA mutations do not have any abnormality in sex development and reproduction.


Asunto(s)
Hipogonadismo , Hipospadias , Inhibinas/genética , Femenino , Humanos , Hipogonadismo/metabolismo , Hipospadias/genética , Hipospadias/metabolismo , Masculino , Mutación/genética , Hermanos , Testículo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA