Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 150(24)2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38078651

RESUMO

To investigate the role of the nuclear receptor NR5A1 in the testis after sex determination, we analyzed mice lacking NR5A1 in Sertoli cells (SCs) from embryonic day (E) 13.5 onwards. Ablation of Nr5a1 impaired the expression of genes characteristic of SC identity (e.g. Sox9 and Amh), caused SC death from E14.5 onwards through a Trp53-independent mechanism related to anoikis, and induced disorganization of the testis cords. Together, these effects caused germ cells to enter meiosis and die. Single-cell RNA-sequencing experiments revealed that NR5A1-deficient SCs changed their molecular identity: some acquired a 'pre-granulosa-like' cell identity, whereas other reverted to a 'supporting progenitor-like' cell identity, most of them being 'intersex' because they expressed both testicular and ovarian genes. Fetal Leydig cells (LCs) did not display significant changes, indicating that SCs are not required beyond E14.5 for their emergence or maintenance. In contrast, adult LCs were absent from postnatal testes. In addition, adult mutant males displayed persistence of Müllerian duct derivatives, decreased anogenital distance and reduced penis length, which could be explained by the loss of AMH and testosterone synthesis due to SC failure.


Assuntos
Anoikis , Células de Sertoli , Animais , Masculino , Camundongos , Anoikis/genética , Morte Celular/genética , Células de Sertoli/metabolismo , Testículo/metabolismo
2.
Development ; 2023 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-38063846

RESUMO

To investigate the role of the nuclear receptor NR5A1 in testis after sex determination, we have analyzed mice lacking NR5A1 in Sertoli cells (SC) from embryonic day (E) 13.5 onwards. Ablation of Nr5a1 impairs the expression of genes characteristic of the SC identity (e.g., Sox9, Amh), causes SC death from E14.5 through a Trp53-independent mechanism related to anoikis, and induces disorganization of the testis cords. Together, these effects cause germ cells to enter meiosis and die. Single-cell RNA-sequencing experiments revealed that NR5A1-deficient SC change their molecular identity: some acquire a "pre-granulosa-like" identity, while other revert to a "supporting progenitor-like" cell identity, most of them being "intersex" because they express both testicular and ovarian genes. Fetal Leydig cells (LC) do not display significant changes, indicating that SC are not required beyond E14.5 for their emergence or maintenance. In contrast, adult LC were absent from the postnatal testes. In addition, adult mutant males display persistence of Müllerian duct derivatives, decreased anogenital distance and reduced penis length, which can be explained by the loss of AMH and testosterone synthesis due to SC failure.

3.
PLoS Genet ; 19(3): e1010656, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36857387

RESUMO

DND1 is essential to maintain germ cell identity. Loss of Dnd1 function results in germ cell differentiation to teratomas in some inbred strains of mice or to somatic fates in zebrafish. Using our knock-in mouse line in which a functional fusion protein between DND1 and GFP is expressed from the endogenous locus (Dnd1GFP), we distinguished two male germ cell (MGC) populations during late gestation cell cycle arrest (G0), consistent with recent reports of heterogeneity among MGCs. Most MGCs express lower levels of DND1-GFP (DND1-GFP-lo), but some MGCs express elevated levels of DND1-GFP (DND1-GFP-hi). A RNA-seq time course confirmed high Dnd1 transcript levels in DND1-GFP-hi cells along with 5-10-fold higher levels for multiple epigenetic regulators. Using antibodies against DND1-GFP for RNA immunoprecipitation (RIP)-sequencing, we identified multiple epigenetic and translational regulators that are binding targets of DND1 during G0 including DNA methyltransferases (Dnmts), histone deacetylases (Hdacs), Tudor domain proteins (Tdrds), actin dependent regulators (Smarcs), and a group of ribosomal and Golgi proteins. These data suggest that in DND1-GFP-hi cells, DND1 hosts coordinating mRNA regulons that consist of functionally related and localized groups of epigenetic enzymes and translational components.


Assuntos
Espermatogônias , Peixe-Zebra , Animais , Feminino , Masculino , Camundongos , Gravidez , Cromatina/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Ligação a RNA/genética , Espermatogônias/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
4.
Metabolomics ; 19(6): 53, 2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-37271779

RESUMO

INTRODUCTION: A decrease in sperm cell count has been observed along the last several decades, especially in the most developed regions of the world. The use of metabolomics to study the composition of the seminal fluid is a promising approach to gain access to the molecular mechanisms underlying this fact. OBJECTIVES: In the present work, we aimed at relating metabolomic profiles of young healthy men to their semen quality parameters obtained from conventional microscopic analysis. METHODS: An untargeted metabolomics approach focusing on low- to mid-polarity compounds was used to analyze a subset of seminal fluid samples from a cohort of over 2700 young healthy men. RESULTS: Our results show that a broad metabolic profiling comprising several families of compounds (including acyl-carnitines, steroids, and other lipids) can contribute to effectively distinguish samples provided by individuals exhibiting low or high absolute sperm counts. CONCLUSION: A number of metabolites involved in sexual development and function, signaling, and energy metabolism were highlighted as being distinctive of samples coming from either group, proving untargeted metabolomics as a promising tool to better understand the pathophysiological processes responsible for male fertility impairment.


Assuntos
Análise do Sêmen , Sêmen , Humanos , Masculino , Sêmen/metabolismo , Metabolômica/métodos , Espermatozoides/metabolismo , Contagem de Espermatozoides
5.
Trends Genet ; 35(5): 346-358, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30902461

RESUMO

Sex determination is the process by which the bipotential gonads develop as either testes or ovaries. With two distinct potential outcomes, the gonadal primordium offers a unique model for the study of cell fate specification and how distinct cell populations diverge from multipotent progenitors. This review focuses on recent advances in our understanding of the genetic programs and epigenetic mechanisms that regulate gonadal sex determination and the regulation of cell fate commitment in the bipotential gonads. We rely primarily on mouse data to illuminate the complex and dynamic genetic programs controlling cell fate decision and sex-specific cell differentiation during gonadal formation and gonadal sex determination.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Gônadas/embriologia , Gônadas/metabolismo , Organogênese/genética , Processos de Determinação Sexual/genética , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Desenvolvimento Embrionário/genética , Feminino , Humanos , Masculino , Ovário , Fenótipo , Testículo
6.
Am J Hum Genet ; 104(2): 331-340, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30686508

RESUMO

Male infertility is a major health concern. Among its different causes, multiple morphological abnormalities of the flagella (MMAF) induces asthenozoospermia and is one of the most severe forms of qualitative sperm defects. Sperm of affected men display short, coiled, absent, and/or irregular flagella. To date, six genes (DNAH1, CFAP43, CFAP44, CFAP69, FSIP2, and WDR66) have been found to be recurrently associated with MMAF, but more than half of the cases analyzed remain unresolved, suggesting that many yet-uncharacterized gene defects account for this phenotype. Here, whole-exome sequencing (WES) was performed on 168 infertile men who had a typical MMAF phenotype. Five unrelated affected individuals carried a homozygous deleterious mutation in ARMC2, a gene not previously linked to the MMAF phenotype. Using the CRISPR-Cas9 technique, we generated homozygous Armc2 mutant mice, which also presented an MMAF phenotype, thus confirming the involvement of ARMC2 in human MMAF. Immunostaining experiments in AMRC2-mutated individuals and mutant mice evidenced the absence of the axonemal central pair complex (CPC) proteins SPAG6 and SPEF2, whereas the other tested axonemal and peri-axonemal components were present, suggesting that ARMC2 is involved in CPC assembly and/or stability. Overall, we showed that bi-allelic mutations in ARMC2 cause male infertility in humans and mice by inducing a typical MMAF phenotype, indicating that this gene is necessary for sperm flagellum structure and assembly.


Assuntos
Alelos , Astenozoospermia/genética , Astenozoospermia/patologia , Proteínas do Citoesqueleto/genética , Flagelos/genética , Mutação , Espermatozoides/anormalidades , Espermatozoides/patologia , Animais , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/deficiência , Humanos , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Masculino , Camundongos , Proteínas dos Microtúbulos/deficiência , Proteínas
7.
FASEB J ; 35(4): e21452, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33749946

RESUMO

Despite the importance of germ cell (GC) differentiation for sexual reproduction, the gene networks underlying their fate remain unclear. Here, we comprehensively characterize the gene expression dynamics during sex determination based on single-cell RNA sequencing of 14 914 XX and XY mouse GCs between embryonic days (E) 9.0 and 16.5. We found that XX and XY GCs diverge transcriptionally as early as E11.5 with upregulation of genes downstream of the bone morphogenic protein (BMP) and nodal/Activin pathways in XY and XX GCs, respectively. We also identified a sex-specific upregulation of genes associated with negative regulation of mRNA processing and an increase in intron retention consistent with a reduction in mRNA splicing in XY testicular GCs by E13.5. Using computational gene regulation network inference analysis, we identified sex-specific, sequential waves of putative key regulator genes during GC differentiation and revealed that the meiotic genes are regulated by positive and negative master modules acting in an antagonistic fashion. Finally, we found that rare adrenal GCs enter meiosis similarly to ovarian GCs but display altered expression of master genes controlling the female and male genetic programs, indicating that the somatic environment is important for GC function. Our data are available on a web platform and provide a molecular roadmap of GC sex determination at single-cell resolution, which will serve as a valuable resource for future studies of gonad development, function, and disease.


Assuntos
Perfilação da Expressão Gênica/métodos , Processos de Determinação Sexual , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas , Masculino , Camundongos , Camundongos Transgênicos , Análise de Célula Única , Fatores de Tempo , Cromossomo X , Cromossomo Y
8.
Hum Reprod ; 36(10): 2638-2648, 2021 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-34486673

RESUMO

STUDY QUESTION: Do selective serotonin reuptake inhibitor (SSRI) antidepressants affect the function of human sperm? SUMMARY ANSWER: The SSRI antidepressant Sertraline (e.g. Zoloft) inhibits the sperm-specific Ca2+ channel CatSper and affects human sperm function in vitro. WHAT IS KNOWN ALREADY: In human sperm, CatSper translates changes of the chemical microenvironment into changes of the intracellular Ca2+ concentration ([Ca2+]i) and swimming behavior. CatSper is promiscuously activated by oviductal ligands, but also by synthetic chemicals that might disturb the fertilization process. It is well known that SSRIs have off-target actions on Ca2+, Na+ and K+ channels in somatic cells. Whether SSRIs affect the activity of CatSper is, however, unknown. STUDY DESIGN, SIZE, DURATION: We studied the action of the seven drugs belonging to the most commonly prescribed class of antidepressants, SSRIs, on resting [Ca2+]i and Ca2+ influx via CatSper in human sperm. The SSRI Sertraline was selected for in-depth analysis of its action on steroid-, prostaglandin-, pH- and voltage-activation of human CatSper. Moreover, the action of Sertraline on sperm acrosomal exocytosis and penetration into viscous media was evaluated. PARTICIPANTS/MATERIALS, SETTING, METHODS: The activity of CatSper was investigated in sperm of healthy volunteers, using kinetic Ca2+ fluorimetry and patch-clamp recordings. Acrosomal exocytosis was investigated using Pisum sativum agglutinin and image cytometry. Sperm penetration in viscous media was evaluated using the Kremer test. MAIN RESULTS AND THE ROLE OF CHANCE: Several SSRIs affected [Ca2+]i and attenuated ligand-induced Ca2+ influx via CatSper. In particular, the SSRI Sertraline almost completely suppressed Ca2+ influx via CatSper. Remarkably, the drug was about four-fold more potent to suppress prostaglandin- versus steroid-induced Ca2+ influx. Sertraline also suppressed alkaline- and voltage-activation of CatSper, indicating that the drug directly inhibits the channel. Finally, Sertraline impaired ligand-induced acrosome reaction and sperm penetration into viscous media. LIMITATIONS, REASONS FOR CAUTION: This is an in vitro study. Future studies have to assess the physiological relevance in vivo. WIDER IMPLICATIONS OF THE FINDINGS: The off-target action of Sertraline on CatSper in human sperm might impair the fertilization process. In a research setting, Sertraline may be used to selectively inhibit prostaglandin-induced Ca2+ influx. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the Swiss Centre for Applied Human Toxicology (SCAHT), the Département de l'Instruction Publique of the State of Geneva, the German Research Foundation (CRU326), the Interdisciplinary Center for Clinical Research, Münster (IZKF; Str/014/21), the Innovation Fund Denmark (grant numbers 14-2013-4) and the EDMaRC research grant from the Kirsten and Freddy Johansen's Foundation. The authors declare that no conflict of interest could be perceived as prejudicing the impartiality of the research reported. TRIAL REGISTRATION NUMBER: NA.


Assuntos
Cálcio , Sertralina , Antidepressivos/metabolismo , Antidepressivos/farmacologia , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Sinalização do Cálcio , Humanos , Masculino , Progesterona/farmacologia , Sertralina/metabolismo , Sertralina/farmacologia , Motilidade dos Espermatozoides , Espermatozoides/metabolismo
9.
FASEB J ; 34(8): 10373-10386, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32557858

RESUMO

Male development, fertility, and lifelong health are all androgen-dependent. Approximately 95% of circulating testosterone is synthesized by the testis and the final step in this canonical pathway is controlled by the activity of the hydroxysteroid-dehydrogenase-17-beta-3 (HSD17B3). To determine the role of HSD17B3 in testosterone production and androgenization during male development and function we have characterized a mouse model lacking HSD17B3. The data reveal that developmental masculinization and fertility are normal in mutant males. Ablation of HSD17B3 inhibits hyperstimulation of testosterone production by hCG, although basal testosterone levels are maintained despite the absence of HSD17B3. Reintroduction of HSD17B3 via gene-delivery to Sertoli cells in adulthood partially rescues the adult phenotype, showing that, as in development, different cell-types in the testis are able to work together to produce testosterone. Together, these data show that HS17B3 acts as a rate-limiting-step for the maximum level of testosterone production by the testis but does not control basal testosterone production. Measurement of other enzymes able to convert androstenedione to testosterone identifies HSD17B12 as a candidate enzyme capable of driving basal testosterone production in the testis. Together, these findings expand our understanding of testosterone production in males.


Assuntos
17-Hidroxiesteroide Desidrogenases/metabolismo , Células de Sertoli/metabolismo , Testículo/metabolismo , Testosterona/metabolismo , Androgênios/metabolismo , Animais , Feminino , Fertilidade/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
10.
Proc Natl Acad Sci U S A ; 115(21): 5474-5479, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29735715

RESUMO

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.


Assuntos
Transtornos do Desenvolvimento Sexual/genética , Diferenciação Sexual , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/fisiologia , Proteínas Wnt/antagonistas & inibidores , beta Catenina/antagonistas & inibidores , Adolescente , Adulto , Animais , Células Cultivadas , Transtornos do Desenvolvimento Sexual/patologia , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/metabolismo , Gônadas/patologia , Humanos , Masculino , Camundongos , Mutação de Sentido Incorreto , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Testículo/metabolismo , Testículo/patologia , Trombospondinas/genética , Trombospondinas/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Adulto Jovem , Peixe-Zebra , beta Catenina/genética , beta Catenina/metabolismo
11.
Bioinformatics ; 35(17): 3133-3139, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30668675

RESUMO

MOTIVATION: Recent advances in transcriptomics have enabled unprecedented insight into gene expression analysis at a single-cell resolution. While it is anticipated that the number of publications based on such technologies will increase in the next decade, there is currently no public resource to centralize and enable scientists to explore single-cell datasets published in the field of reproductive biology. RESULTS: Here, we present a major update of the ReproGenomics Viewer, a cross-species and cross-technology web-based resource of manually-curated sequencing datasets related to reproduction. The redesign of the ReproGenomics Viewer's architecture is accompanied by significant growth of the database content including several landmark single-cell RNA-sequencing datasets. The implementation of additional tools enables users to visualize and browse the complex, high-dimensional data now being generated in the reproductive field. AVAILABILITY AND IMPLEMENTATION: The ReproGenomics Viewer resource is freely accessible at http://rgv.genouest.org. The website is implemented in Python, JavaScript and MongoDB, and is compatible with all major browsers. Source codes can be downloaded from https://github.com/fchalmel/RGV. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Software , Biologia Computacional , Bases de Dados Factuais , Genômica , Análise de Sequência de RNA
12.
Genet Med ; 22(1): 150-159, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31337883

RESUMO

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.


Assuntos
Disgenesia Gonadal 46 XY/genética , Mutação de Sentido Incorreto , RNA Helicases/genética , Análise de Sequência de DNA/métodos , Testículo/crescimento & desenvolvimento , Adolescente , Animais , Pré-Escolar , Feminino , Predisposição Genética para Doença , Heterozigoto , Humanos , Recém-Nascido , Masculino , Camundongos , Mutagênese Sítio-Dirigida , Taxa de Mutação , Domínios Proteicos , RNA Helicases/química , Testículo/metabolismo , Adulto Jovem
13.
Int J Mol Sci ; 21(21)2020 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-33182400

RESUMO

Androgen insensitivity syndrome (AIS), manifesting incomplete virilization in 46,XY individuals, is caused mostly by androgen receptor (AR) gene mutations. Therefore, a search for AR mutations is a routine approach in AIS diagnosis. However, some AIS patients lack AR mutations, which complicates the diagnosis. Here, we describe a patient suffering from partial androgen insensitivity syndrome (PAIS) and lacking AR mutations. The whole exome sequencing of the patient and his family members identified a heterozygous FKBP4 gene mutation, c.956T>C (p.Leu319Pro), inherited from the mother. The gene encodes FKBP prolyl isomerase 4, a positive regulator of the AR signaling pathway. This is the first report describing a FKBP4 gene mutation in association with a human disorder of sexual development (DSD). Importantly, the dysfunction of a homologous gene was previously reported in mice, resulting in a phenotype corresponding to PAIS. Moreover, the Leu319Pro amino acid substitution occurred in a highly conserved position of the FKBP4 region, responsible for interaction with other proteins that are crucial for the AR functional heterocomplex formation and therefore the substitution is predicted to cause the disease. We proposed the FKBP4 gene as a candidate AIS gene and suggest screening that gene for the molecular diagnosis of AIS patients lacking AR gene mutations.


Assuntos
Síndrome de Resistência a Andrógenos/genética , Receptores Androgênicos/genética , Transdução de Sinais/genética , Proteínas de Ligação a Tacrolimo/genética , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Criança , Exoma/genética , Humanos , Masculino , Mutação/genética , Desenvolvimento Sexual/genética
14.
Biol Reprod ; 100(1): 253-267, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30084987

RESUMO

Controlled changes in mitochondrial biogenesis and morphology are required for cell survival and homeostasis, but the molecular mechanisms are largely unknown. Here, male and female prepubertal mice (P21) with insulin and IGF1 receptors deletions in steroidogenic tissues (Insr/Igf1r-DKO) were used to investigate transcription of the key regulators of mitochondrial biogenesis (Ppargc1a, Ppargc1b, Pparg, Nrf1, Tfam) and architecture in Leydig cells, ovaries, and adrenals. Results showed that the expression of PGC1, a master regulator of mitochondrial biogenesis and integrator of environmental signals, and its downstream target Tfam, significantly decreased in androgen-producing Leydig cells. This is followed by reduction of Mtnd1, a mitochondrial DNA encoded transcript whose core subunit belongs to the minimal assembly required for catalysis. The same markers remained unchanged in ovaries. In contrast, in adrenals, the pattern of transcripts for mitochondrial biogenesis markers was the same in both sexes, but opposite from that observed in Leydig cells. The level of transcripts for markers of mitochondrial architecture (Mfn1, Mfn2) significantly increased in Leydig cells from Insr/Igf1r-DKO, but not in ovaries. This was followed by mitochondrial morphology disturbance, suggesting that the mitochondrial phase of steroidogenesis could be affected. Indeed, basal and pregnenolone stimulated progesterone productions in the mitochondria of Leydig cells from Insr/Igf1r-DKO decreased more than androgen production, and were barely detectable. Our results are the first to show that INSR/IGF1R are important for mitochondrial biogenesis in gonadal steroidogenic cells of prepubertal males, but not females and they serve as important regulators of mitochondrial architecture and biogenesis markers in Leydig cells.


Assuntos
Fator de Crescimento Insulin-Like I/farmacologia , Insulina/farmacologia , Células Intersticiais do Testículo/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Biogênese de Organelas , Testículo/efeitos dos fármacos , Animais , Biomarcadores/metabolismo , Células Cultivadas , Feminino , Hormônios Esteroides Gonadais/metabolismo , Células Intersticiais do Testículo/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Mitocôndrias/fisiologia , Ovário/efeitos dos fármacos , Ovário/metabolismo , Maturidade Sexual/efeitos dos fármacos , Maturidade Sexual/fisiologia , Transdução de Sinais/efeitos dos fármacos , Testículo/citologia , Testículo/metabolismo
15.
FASEB J ; 32(6): 3321-3335, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29401624

RESUMO

The insulin family of growth factors (insulin, IGF1, and IGF2) are critical in sex determination, adrenal differentiation, and testicular function. Notably, the IGF system has been reported to mediate the proliferation of steroidogenic cells. However, the precise role and contribution of the membrane receptors mediating those effects, namely, insulin receptor (INSR) and type-I insulin-like growth factor receptor (IGF1R), have not, to our knowledge, been investigated. We show here that specific deletion of both Insr and Igf1r in steroidogenic cells in mice leads to severe alterations of adrenocortical and testicular development. Double-mutant mice display drastic size reduction of both adrenocortex and testes, with impaired corticosterone, testosterone, and sperm production. Detailed developmental analysis of the testes revealed that fetal Leydig cell (LC) function is normal, but there is a failure of adult LC maturation and steroidogenic function associated with accumulation of progenitor LCs (PLCs). Cell-lineage tracing revealed PLC enrichment is secondary to Insr and Igf1r deletion in differentiated adult LCs, suggesting a feedback mechanism between cells at different steps of differentiation. Taken together, these data reveal the cell-autonomous and nonautonomous roles of the IGF system for proper development and maintenance of steroidogenic lineages.-Neirijnck, Y., Calvel, P., Kilcoyne, K. R., Kühne, F., Stévant, I., Griffeth, R. J., Pitetti, J.-L., Andric, S. A., Hu, M.-C., Pralong, F., Smith, L. B., Nef, S. Insulin and IGF1 receptors are essential for the development and steroidogenic function of adult Leydig cells.


Assuntos
Diferenciação Celular , Células Intersticiais do Testículo/metabolismo , Receptor de Insulina/metabolismo , Receptores de Somatomedina/metabolismo , Células-Tronco/metabolismo , Córtex Suprarrenal/citologia , Córtex Suprarrenal/metabolismo , Animais , Corticosterona/genética , Corticosterona/metabolismo , Células Intersticiais do Testículo/citologia , Masculino , Camundongos , Camundongos Knockout , Receptor de Insulina/genética , Receptores de Somatomedina/genética , Células-Tronco/citologia , Testosterona/genética , Testosterona/metabolismo
16.
Int J Mol Sci ; 20(18)2019 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-31505893

RESUMO

Persistent research over the past few decades has clearly established that the insulin-like family of growth factors, which is composed of insulin and insulin-like growth factors 1 (IGF1) and 2 (IGF2), plays essential roles in sexual development and reproduction of both males and females. Within the male and female reproductive organs, ligands of the family act in an autocrine/paracrine manner, in order to guide different aspects of gonadogenesis, sex determination, sex-specific development or reproductive performance. Although our knowledge has greatly improved over the last years, there are still several facets that remain to be deciphered. In this review, we first briefly outline the principles of sexual development and insulin/IGF signaling, and then present our current knowledge, both in rodents and humans, about the involvement of insulin/IGFs in sexual development and reproductive functions. We conclude by highlighting some interesting remarks and delineating certain unanswered questions that need to be addressed in future studies.


Assuntos
Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Insulina/metabolismo , Reprodução/fisiologia , Diferenciação Sexual/fisiologia , Transdução de Sinais/fisiologia , Animais , Feminino , Humanos , Masculino
17.
Hum Mol Genet ; 25(5): 878-91, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26721930

RESUMO

In mammals, sperm-oocyte fusion initiates Ca(2+) oscillations leading to a series of events called oocyte activation, which is the first stage of embryo development. Ca(2+) signaling is elicited by the delivery of an oocyte-activating factor by the sperm. A sperm-specific phospholipase C (PLCZ1) has emerged as the likely candidate to induce oocyte activation. Recently, PAWP, a sperm-born tryptophan domain-binding protein coded by WBP2NL, was proposed to serve the same purpose. Here, we studied two infertile brothers exhibiting normal sperm morphology but complete fertilization failure after intracytoplasmic sperm injection. Whole exomic sequencing evidenced a missense homozygous mutation in PLCZ1, c.1465A>T; p.Ile489Phe, converting Ile 489 into Phe. We showed the mutation is deleterious, leading to the absence of the protein in sperm, mislocalization of the protein when injected in mouse GV and MII oocytes, highly abnormal Ca(2+) transients and early embryonic arrest. Altogether these alterations are consistent with our patients' sperm inability to induce oocyte activation and initiate embryo development. In contrast, no deleterious variants were identified in WBP2NL and PAWP presented normal expression and localization. Overall we demonstrate in humans, the absence of PLCZ1 alone is sufficient to prevent oocyte activation irrespective of the presence of PAWP. Additionally, it is the first mutation located in the C2 domain of PLCZ1, a domain involved in targeting proteins to cell membranes. This opens the door to structure-function studies to identify the conserved amino acids of the C2 domain that regulate the targeting of PLCZ1 and its selectivity for its lipid substrate(s).


Assuntos
Proteínas de Transporte/genética , Infertilidade Masculina/genética , Mutação , Fosfoinositídeo Fosfolipase C/genética , Proteínas de Plasma Seminal/genética , Interações Espermatozoide-Óvulo/genética , Espermatozoides/metabolismo , Adulto , Sequência de Aminoácidos , Animais , Sequência de Bases , Sinalização do Cálcio , Proteínas de Transporte/metabolismo , Perda do Embrião , Feminino , Regulação da Expressão Gênica , Homozigoto , Humanos , Técnicas de Maturação in Vitro de Oócitos , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Masculino , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Oócitos/citologia , Oócitos/metabolismo , Fosfoinositídeo Fosfolipase C/deficiência , Transporte Proteico , Proteínas de Plasma Seminal/metabolismo , Alinhamento de Sequência , Irmãos , Motilidade dos Espermatozoides , Espermatozoides/patologia
18.
J Biol Chem ; 291(32): 16448-61, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27317664

RESUMO

Selective transport of pyruvate across the inner mitochondrial membrane by the mitochondrial pyruvate carrier (MPC) is a fundamental step that couples cytosolic and mitochondrial metabolism. The recent molecular identification of the MPC complex has revealed two interacting subunits, MPC1 and MPC2. Although in yeast, an additional subunit, MPC3, can functionally replace MPC2, no alternative MPC subunits have been described in higher eukaryotes. Here, we report for the first time the existence of a novel MPC subunit termed MPC1-like (MPC1L), which is present uniquely in placental mammals. MPC1L shares high sequence, structural, and topological homology with MPC1. In addition, we provide several lines of evidence to show that MPC1L is functionally equivalent to MPC1: 1) when co-expressed with MPC2, it rescues pyruvate import in a MPC-deleted yeast strain; 2) in mammalian cells, it can associate with MPC2 to form a functional carrier as assessed by bioluminescence resonance energy transfer; 3) in MPC1 depleted mouse embryonic fibroblasts, MPC1L rescues the loss of pyruvate-driven respiration and stabilizes MPC2 expression; and 4) MPC1- and MPC1L-mediated pyruvate imports show similar efficiency. However, we show that MPC1L has a highly specific expression pattern and is localized almost exclusively in testis and more specifically in postmeiotic spermatids and sperm cells. This is in marked contrast to MPC1/MPC2, which are ubiquitously expressed throughout the organism. To date, the biological importance of this alternative MPC complex during spermatogenesis in placental mammals remains unknown. Nevertheless, these findings open up new avenues for investigating the structure-function relationship within the MPC complex.


Assuntos
Proteínas de Transporte de Ânions/biossíntese , Regulação da Expressão Gênica/fisiologia , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Espermátides/metabolismo , Espermatogênese/fisiologia , Testículo/metabolismo , Animais , Proteínas de Transporte de Ânions/genética , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas Mitocondriais/biossíntese , Proteínas Mitocondriais/genética , Transportadores de Ácidos Monocarboxílicos , Espermátides/citologia , Testículo/citologia
19.
Development ; 141(10): 2139-49, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24803659

RESUMO

Sertoli cells (SCs) regulate testicular fate in the differentiating gonad and are the main regulators of spermatogenesis in the adult testis; however, their role during the intervening period of testis development, in particular during adult Leydig cell (ALC) differentiation and function, remains largely unknown. To examine SC function during fetal and prepubertal development we generated two transgenic mouse models that permit controlled, cell-specific ablation of SCs in pre- and postnatal life. Results show that SCs are required: (1) to maintain the differentiated phenotype of peritubular myoid cells (PTMCs) in prepubertal life; (2) to maintain the ALC progenitor population in the postnatal testis; and (3) for development of normal ALC numbers. Furthermore, our data show that fetal LCs function independently from SC, germ cell or PTMC support in the prepubertal testis. Together, these findings reveal that SCs remain essential regulators of testis development long after the period of sex determination. These findings have significant implications for our understanding of male reproductive disorders and wider androgen-related conditions affecting male health.


Assuntos
Diferenciação Celular , Células Intersticiais do Testículo/fisiologia , Epitélio Seminífero/citologia , Células de Sertoli/fisiologia , Maturidade Sexual/fisiologia , Testículo/citologia , Testículo/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Masculino , Camundongos , Camundongos Nus , Camundongos Transgênicos , Túbulos Seminíferos/citologia
20.
PLoS Genet ; 10(5): e1004340, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24784881

RESUMO

The Hedgehog (Hh) family of secreted proteins act as morphogens to control embryonic patterning and development in a variety of organ systems. Post-translational covalent attachment of cholesterol and palmitate to Hh proteins are critical for multimerization and long range signaling potency. However, the biological impact of lipid modifications on Hh ligand distribution and signal reception in humans remains unclear. In the present study, we report a unique case of autosomal recessive syndromic 46,XY Disorder of Sex Development (DSD) with testicular dysgenesis and chondrodysplasia resulting from a homozygous G287V missense mutation in the hedgehog acyl-transferase (HHAT) gene. This mutation occurred in the conserved membrane bound O-acyltransferase (MBOAT) domain and experimentally disrupted the ability of HHAT to palmitoylate Hh proteins such as DHH and SHH. Consistent with the patient phenotype, HHAT was found to be expressed in the somatic cells of both XX and XY gonads at the time of sex determination, and Hhat loss of function in mice recapitulates most of the testicular, skeletal, neuronal and growth defects observed in humans. In the developing testis, HHAT is not required for Sertoli cell commitment but plays a role in proper testis cord formation and the differentiation of fetal Leydig cells. Altogether, these results shed new light on the mechanisms of action of Hh proteins. Furthermore, they provide the first clinical evidence of the essential role played by lipid modification of Hh proteins in human testicular organogenesis and embryonic development.


Assuntos
Aciltransferases/genética , Transtorno 46,XY do Desenvolvimento Sexual/genética , Proteínas Hedgehog/metabolismo , Lipoilação/genética , Mutação de Sentido Incorreto , Transdução de Sinais/genética , Aciltransferases/química , Aciltransferases/metabolismo , Sequência de Aminoácidos , Animais , Feminino , Homozigoto , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Linhagem , Homologia de Sequência de Aminoácidos , Testículo/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA