Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell ; 150(4): 673-84, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22901802

RESUMEN

A pharmacologic approach to male contraception remains a longstanding challenge in medicine. Toward this objective, we explored the spermatogenic effects of a selective small-molecule inhibitor (JQ1) of the bromodomain and extraterminal (BET) subfamily of epigenetic reader proteins. Here, we report potent inhibition of the testis-specific member BRDT, which is essential for chromatin remodeling during spermatogenesis. Biochemical and crystallographic studies confirm that occupancy of the BRDT acetyl-lysine binding pocket by JQ1 prevents recognition of acetylated histone H4. Treatment of mice with JQ1 reduced seminiferous tubule area, testis size, and spermatozoa number and motility without affecting hormone levels. Although JQ1-treated males mate normally, inhibitory effects of JQ1 evident at the spermatocyte and round spermatid stages cause a complete and reversible contraceptive effect. These data establish a new contraceptive that can cross the blood:testis boundary and inhibit bromodomain activity during spermatogenesis, providing a lead compound targeting the male germ cell for contraception.


Asunto(s)
Azepinas/farmacología , Anticonceptivos Masculinos/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Triazoles/farmacología , Animales , Azepinas/química , Barrera Hematotesticular , Anticonceptivos Masculinos/química , Femenino , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Modelos Moleculares , Proteínas Nucleares/química , Estructura Terciaria de Proteína , Recuento de Espermatozoides , Motilidad Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Testículo/citología , Testículo/efectos de los fármacos , Triazoles/química
2.
Development ; 145(15)2018 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-29980567

RESUMEN

Throughout the male reproductive lifespan, spermatogonial stem cells (SSCs) produce committed progenitors that proliferate and then remain physically connected in growing clones via short cylindrical intercellular bridges (ICBs). These ICBs, which enlarge in meiotic spermatocytes, have been demonstrated to provide a conduit for postmeiotic haploid spermatids to share sex chromosome-derived gene products. In addition to ICBs, spermatogonia exhibit multiple thin cytoplasmic projections. Here, we have explored the nature of these projections in mice and find that they are dynamic, span considerable distances from their cell body (≥25 µm), either terminate or physically connect multiple adjacent spermatogonia, and allow for sharing of macromolecules. Our results extend the current model that subsets of spermatogonia exist as isolated cells or clones, and support a model in which spermatogonia of similar developmental fates are functionally connected through a shared dynamic cytoplasm mediated by thin cytoplasmic projections.


Asunto(s)
Citoplasma/metabolismo , Mamíferos/metabolismo , Espermatogonias/metabolismo , Animales , Diferenciación Celular , Citoplasma/ultraestructura , Difusión , Proteínas Fluorescentes Verdes/metabolismo , Espacio Intracelular/metabolismo , Sustancias Macromoleculares/metabolismo , Masculino , Meiosis , Ratones Transgénicos , Papio , Ratas , Espermatocitos/citología , Espermatocitos/metabolismo , Espermatogonias/citología , Espermatogonias/ultraestructura
3.
PLoS Genet ; 7(10): e1002320, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22028666

RESUMEN

The transforming growth factor ß (TGFß) superfamily proteins are principle regulators of numerous biological functions. Although recent studies have gained tremendous insights into this growth factor family in female reproduction, the functions of the receptors in vivo remain poorly defined. TGFß type 1 receptor (TGFBR1), also known as activin receptor-like kinase 5, is the major type 1 receptor for TGFß ligands. Tgfbr1 null mice die embryonically, precluding functional characterization of TGFBR1 postnatally. To study TGFBR1-mediated signaling in female reproduction, we generated a mouse model with conditional knockout (cKO) of Tgfbr1 in the female reproductive tract using anti-Müllerian hormone receptor type 2 promoter-driven Cre recombinase. We found that Tgfbr1 cKO females are sterile. However, unlike its role in growth differentiation factor 9 (GDF9) signaling in vitro, TGFBR1 seems to be dispensable for GDF9 signaling in vivo. Strikingly, we discovered that the Tgfbr1 cKO females develop oviductal diverticula, which impair embryo development and transit of embryos to the uterus. Molecular analysis further demonstrated the dysregulation of several cell differentiation and migration genes (e.g., Krt12, Ace2, and MyoR) that are potentially associated with female reproductive tract development. Moreover, defective smooth muscle development was also revealed in the uteri of the Tgfbr1 cKO mice. Thus, TGFBR1 is required for female reproductive tract integrity and function, and disruption of TGFBR1-mediated signaling leads to catastrophic structural and functional consequences in the oviduct and uterus.


Asunto(s)
Desarrollo Embrionario/genética , Factor 9 de Diferenciación de Crecimiento/metabolismo , Músculo Liso/crecimiento & desarrollo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Fenómenos Fisiológicos Reproductivos/genética , Útero/embriología , Animales , Células Cultivadas , Divertículo/genética , Divertículo/patología , Trompas Uterinas/metabolismo , Trompas Uterinas/patología , Femenino , Fertilidad/genética , Factor 9 de Diferenciación de Crecimiento/genética , Células HEK293 , Humanos , Integrasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso/metabolismo , Progesterona/sangre , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Útero/anomalías
4.
PLoS Biol ; 8(8)2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-20808952

RESUMEN

During gametogenesis and pre-implantation development, the mammalian epigenome is reprogrammed to establish pluripotency in the epiblast. Here we show that the histone 3 lysine 4 (H3K4) methyltransferase, MLL2, controls most of the promoter-specific chromatin modification, H3K4me3, during oogenesis and early development. Using conditional knockout mutagenesis and a hypomorph model, we show that Mll2 deficiency in oocytes results in anovulation and oocyte death, with increased transcription of p53, apoptotic factors, and Iap elements. MLL2 is required for (1) bulk H3K4me3 but not H3K4me1, indicating that MLL2 controls most promoters but monomethylation is regulated by a different H3K4 methyltransferase; (2) the global transcriptional silencing that preceeds resumption of meiosis but not for the concomitant nuclear reorganization into the surrounded nucleolus (SN) chromatin configuration; (3) oocyte survival; and (4) normal zygotic genome activation. These results reveal that MLL2 is autonomously required in oocytes for fertility and imply that MLL2 contributes to the epigenetic reprogramming that takes place before fertilization. We propose that once this task has been accomplished, MLL2 is not required until gastrulation and that other methyltransferases are responsible for bulk H3K4me3, thereby revealing an unexpected epigenetic control switch amongst the H3K4 methyltransferases during development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Lisina/metabolismo , Metiltransferasas/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Oocitos/enzimología , Animales , Epigenómica , Femenino , N-Metiltransferasa de Histona-Lisina , Metilación , Metiltransferasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/metabolismo , Oogénesis
5.
Nat Commun ; 12(1): 3386, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099644

RESUMEN

During early pregnancy in the mouse, nidatory estrogen (E2) stimulates endometrial receptivity by activating a network of signaling pathways that is not yet fully characterized. Here, we report that bone morphogenetic proteins (BMPs) control endometrial receptivity via a conserved activin receptor type 2 A (ACVR2A) and SMAD1/5 signaling pathway. Mice were generated to contain single or double conditional deletion of SMAD1/5 and ACVR2A/ACVR2B receptors using progesterone receptor (PR)-cre. Female mice with SMAD1/5 deletion display endometrial defects that result in the development of cystic endometrial glands, a hyperproliferative endometrial epithelium during the window of implantation, and impaired apicobasal transformation that prevents embryo implantation and leads to infertility. Analysis of Acvr2a-PRcre and Acvr2b-PRcre pregnant mice determined that BMP signaling occurs via ACVR2A and that ACVR2B is dispensable during embryo implantation. Therefore, BMPs signal through a conserved endometrial ACVR2A/SMAD1/5 pathway that promotes endometrial receptivity during embryo implantation.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Implantación del Embrión , Infertilidad Femenina/genética , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Animales , Biopsia , Modelos Animales de Enfermedad , Endometrio/metabolismo , Endometrio/patología , Estrógenos/metabolismo , Femenino , Humanos , Ratones , Ratones Noqueados , Embarazo , Transducción de Señal/fisiología , Proteína Smad1/análisis , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/análisis , Proteína Smad5/genética , Proteína Smad5/metabolismo
6.
Reprod Biol Endocrinol ; 8: 69, 2010 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-20565978

RESUMEN

BACKGROUND: Inhibin is a tumor-suppressor and activin antagonist. Inhibin-deficient mice develop gonadal tumors and a cachexia wasting syndrome due to enhanced activin signaling. Because activins signal through SMAD2 and SMAD3 in vitro and loss of SMAD3 attenuates ovarian tumor development in inhibin-deficient females, we sought to determine the role of SMAD2 in the development of ovarian tumors originating from the granulosa cell lineage. METHODS: Using an inhibin alpha null mouse model and a conditional knockout strategy, double conditional knockout mice of Smad2 and inhibin alpha were generated in the current study. The survival rate and development of gonadal tumors and the accompanying cachexia wasting syndrome were monitored. RESULTS: Nearly identical to the controls, the Smad2 and inhibin alpha double knockout mice succumbed to weight loss, aggressive tumor progression, and death. Furthermore, elevated activin levels and activin-induced pathologies in the liver and stomach characteristic of inhibin deficiency were also observed in these mice. Our results indicate that SMAD2 ablation does not protect inhibin-deficient females from the development of ovarian tumors or the cachexia wasting syndrome. CONCLUSIONS: SMAD2 is not required for mediating tumorigenic signals of activin in ovarian tumor development caused by loss of inhibin.


Asunto(s)
Carcinoma/genética , Inhibinas/genética , Neoplasias Ováricas/genética , Proteína Smad2/fisiología , Neoplasias Testiculares/genética , Activinas/sangre , Activinas/metabolismo , Animales , Carcinoma/metabolismo , Carcinoma/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Proteína Smad2/genética , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patología
7.
Mol Reprod Dev ; 76(5): 453-9, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18951373

RESUMEN

Tektins are evolutionarily conserved flagellar (and ciliary) filamentous proteins present in the axoneme and peri-axonemal structures in diverse metazoan species. We have previously shown that tektin 3 (TEKT3) and tektin 4 (TEKT4) are male germ cell-enriched proteins, and that TEKT4 is essential for coordinated and progressive sperm motility in mice. Here we report that male mice null for TEKT3 produce sperm with reduced motility (47.2% motility) and forward progression, and increased flagellar structural bending defects. Male TEKT3-null mice however maintain normal fertility in two different genetic backgrounds tested, in contrast to TEKT4-null mice. Furthermore, male mice null for both TEKT3 and TEKT4 show subfertility on a mixed B6;129 genetic background, significantly different from either single knockouts, suggesting partial nonredundant roles for these two proteins in sperm physiology. Our results suggest that tektins are potential candidate genes for nonsyndromic asthenozoospermia in humans.


Asunto(s)
Fertilidad , Proteínas de Microtúbulos/genética , Proteínas de Microtúbulos/metabolismo , Motilidad Espermática , Espermatozoides/fisiología , Análisis de Varianza , Animales , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Fertilidad/genética , Flagelos/fisiología , Masculino , Ratones , Ratones Noqueados , Espermatozoides/patología
8.
Mol Endocrinol ; 22(10): 2336-52, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18687735

RESUMEN

Dicer is an evolutionarily conserved ribonuclease III that is necessary for microRNA (miRNA) processing and the synthesis of small interfering RNAs from long double-stranded RNA. Although it has been shown that Dicer plays important roles in the mammalian germline and early embryogenesis, the functions of Dicer-dependent pathways in the somatic cells of the female reproductive tract are unknown. Using a transgenic line in which Cre recombinase is driven by the anti-Müllerian hormone receptor type 2 promoter, we conditionally inactivated Dicer1 in the mesenchyme of the developing Müllerian ducts and postnatally in ovarian granulosa cells and mesenchyme-derived cells of the oviducts and uterus. Deletion of Dicer in these cell types results in female sterility and multiple reproductive defects including decreased ovulation rates, compromised oocyte and embryo integrity, prominent bilateral paratubal (oviductal) cysts, and shorter uterine horns. The paratubal cysts act as a reservoir for spermatozoa and oocytes and prevent embryos from transiting the oviductal isthmus and passing the uterotubal junction to enter the uterus for implantation. Deep sequencing of small RNAs in oviduct revealed down-regulation of specific miRNAs in Dicer conditional knockout females compared with wild type. The majority of these differentially expressed miRNAs are predicted to regulate genes important for Müllerian duct differentiation and mesenchyme-derived structures, and several of these putative target genes were significantly up-regulated upon conditional deletion of Dicer1. Thus, our findings reveal diverse and critical roles for Dicer and its miRNA products in the development and function of the female reproductive tract.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Endorribonucleasas/metabolismo , Genitales Femeninos/fisiología , Infertilidad/genética , Animales , ARN Helicasas DEAD-box/genética , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/anatomía & histología , Endorribonucleasas/genética , Estradiol/sangre , Ciclo Estral/fisiología , Femenino , Hormona Folículo Estimulante/sangre , Genitales Femeninos/anatomía & histología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , MicroARNs/genética , MicroARNs/metabolismo , Oocitos/patología , Oviductos/anatomía & histología , Oviductos/patología , Ovulación/fisiología , Embarazo , Regiones Promotoras Genéticas , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Ribonucleasa III , Espermatozoides/citología
9.
Mol Cell Endocrinol ; 294(1-2): 19-28, 2008 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-18657590

RESUMEN

The inhibins are secreted alpha:beta heterodimers of the TGF-beta superfamily that are mainly synthesized in Sertoli cells and granulosa cells, and are critical regulators of testicular and ovarian development and function. Mice homozygous for a targeted deletion of the inhibin alpha subunit gene (Inha(-/-)) develop sex cord-stromal tumors in a gonadotropin-dependent manner. Here, we determine the contribution of LH to gonadal tumorigenesis by generating mice deficient in both inhibins and LH. Inha(-/-)Lhb(-/-) mice have increased survival and delayed tumor progression, and these observations correlate with lower serum FSH and estradiol levels compared to Inha(-/-) controls. Double mutant testicular tumors demonstrate decreased expression of cyclin D2, while double mutant ovarian tumors have elevated expression of p15(INK4b) and trend toward higher levels of p27(Kip1). We conclude that LH is not required for tumor formation in the absence of inhibins but promotes tumor progression, likely through alterations in serum hormone levels and cell cycle regulators.


Asunto(s)
Inhibinas/deficiencia , Hormona Luteinizante/metabolismo , Neoplasias Ováricas/patología , Neoplasias Testiculares/patología , Animales , Caquexia/patología , Progresión de la Enfermedad , Estradiol/sangre , Femenino , Hormona Folículo Estimulante/sangre , Regulación Neoplásica de la Expresión Génica , Inhibinas/metabolismo , Longevidad , Hormona Luteinizante/deficiencia , Masculino , Ratones , Mutación/genética , Especificidad de Órganos , Neoplasias Ováricas/genética , Análisis de Supervivencia , Síndrome , Neoplasias Testiculares/genética , Testosterona/sangre
10.
FASEB J ; 21(4): 1013-25, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17244819

RESUMEN

Sperm flagellar motion is the outcome of a dynamic interplay between the axonemal cytoskeleton, the peri-axonemal accessory structures, and multiple regulatory networks that coordinate to produce flagellar beat and waveform. Tektins are conserved components of the flagellar proteome in evolutionarily diverse species and are believed to play essential roles in the mechanics of sperm motility. Using database mining, we identified multiple new paralogs of previously annotated tektins, including tektin 4 (TEKT4), which shares 77.1% identity with its nearest human homologue. Mouse Tekt4 is a germ cell-enriched gene, most abundantly expressed in haploid round spermatids in the testis, and the protein is localized to the sperm flagella. Male mice lacking TEKT4 on a 129S5/SvEvBrd inbred background are subfertile. Tekt4-null sperm exhibit drastically reduced forward progressive velocity and uncoordinated waveform propagation along the flagellum. In Tekt4-null sperm, flagellar ultrastructure is grossly unaltered as revealed by transmission electron microscopy. However, the ineffective flagellar strokes lead to approximately 10-fold higher consumption of intracellular ATP in Tekt4-null sperm as compared to wild-type, and null spermatozoa rapidly lose progressive motility when incubated for > or = 1.5 h. Our studies demonstrate that TEKT4 is necessary for the proper coordinated beating of the sperm flagellum and male reproductive physiology.


Asunto(s)
Astenozoospermia/genética , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/fisiología , Regulación de la Expresión Génica , Proteínas de Microtúbulos/genética , Proteínas de Microtúbulos/fisiología , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Astenozoospermia/metabolismo , Citoesqueleto/metabolismo , Flagelos/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Espermatozoides/metabolismo
11.
Mol Endocrinol ; 17(10): 2053-69, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12855747

RESUMEN

Inhibins are heterodimeric (alpha:betaA and alpha:betaB) endocrine, paracrine, and autocrine factors of the TGFbeta superfamily that are produced predominantly by ovarian granulosa cells in females and testicular Sertoli cells in males. Control of granulosa and Sertoli cell proliferation is lost in the inhibin alpha (Inhalpha) knockout mouse model, leading to gonadotropin-dependent gonadal tumors of the granulosa/Sertoli cell lineage in both females and males. Castrate Inhalpha knockout mice develop sex steroidogenic tumors of the adrenal cortex. Physiological control of granulosa/Sertoli cell cycle progression depends on p27Kip1 and cyclin D2, which function in the G1-->S phase transition. To study the cell cycle-regulatory factors involved in ovarian, testicular, and adrenal tumor development in vivo, we have bred Inhalpha mutant mice to mice with targeted disruptions of the p27 and cyclin D2 genes. Our previous studies demonstrated that inhibins act cooperatively with p27 to negatively regulate granulosa cell proliferation, as double mutant mice lacking inhibins and p27 develop and succumb to ovarian tumors more rapidly than Inhalpha knockout mice. Here, we report that cyclin D2 antagonizes this inhibition and is key in promoting gonadal growth and tumor development, and tumor development is markedly suppressed in double-mutant mice. We found that double-knockout females lacking cyclin D2 and Inhalpha lived longer than mice lacking inhibins alone; the majority of these double-knockout mice lived longer than 17 wk, as opposed to inhibin alpha single-knockout females with 50% survival at between 12 and 13 wk of age. Moreover, 95% of inhibin alpha knockout males succumb to testicular tumor development by 12 wk of age, whereas double knockouts were protected from early signs of tumor development and had a 50% survival of 40 wk. Interestingly, the results of these studies reflect tissue-specific consequences of loss of these cell cycle regulators. In castrate mice, loss of p27 has little effect on adrenal cortical tumor progression in the absence of inhibins, whereas loss of cyclin D2 prolongs the lifespan of cyclin D2, Inhalpha double knockouts. After gonadectomy, 50% of cyclin D2, Inhalpha double-knockout males live to more than 46 wk of age, 10 wk longer than 50% of littermates lacking only inhibins. Similarly, 50% of female cyclin D2, inhibin alpha double knockouts live to 47 wk of age before succumbing to adrenal tumor development, in contrast to the 50% survival of Inhalpha single-knockout females at between 27 and 28 wk. Thus, identification of genetic modifiers of the Inhalpha knockout tumor phenotype has led us to a better appreciation of how specific components of the cell cycle machinery contribute to tumorigenesis in the ovary, testis, and adrenal gland.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/fisiopatología , Proteínas de Ciclo Celular/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Neoplasias Ováricas/fisiopatología , Neoplasias Testiculares/fisiopatología , Proteínas Supresoras de Tumor/metabolismo , Neoplasias de la Corteza Suprarrenal/patología , Animales , Caquexia/fisiopatología , Castración , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , División Celular , Transformación Celular Neoplásica , Ciclina D2 , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/genética , Ciclinas/biosíntesis , Ciclinas/genética , Progresión de la Enfermedad , Femenino , Hormona Folículo Estimulante/antagonistas & inhibidores , Hormona Folículo Estimulante/sangre , Células de la Granulosa/metabolismo , Células de la Granulosa/patología , Inhibinas/genética , Inhibinas/metabolismo , Masculino , Ratones , Ratones Noqueados , Neoplasias Ováricas/patología , Ovario/metabolismo , Ovario/patología , Tumor de Células de Sertoli/patología , Tumor de Células de Sertoli/fisiopatología , Neoplasias Testiculares/patología , Testículo/metabolismo , Testículo/patología , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética
12.
Mol Endocrinol ; 17(10): 2039-52, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12855748

RESUMEN

Sex steroids control cellular phenotypes by binding to receptor proteins that in turn regulate downstream gene expression. They are important tropic factors in hormonally responsive tissues and have been implicated in the pathogenesis of both benign proliferations and malignancies at some of these sites. Knockout mice lacking inhibins, alpha:beta heterodimeric peptide hormones of the TGFbeta superfamily, develop gonadal tumors that produce sex steroids and depend on pituitary gonadotropin hormones. To better appreciate how sex steroid receptor signaling pathways contribute to the loss of granulosa/Sertoli cell proliferation in the ovary and testis of inhibin alpha (Inhalpha) knockout mice, we are using both pharmacologic and genetic approaches. Roles of androgens in testicular tumor development have been investigated in our previous studies using double-mutant mice lacking inhibins and carrying the null testicular feminization (tfm) mutation of the androgen receptor. Herein, we report that androgens also participate in the development of ovarian tumors, as tumor development is forestalled in mice treated with flutamide, a nonsteroidal inhibitor of androgen actions. Additionally, we generated double-mutant mice lacking estrogen receptor alpha (ERalpha) and Inhalpha or ERbeta and Inhalpha, as well as triple-mutant mice lacking ERalpha, ERbeta, and Inhalpha to determine the effects of individual and combined ER signaling pathways on tumor development. Although estrogens may have proliferative effects during follicle development and are important in specifying the granulosa cell phenotype, ERalpha and ERbeta signaling are not essential for timely granulosa cell tumor development or granulosa cell-like morphological features in ovarian tumors. However, redundant ER signaling through ERalpha and ERbeta in males is critical for testicular tumor formation, as triple-knockout, but not double-knockout, males are protected from early Sertoli cell tumorigenesis and death. Together, these studies indicate important and sexually dimorphic functions of estrogens and androgens in tumor development in this mouse model and indicate, for the first time, overlapping functions of ERalpha and ERbeta in Sertoli cell pathophysiology.


Asunto(s)
Andrógenos/fisiología , Neoplasias Ováricas/metabolismo , Receptores de Estrógenos/fisiología , Neoplasias Testiculares/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Caquexia/fisiopatología , Ciclina D2 , Ciclinas/genética , Ciclinas/metabolismo , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Flutamida/farmacología , Genotipo , Tumor de Células de la Granulosa/metabolismo , Tumor de Células de la Granulosa/patología , Inhibinas/genética , Inhibinas/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes , Neoplasias Ováricas/patología , Ovario/citología , Ovario/efectos de los fármacos , Ovario/metabolismo , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Estrógenos/genética , Tumor de Células de Sertoli/metabolismo , Tumor de Células de Sertoli/patología , Caracteres Sexuales , Transducción de Señal , Neoplasias Testiculares/patología , Testículo/citología , Testículo/efectos de los fármacos
13.
Biol Reprod ; 80(3): 449-57, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19020301

RESUMEN

A conserved feature of germ cell cytokinesis is the formation of stable intercellular bridges between daughter cells. These intercellular bridges are seen in diverse species from Drosophila melanogaster to Homo sapiens and have been shown to have roles in communication of large numbers of germ cells. In testis expressed gene 14 (Tex14) knockout mice, intercellular bridges do not form during spermatogenesis, and male mice are sterile, demonstrating an essential role for intercellular bridges in postnatal spermatogenesis in mammals. Intercellular bridges also form between dividing germ cells in both male and female embryos. However, little is known about the formation or role of the embryonic intercellular bridges in mammals. In females, embryonic intercellular bridges have been proposed to have a role in development of the presumptive oocyte. Herein, we show that TEX14 is an essential component of male and female embryonic intercellular bridges. In addition, we demonstrate that mitotic kinesin-like protein 1 (MKLP1, official symbol KIF23), which we have discovered is a component of intercellular bridges during spermatogenesis, is also a component of male and female embryonic intercellular bridges. Germ cell intercellular bridges are readily identified by KIF23 immunofluorescence between the gonocytes and oogonia of control mice but are absent between germ cells of Tex14-null mice. Furthermore, by electron microscopy, intercellular bridges are present in all control newborn ovaries but are absent in the Tex14 knockout ovaries. Despite the absence of embryonic intercellular bridges in the Tex14-null mice, male mice initiate spermatogenesis, and female mice are fertile. Although fewer oocytes were present in Tex14-null neonatal ovaries, folliculogenesis was still active at 1 yr of age. Thus, while TEX14 and intercellular bridges have an essential role in postnatal spermatogenesis, they are not required in the embryo.


Asunto(s)
Comunicación Celular/fisiología , Fertilidad/fisiología , Células Germinativas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Citocinesis/fisiología , Femenino , Células Germinativas/citología , Cinesinas/metabolismo , Masculino , Ratones , Ratones Noqueados , Oocitos/citología , Oogénesis/fisiología , Ovario/citología , Ovario/embriología , Ovario/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Espermatogénesis/fisiología , Testículo/citología , Testículo/embriología , Testículo/metabolismo , Factores de Transcripción/genética
14.
Proc Natl Acad Sci U S A ; 103(13): 4982-7, 2006 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-16549803

RESUMEN

Cytokinesis in somatic cells concludes with the formation of a midbody, which is abscised to form individual daughter cells. In contrast, germ cell cytokinesis results in a permanent intercellular bridge connecting the daughter cells through a large cytoplasmic channel. During spermatogenesis, proposed roles for the intercellular bridge include germ cell communication, synchronization, and chromosome dosage compensation in haploid cells. Although several essential components of the midbody have recently been identified, essential components of the vertebrate germ cell intercellular bridge have until now not been described. Herein, we show that testis-expressed gene 14 (TEX14) is a novel protein that localizes to germ cell intercellular bridges. In the absence of TEX14, intercellular bridges are not observed by using electron microscopy and other markers. Spermatogenesis in Tex14(-/-) mice progresses through the transit amplification of diploid spermatogonia and the expression of early meiotic markers but halts before the completion of the first meiotic division. Thus, TEX14 is required for intercellular bridges in vertebrate germ cells, and these studies provide evidence that the intercellular bridge is essential for spermatogenesis and fertility.


Asunto(s)
Fertilidad/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Alelos , Animales , Comunicación Celular , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Espermatogénesis , Testículo/citología , Testículo/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
15.
Proc Natl Acad Sci U S A ; 102(16): 5755-60, 2005 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-15824319

RESUMEN

MSY2, a germ-cell-specific member of the Y-box family of DNA-/RNA-binding proteins, is proposed to function as a coactivator of transcription in the nucleus and to stabilize and store maternal and paternal mRNAs in the cytoplasm. In mice lacking Msy2, a normal Mendelian ratio is observed after matings between heterozygotes with equal numbers of phenotypically normal but sterile male and female homozygotes (Msy2-/-). Spermatogenesis is disrupted in postmeiotic null germ cells with many misshapen and multinucleated spermatids, and no spermatozoa are detected in the epididymis. Apoptosis is increased in the testes of homozygotes, and real-time RT-PCR assays reveal large reductions in the mRNA levels of postmeiotic male germ cell mRNAs and smaller reductions of meiotic germ cell transcripts. In females, there is no apparent decrease in either the number of follicles or their morphology in ovaries obtained from 2- and 8-day-old Msy2-/- mice. In contrast, follicle number and progression are reduced in 21-day-old Msy2-/- ovaries. In adult Msy2-/- females, oocyte loss increases, anovulation is observed, and multiple oocyte and follicle defects are seen. Thus, Msy2 represents one of a small number of germ-cell-specific genes whose deletion leads to the disruption of both spermatogenesis and oogenesis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Infertilidad , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Femenino , Marcación de Gen , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Noqueados , Oocitos/citología , Oocitos/fisiología , Oogénesis/fisiología , Ovario/anomalías , Ovario/citología , Ovario/fisiología , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Espermatogénesis/fisiología , Espermatozoides/anomalías , Espermatozoides/citología , Espermatozoides/fisiología , Testículo/citología , Testículo/fisiología , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA