Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Arch Pharm (Weinheim) ; 349(4): 233-41, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26948688

RESUMEN

Two photo-crosslinking biarsenical (CrAsH-EDT2 )-modified probes were synthesized that are expected to be useful tools for tetracysteine-labeled proteins to facilitate the co-affinity purification of their DNA binding sequences and interacting proteins. In addition, improvements for the synthesis of CrAsH-EDT2 and N(1) -(4-azido-2-nitrophenyl)hexane-1,6-diamine are reported. Both photoprobes effectively entered HeLa cells (and the nucleus) and were dependent on the tetracysteine motif in recombinant DMRT1 (doublesex and Mab3-related transcription factor) to induce fluorescence, suggesting that their crosslinking abilities can be exploited for the identification of nucleic acids and proteins associated with a protein of interest.


Asunto(s)
Arsénico , Arsenicales/química , Azidas/química , Reactivos de Enlaces Cruzados/química , Diaminas/química , Diazometano/análogos & derivados , Diazometano/química , Fluoresceínas/química , Mercaptoetanol/análogos & derivados , Etiquetas de Fotoafinidad/química , Arsenicales/síntesis química , Azidas/síntesis química , Diaminas/síntesis química , Diazometano/síntesis química , Fluoresceínas/síntesis química , Células HeLa , Humanos , Mercaptoetanol/química , Etiquetas de Fotoafinidad/síntesis química , Relación Estructura-Actividad , Factores de Transcripción/metabolismo
2.
Biol Reprod ; 93(4): 83, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26269506

RESUMEN

The nuclear receptor steroidogenic factor 1 (SF-1, AD4BP, NR5A1) is a key regulator of the endocrine axes and is essential for adrenal and gonad development. Partial rescue of Nr5a1(-/-) mice with an SF-1-expressing transgene caused a hypomorphic phenotype that revealed its roles in Leydig cell development. In contrast to controls, all male rescue mice (Nr5a1(-/-);tg(+/0)) showed varying signs of androgen deficiency, including spermatogenic arrest, cryptorchidism, and poor virilization. Expression of various Leydig cell markers measured by immunohistochemistry, Western blot analysis, and RT-PCR indicated fetal and adult Leydig cell development were differentially impaired. Whereas fetal Leydig cell development was delayed in Nr5a1(-/-);tg(+/0) embryos, it recovered to control levels by birth. In contrast, Sult1e1, Vcam1, and Hsd3b6 transcript levels in adult rescue testes indicated complete blockage in adult Leydig cell development. In addition, between Postnatal Days 8 and 12, peritubular cells expressing PTCH1, SF-1, and CYP11A1 were observed in control testes but not in rescue testes, indicating SF-1 is needed for either survival or differentiation of adult Leydig cell progenitors. Cultured prepubertal rat peritubular cells also expressed SF-1 and PTCH1, but Cyp11a1 was expressed only after treatment with cAMP and retinoic acid. Together, data show SF-1 is needed for proper development of fetal and adult Leydig cells but with distinct primary functions; in fetal Leydig cells, it regulates differentiation, whereas in adult Leydig cells it regulates progenitor cell formation and/or survival.


Asunto(s)
Células Intersticiales del Testículo/fisiología , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/fisiología , Testículo/crecimiento & desarrollo , Andrógenos/deficiencia , Animales , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Hormonas Esteroides Gonadales/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Ratas , Túbulos Seminíferos/embriología , Túbulos Seminíferos/crecimiento & desarrollo , Túbulos Seminíferos/metabolismo , Células Madre , Factor Esteroidogénico 1/biosíntesis , Testículo/embriología , Testículo/metabolismo
3.
Biol Reprod ; 88(2): 51, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23255335

RESUMEN

DMRT1 is an evolutionarily conserved transcriptional factor expressed only in the postnatal testis, where it is produced in Sertoli cells and germ cells. While deletion of Dmrt1 in mice demonstrated it is required for postnatal testis development and fertility, much is still unknown about its temporal- and cell-specific functions. This study characterized a novel mouse model of DMRT1-deficient germ cells that was generated by breeding Dmrt1-null (Dmrt1(-/-)) mice with Wt1-Dmrt1 transgenic (Dmrt1(+/-;tg)) mice, which express a rat Dmrt1 cDNA in gonadal supporting cells by directing it from the Wilms tumor 1 locus in a yeast artificial chromosome transgene. Like Dmrt1(-/-) mice, male Dmrt1(-/-) transgenic mice (Dmrt1(-/-;tg)) were infertile, while female mice were fertile. Immunohistochemistry and Western blot analysis showed transgenic DMRT1 expressed in supporting cells of the newborn gonads of both sex and in Sertoli cells of the testis afterbirth. Sertoli cells were evaluated by electron microscopy, revealing that maturation of Dmrt1(-/-;tg) Sertoli cells was incomplete. Morphological analysis of testes from 42-day-old mice showed that, compared to Dmrt1(-/-) mice, Dmrt1(-/-;tg) mice have improved seminiferous tubule structure, with lumens present in many. Immunohistochemistry of the polarity markers ESPIN and NECTIN-2 showed that DMRT1 in Sertoli cells is required for NECTIN-2 expression and influences organization of ectoplasmic specializations. Further functional analyses of the transgene on a Dmrt1(-/-) background showed that it did not rescue the decrease in Dmrt1(-/-) testis size, but when expressed on a wild-type background, exogenous DMRT1 prevented the normal age-related decline in testis size and enhanced sperm progressive motility. The studies suggest that DMRT1 in Sertoli cells regulates tubule morphology, spermatogenesis, and sperm function via its effects on Sertoli cell maturation and polarity. Furthermore, expression and function of transgenic DMRT1 in Sertoli cells establishes a novel mouse model of DMRT1-deficient germ cells generated by breeding Dmrt1-null mice with Wt1-Dmrt1 transgenic mice (rescue; Dmrt1(-/-;tg)).


Asunto(s)
Modelos Animales , Células de Sertoli/metabolismo , Testículo/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transgenes/genética , Animales , Moléculas de Adhesión Celular/metabolismo , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Microscopía Electrónica de Transmisión , Nectinas , Células de Sertoli/patología , Células de Sertoli/ultraestructura , Motilidad Espermática/fisiología , Testículo/patología , Testículo/ultraestructura , Factores de Transcripción/metabolismo
4.
Proc Natl Acad Sci U S A ; 107(30): 13360-5, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20616082

RESUMEN

The DM domain proteins Doublesex- and MAB-3-related transcription factors (DMRTs) are widely conserved in metazoan sex determination and sexual differentiation. One of these proteins, DMRT1, plays diverse and essential roles in development of the vertebrate testis. In mammals DMRT1 is expressed and required in both germ cells and their supporting Sertoli cells. Despite its critical role in testicular development, little is known about how DMRT1 functions as a transcription factor or what genes it binds and regulates. We combined ChIP methods with conditional gene targeting and mRNA expression analysis and identified almost 1,400 promoter-proximal regions bound by DMRT1 in the juvenile mouse testis and determined how expression of the associated mRNAs is affected when Dmrt1 is selectively mutated in germ cells or Sertoli cells. These analyses revealed that DMRT1 is a bifunctional transcriptional regulator, activating some genes and repressing others. ChIP analysis using conditional mutant testes showed that DNA binding and transcriptional regulation of individual target genes can differ between germ cells and Sertoli cells. Genes bound by DMRT1 in vivo were enriched for a motif closely resembling the sequence DMRT1 prefers in vitro. Differential response of genes to loss of DMRT1 corresponded to differences in the enriched motif, suggesting that other transacting factors may modulate DMRT1 activity. DMRT1 bound its own promoter and those of six other Dmrt genes, indicating auto- and cross-regulation of these genes. Many of the DMRT1 target genes identified here are known to be important for a variety of functions in testicular development; the others are candidates for further investigation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Genoma , Testículo/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Sitios de Unión/genética , Proteínas Portadoras/clasificación , Proteínas Portadoras/genética , Línea Celular , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Unión Proteica , Testículo/crecimiento & desarrollo , Factores de Transcripción/genética , Activación Transcripcional , Transfección
5.
Biol Reprod ; 84(1): 7-17, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20739665

RESUMEN

Follicle-stimulating hormone (FSH), a pituitary glycoprotein hormone, is an integral component of the endocrine axis that regulates gonadal function and fertility. To transmit its signal, FSH must bind to its receptor (FSHR) located on Sertoli cells of the testis and granulosa cells of the ovary. Thus, both the magnitude and the target of hormone response are controlled by mechanisms that determine FSHR levels and cell-specific expression, which are supported by transcription of its gene. The present review examines the status of FSHR/Fshr gene regulation, emphasizing the importance of distal sequences in FSHR/Fshr transcription, new insights gained from the influx of genomics data and bioinformatics, and emerging trends that offer direction in deciphering the FSHR/Fshr regulatory landscape.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Receptores de HFE/metabolismo , Animales , Receptores de HFE/genética , Elementos Reguladores de la Transcripción/fisiología
6.
Biol Reprod ; 84(3): 422-34, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20962249

RESUMEN

Targets of steroidogenic factor 1 (SF1; also known as NR5A1 and AD4BP) have been identified within cells at every level of the hypothalamic-pituitary-gonadal and -adrenal axes, revealing SF1 to be a master regulator of major endocrine systems. Mouse embryos express SF1 in the genital ridge until Embryonic Day 13.5 (E13.5). Thereafter, expression persists in the male and is substantially lower in the female gonad until birth. We hypothesize that the sexually dimorphic expression of Sf1 during gonadogenesis is mediated by sex-specific regulation of its promoter. To investigate dimorphic regulation within the fetal gonad, we developed an experimental strategy using transient transfection of E13.5 gonad explant cultures and evaluated various Sf1 promoter constructs for sexually dimorphic DNA elements. The proximal Sf1 promoter correctly targeted reporter activity to SF1-expressing cells in both XY and XX gonads. Stepwise deletion of sequences from the Sf1 promoter revealed two regions that affected regulation within female gonads. Mutation of both sequences together did not cause further disruption of reporter activity, suggesting the two sites might work in concert to promote activity in female somatic cells. Results from gel mobility shift assays and fetal gonad-chromatin immunoprecipitation showed that TCFAP2 binds to one of the two female-specific sites within the proximal promoter of Sf1. Together, we show that transient transfection experiments performed within developing testes and ovaries are a powerful tool to uncover elements within the Sf1 promoter that contribute to sex-specific expression.


Asunto(s)
Ovario/embriología , Ovario/metabolismo , Regiones Promotoras Genéticas/fisiología , Factor Esteroidogénico 1/genética , Animales , Secuencia de Bases , Células Cultivadas , Electroporación , Femenino , Gónadas/embriología , Gónadas/metabolismo , Masculino , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Especificidad de Órganos/genética , Plásmidos/genética , Regiones Promotoras Genéticas/genética , Factor Esteroidogénico 1/metabolismo
8.
Endocrinology ; 161(5)2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32010941

RESUMEN

Differences of sex development (DSDs) are a constellation of conditions that result in genital ambiguity or complete sex reversal. Although determining the underlying genetic variants can affect clinical management, fewer than half of undermasculinized males ever receive molecular diagnoses. Next-generation sequencing (NGS) technology has improved diagnostic capabilities in several other diseases, and a few small studies suggest that it may improve molecular diagnostic capabilities in DSDs. However, the overall diagnostic rate that can be achieved with NGS for larger groups of patients with DSDs remains unknown. In this study, we aimed to implement a tiered approach to genetic testing in undermasculinized males seen in an interdisciplinary DSD clinic to increase the molecular diagnosis rate in this group. We determined the diagnosis rate in patients undergoing all clinically available testing. Patients underwent a stepwise approach to testing beginning with a karyotype and progressing through individual gene testing, microarray, panel testing, and then to whole-exome sequencing (WES) if no molecular cause was found. Deletion/duplication studies were also done if deletions were suspected. Sixty undermasculinized male participants were seen in an interdisciplinary DSD clinic from 2008 to 2016. Overall, 37/60 (62%) of patients with Y chromosomes and 46% of those who were 46XY received molecular diagnoses. Of the 46,XY patients who underwent all available genetic testing, 18/28 (64%) achieved molecular diagnoses. This study suggests that the addition of WES testing can result in a higher rate of molecular diagnoses compared to genetic panel testing.


Asunto(s)
Trastorno del Desarrollo Sexual 46,XY/genética , Trastornos del Desarrollo Sexual/genética , Secuenciación del Exoma/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Cariotipificación/métodos , Desarrollo Sexual/genética , Adolescente , Niño , Preescolar , Trastorno del Desarrollo Sexual 46,XY/diagnóstico , Trastornos del Desarrollo Sexual/diagnóstico , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Sensibilidad y Especificidad
9.
Endocrinology ; 149(10): 5297-306, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18566134

RESUMEN

Pituitary FSH promotes pubertal timing and normal gametogenesis by binding its receptor (FSHR) located on Sertoli and granulosa cells of the testis and ovary, respectively. Studies on Fshr transcription provide substantial evidence that upstream stimulatory factor (USF) 1 and USF2, basic helix-loop-helix leucine zipper proteins, regulate Fshr through an E-box within its promoter. However, despite the strong in vitro support for USF1 and USF2 in Fshr regulation, there is currently no in vivo corroborating evidence. In the present study, chromatin immunoprecipitation demonstrated specific binding of USF1 and USF2 to the Fshr promoter in both Sertoli and granulosa cells, in vivo. Control cells lacking Fshr expression showed no USF-Fshr promoter binding, thus correlating USF-promoter binding to gene activity. Evaluation of Fshr expression in Usf1 and Usf2 null mice further explored USF's role in Fshr transcription. Loss of either gene significantly reduced ovarian Fshr levels, whereas testis levels were unaltered. Chromatin immunoprecipitation analysis of USF-Fshr promoter binding in Usf-null mice indicated differences in the composition of promoter-bound USF dimers in granulosa and Sertoli cells. Promoter-bound USF dimer levels declined in granulosa cells from both null mice, despite increased USF2 levels in Usf1-null ovaries. However, compensatory increases in promoter-bound USF homodimers were evident in Usf-null Sertoli cells. In summary, this study provides the first in vivo evidence that USF1 and USF2 bind the Fshr promoter and revealed differences between Sertoli and granulosa cells in compensatory responses to USF loss and the USF dimeric composition required for Fshr transcription.


Asunto(s)
Células de la Granulosa/fisiología , Receptores de HFE/genética , Células de Sertoli/fisiología , Factores Estimuladores hacia 5'/metabolismo , Animales , Células Cultivadas , Femenino , Regulación de la Expresión Génica/fisiología , Células de la Granulosa/citología , Masculino , Ratones , Ratones Mutantes , Ovario/citología , Ovario/fisiología , Regiones Promotoras Genéticas/fisiología , Células de Sertoli/citología , Caracteres Sexuales , Testículo/citología , Testículo/fisiología , Factores Estimuladores hacia 5'/genética
10.
Mol Endocrinol ; 21(12): 2968-87, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17761949

RESUMEN

Steroidogenic factor-1 (SF-1/Ad4BP; NR5A1), a nuclear receptor transcription factor, has a pivotal role in adrenal and gonadal development in humans and mice. A frequent feature of childhood adrenocortical tumors is SF-1 amplification and overexpression. Here we show that an increased SF-1 dosage can by itself augment human adrenocortical cell proliferation through concerted actions on the cell cycle and apoptosis. This effect is dependent on an intact SF-1 transcriptional activity. Gene expression profiling showed that an increased SF-1 dosage regulates transcripts involved in steroid metabolism, the cell cycle, apoptosis, and cell adhesion to the extracellular matrix. Consistent with these results, increased SF-1 levels selectively modulate the steroid secretion profile of adrenocortical cells, reducing cortisol and aldosterone production and maintaining dehydroepiandrosterone sulfate secretion. As a model to understand the mechanisms of transcriptional regulation by increased SF-1 dosage, we studied FATE1, coding for a cancer-testis antigen implicated in the control of cell proliferation. Increased SF-1 levels increase its binding to a consensus site in FATE1 promoter and stimulate its activity through modulation of the recruitment of specific cofactors. On the other hand, sphingosine, which can compete with phospholipids for binding to SF-1, had no effect on the SF-1 dosage-dependent increase of adrenocortical cell proliferation and expression of the FATE1 promoter. In mice, increased Sf-1 dosage produces adrenocortical hyperplasia and formation of tumors expressing gonadal markers (Amh, Gata-4), which originate from the subcapsular region of the adrenal cortex. Gene expression profiling revealed that genes involved in cell adhesion and the immune response and transcription factor signal transducer and activator of transcription-3 (Stat3) are differentially expressed in Sf-1 transgenic mouse adrenals compared with wild-type adrenals. Our studies reveal a critical role for SF-1 dosage in adrenocortical tumorigenesis and constitute a rationale for the development of drugs targeting SF-1 transcriptional activity for adrenocortical tumor therapy.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias/metabolismo , Neoplasias/patología , Factor Esteroidogénico 1/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Matriz Extracelular/metabolismo , Histidina/genética , Histidina/metabolismo , Humanos , Metabolismo de los Lípidos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación/genética , Neoplasias/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factor Esteroidogénico 1/genética , Esteroides/metabolismo , Transcripción Genética/genética , Regulación hacia Arriba
11.
Mol Cell Endocrinol ; 260-262: 100-8, 2007 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-17084019

RESUMEN

The cell-surface receptor for the gonadotropin follicle-stimulating hormone (FSH) is expressed exclusively on Sertoli cells of the testis and granulosa cells of the ovary. FSH signal transduction through its receptor (Fshr) is critical for the timing and maintenance of normal gametogenesis in the mammalian gonad. In the 13 years since the gene encoding Fshr was first cloned, the mechanisms controlling its transcription have been extensively examined, but a clear understanding of what drives its unique cell-specificity remains elusive. Current knowledge of basal Fshr transcription highlights the role of an E-box in the proximal promoter which is bound by the basic helix-loop-helix transcription factors upstream stimulatory factor 1 (Usf1) and Usf2. Recent studies utilizing knockout mice and chromatin immunoprecipitation validated the importance of Usf to Fshr transcription and demonstrated a sexually dimorphic requirement for the Usf proteins to maintain normal Fshr expression. Studies have also shown that the promoter region itself is insufficient for appropriate Fshr expression in transgenic mice, indicating Fshr transcription depends on regulatory elements that lie outside of the promoter. Identification of such elements has been propelled by recent availability of genome sequence data, which facilitated studies using comparative genomics, DNase I hypersensitivity mapping, and transgenic analysis with large fragments of DNA. This review will focus on the current understanding of transcriptional regulatory processes that control expression of rat Fshr, including recent advances from our laboratory.


Asunto(s)
Regulación de la Expresión Génica , Receptores de HFE/genética , Transcripción Genética , Animales , Humanos , Modelos Genéticos , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores Estimuladores hacia 5'/metabolismo
12.
Mol Cell Endocrinol ; 260-262: 49-58, 2007 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-17097219

RESUMEN

The gonadotropin follicle-stimulating hormone (FSH) is required for initiation and maintenance of normal gametogenesis and acts through a specific, cell-surface receptor (Fshr) present only on Sertoli and granulosa cells in the gonads. Despite extensive examination of the transcriptional mechanisms regulating Fshr, the sequences directing its expression to these cells remain unidentified. To establish the minimal region necessary for Fshr expression, we generated transgenic mice carrying a yeast artificial chromosome (YAC) that contained 413 kilobases (kb) of the rat Fshr locus (YAC60). Transgene expression, as determined by RT-PCR, was absent from immature testis and Sertoli cells, limited to germ cells of the adult testis, and never observed in the ovary. While the data is limited to only one transgenic line, it suggests that the 413kb region does not specify the normal spatiotemporal expression pattern of Fshr. Comparative genomics was used to identify potential distal regulatory elements, revealing seven regions of high evolutionary conservation (>80% identity over 100bp or more), six of which were absent from the transgene. Functional examination of the evolutionary conserved regions (ECRs) by transient transfection revealed that all of the ECRs had modest transcriptional activity in Sertoli or myoid cells with two, ECR4 and ECR5, showing differential effects in expressing and non-expressing cells. These data reveal that distal regulatory regions (outside the 413kb in YAC60) are required for appropriate temporal and spatial Fshr expression and implicate the identified ECRs in transcriptional regulation of Fshr.


Asunto(s)
Regulación de la Expresión Génica/genética , Receptores de HFE/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Cromosomas Artificiales de Levadura , Secuencia Conservada , Evolución Molecular , Perfilación de la Expresión Génica , Humanos , Integrasas/metabolismo , Ratones , Ratones Transgénicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Recombinación Genética , Saccharomyces cerevisiae/genética , Transcripción Genética
13.
Mol Cell Biol ; 24(1): 377-88, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14673170

RESUMEN

The doublesex and mab-3 related transcription factor 1 (Dmrt1) is a putative transcriptional regulator that is expressed exclusively in the gonads and is required for postnatal testis differentiation. Here we describe the transcriptional mechanisms regulating testis-specific expression of the Dmrt1 gene. Transient-transfection analysis identified a region of the promoter between kb -3.2 and -2.8 that is important for Sertoli cell-specific expression. DNase I footprinting revealed four sites of DNA-protein interaction within this region, three of which were prominent in primary Sertoli cells. Analysis of these sites, using electrophoretic mobility shift assays, revealed that Gata4 and another unknown factor bound within these regions. Further transient-transfection assays of various mutant promoters established the functional relevance of the Gata4-response and unknown factor-response elements, while studies of Dmrt1 expression in 13.5 days postcoitum Fog2 null gonads supported the in vivo importance of Gata4's regulation. As a whole, these studies identify Gata4 as an important regulator in the Dmrt1 transcriptional machinery that is responsible for robust expression of Dmrt1 in the testis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Testículo/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , ADN/metabolismo , Factor de Transcripción GATA4 , Masculino , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Unión Proteica , Ratas , Células de Sertoli/metabolismo , Testículo/embriología , Factores de Transcripción/genética
14.
Mol Endocrinol ; 19(8): 2112-31, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15817654

RESUMEN

Expression of the FSH receptor (Fshr) is restricted to testicular Sertoli cells and ovarian granulosa cells, thereby limiting the direct targets of FSH action to these somatic cells of the gonads. Earlier studies indicate that transcription of Fshr in the gonads requires elements outside the gene's immediate 5' flanking sequence. To help uncover candidate regulatory sequences, comparative genomics and deoxyribonuclease I hypersensitivity mapping were employed. A total of 156 evolutionarily conserved sequences were found, and partial deoxyribonuclease I hypersensitivity mapping across 45 kb of 5' flanking sequence and the first intron identified four hypersensitive sites, DHS1-4. Notably, DHS1 and DHS2 localized to conserved sites in the promoter region and exon 1 and correlated with the active state of the gene. DHS3 also corresponded to a conserved site (site 7) but was more pronounced in nonexpressing myoid cells, suggesting a role in gene silencing. Transient transfection analysis of DHS3 confirmed its role in gene silencing, a function that was promoter, cell type, and position dependent. Protein-DNA binding studies on DHS3 revealed that octamer transcription factor 1 (OCT-1) and GATA-4 bound site 7, in vitro, and transient transfection analysis showed that their binding sites were required for silencing activity. Furthermore, chromatin immunoprecipitation revealed that OCT-1 bound to site 7 in the endogenous gene, but only in myoid cells. In contrast, GATA-1 bound site 7 predominantly in Sertoli cells, suggesting that it attenuates silencer activity. The findings reveal that OCT-1 binds within DHS3 to silence Fshr transcription and implicate members of the GATA family in the modulation of this activity.


Asunto(s)
Regulación de la Expresión Génica , Silenciador del Gen , Receptores de HFE/genética , Animales , Secuencia de Bases , Sitios de Unión , Núcleo Celular/metabolismo , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Mapeo Cromosómico , Secuencia Conservada , Desoxirribonucleasa I/metabolismo , Exones , Genes Reporteros , Genoma , Genómica/métodos , Humanos , Intrones , Masculino , Modelos Genéticos , Datos de Secuencia Molecular , Familia de Multigenes , Mutación , Plásmidos/metabolismo , Unión Proteica , Ratas , Células de Sertoli/metabolismo , Transcripción Genética , Transfección
15.
Mol Endocrinol ; 19(10): 2549-63, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15961510

RESUMEN

Steroidogenic factor 1 (SF-1/Nr5a1) is an orphan nuclear receptor encoded by the Ftz-F1 gene and is required for gonad and adrenal development and regulation of hormone production within the reproductive and adrenal axes. To extend our understanding of Ftz-F1 and its role in SF-1 expression, we identified and characterized a yeast artificial chromosome (YAC) containing Ftz-F1. Within this YAC, Ftz-F1 is centrally located and flanked by genes encoding a second orphan nuclear receptor, germ cell nuclear factor, and proteasome (prosome, macropain) subunit beta type 7. Three lines of transgenic mice carrying the YAC were generated and in two lines (lines 7 and 14), RT-PCR and ribonuclease protection analysis showed that expression of transgenic SF-1 mimicked that of endogenous SF-1, both spatially and quantitatively. In the third line (line 15), pituitary and hypothalamic expression were absent. Comparison of the integrated transgenes revealed that line 15 was truncated at the end of intron 4 and revealed a region within the locus that is responsible for SF-1 expression in the pituitary and hypothalamus. The line 14 transgene was introduced into a mouse strain lacking functional SF-1. Examination of SF-1-deficient, transgene-positive mice revealed that the YAC was able to rescue adrenal and gonad development, which normally arrests in the SF-1-null embryos and showed that the 153-kb transgene integrated in line 14 is sufficient to properly direct SF-1 expression and support its biological activity. Thus, the study defines a region of Ftz-F1 that contains the requisite set of regulatory elements to direct SF-1 cell-specific expression and all temporal and quantitative changes need for its biological activity.


Asunto(s)
Cromosomas Artificiales de Levadura/genética , Proteínas de Unión al ADN/genética , Factores de Transcripción/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , ADN/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Genitales/embriología , Genitales/metabolismo , Proteínas de Homeodominio , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Hipófisis/embriología , Hipófisis/metabolismo , Ratas , Receptores Citoplasmáticos y Nucleares , Factor Esteroidogénico 1 , Factores de Transcripción/deficiencia , Factores de Transcripción/fisiología
16.
Ann N Y Acad Sci ; 1061: 55-64, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16467257

RESUMEN

Steroidogenic factor 1 (SF-1, Nr5a1, and Ad4bp) is an orphan nuclear receptor required for adrenal and gonad development and endocrine regulation. To extend our understanding of SF-1 function and the mechanisms controlling its expression, a transgenic rescue strategy was employed to locate important transcriptional control regions and to reveal functional roles of the protein. A rat yeast artificial chromosome containing Ftz-F1, the gene encoding SF-1, was used to generate mice with different transgenes that varied in size. Rat SF-1 mRNA expression was assayed to assess each transgene's targeting ability. SF-1-deficient/transgene-positive (SF-1(-/-); tg/+) "rescue" mice were then generated and the animals' developmental and reproductive status was evaluated. The results identified differences in expression patterns and rescue abilities that provided insight into SF-1 transcriptional control and function. Comparing transgene maps and mRNA profiles placed critical transcriptional elements for pituitary and hypothalamic expression to a region 3' to intron 4, whereas examination of rescued mice revealed that an approximately 153-kb region of the Ftz-F1 locus recapitulates most or all activity ascribed to the endogenous allele. A second line of rescued mice was hypomorphic, with males showing defects in androgen-dependent tissues due to abnormal Leydig cell differentiation. Histological analysis of embryonic (e14.5) and adult testes from these mice implicated SF-1 in roles that are distinct in fetal and adult Leydig cells.


Asunto(s)
Proteínas de Homeodominio/fisiología , Células Intersticiales del Testículo/citología , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/fisiología , Animales , Diferenciación Celular , Cromosomas Artificiales de Levadura/genética , Cromosomas Artificiales de Levadura/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Genéticos , Ratas , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Factor Esteroidogénico 1 , Testículo/citología , Testículo/embriología , Testículo/crecimiento & desarrollo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transgenes/genética
17.
Endocrinology ; 155(7): 2349-54, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24712878

RESUMEN

SF-1 (NR5A1) overexpression can induce adrenocortical tumor formation in transgenic mice and is associated with more severe prognosis in patients with adrenocortical cancer. In this study we have identified Vanin-1 (Vnn1), a SF-1 target gene, as a novel modulator of the tumorigenic effect of Sf-1 overexpression in the adrenal cortex. Vanin-1 is endowed with pantetheinase activity, releasing cysteamine in tissues and regulating cell response to oxidative stress by modulating the production of glutathione. Sf-1 transgenic mice developed adrenocortical neoplastic lesions (both dysplastic and nodular) with a frequency increasing with age. Genetic ablation of the Vnn1 gene in Sf-1 transgenic mice significantly reduced the severity of neoplastic lesions in the adrenal cortex. This effect could be reversed by treatment of Sf-1 transgenic/Vnn1 null mice with cysteamine. These data show that alteration of the mechanisms controlling intracellular redox and detoxification mechanisms is relevant to the pathogenesis of adrenocortical neoplasia induced by SF-1 overexpression.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/metabolismo , Amidohidrolasas/metabolismo , Transformación Celular Neoplásica/metabolismo , Factor Esteroidogénico 1/metabolismo , Corteza Suprarrenal/efectos de los fármacos , Corteza Suprarrenal/metabolismo , Corteza Suprarrenal/patología , Neoplasias de la Corteza Suprarrenal/genética , Amidohidrolasas/genética , Animales , Transformación Celular Neoplásica/genética , Cisteamina/metabolismo , Cisteamina/farmacología , Progresión de la Enfermedad , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Glutatión/metabolismo , Hiperplasia , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Especies Reactivas de Oxígeno/metabolismo , Factor Esteroidogénico 1/genética , Factores de Tiempo
18.
Biol Reprod ; 81(1): 118-25, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19264703

RESUMEN

DMRT1 is a transcription factor expressed only in Sertoli cells and undifferentiated spermatogonia of the postnatal testis, where it is required for proper cellular differentiation and fertility. To elucidate the transcriptional regulatory regions that provide DMRT1's cell-specific expression, transgenic mice containing a LacZ reporter gene driven by variable amounts of rat Dmrt1 5' flanking sequence, 9 kb and smaller, were evaluated. Examination of transgene expression by RT-PCR indicated that multiple promoter regions direct Dmrt1 to the testis and that sequences upstream of 2.8 kb are needed for both Sertoli cell expression and limiting transcriptional influence imposed by surrounding chromatin. Thus, whereas many of the transgenes were expressed in the testis, the ones with smaller promoters were significantly more prone to expression at ectopic sites or to complete silencing. Transgene expression in Sertoli cells and germ cells was assessed by immunohistochemistry and RT-PCR following busulfan treatment to remove germ cells. Both evaluations indicated expression of the 9- and 3.2-kb promoters in Sertoli cells and germ cells, whereas activity of smaller promoters was largely restricted to germ cells. In all, the present study provides in vivo evidence that distinct promoter sequences participate in Dmrt1 regulation in somatic cells and germ cells, with the -3.2 kb/-2.8 kb region directing expression in Sertoli cells and downstream sequences (< or =1.3 kb) directing it in germ cells. Further exploration of the mechanisms restricting Dmrt1 expression to the testis revealed that FOXL2, a transcription factor required for differentiation of the ovary, repressed Dmrt1 promoter through the -3.2 kb/-2.8 kb regulatory region, offering a potential mechanism for Dmrt1 transcriptional silencing in granulosa cells.


Asunto(s)
Células Germinativas/metabolismo , Regiones Promotoras Genéticas , Células de Sertoli/metabolismo , Factores de Transcripción/genética , Animales , Secuencia de Bases , Células Cultivadas , Femenino , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/fisiología , Regulación de la Expresión Génica , Operón Lac , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Especificidad de Órganos/genética , Regiones Promotoras Genéticas/fisiología , Ratas , Factores de Transcripción/metabolismo , Transcripción Genética
19.
Biol Reprod ; 78(6): 1139-52, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18218611

RESUMEN

Gamendazole was recently identified as an orally active antispermatogenic compound with antifertility effects. The cellular mechanism(s) through which these effects occur and the molecular target(s) of gamendazole action are currently unknown. Gamendazole was recently designed as a potent orally active antispermatogenic male contraceptive agent. Here, we report the identification of binding targets and propose a testable mechanism of action for this antispermatogenic agent. Both HSP90AB1 (previously known as HSP90beta [heat shock 90-kDa protein 1, beta]) and EEF1A1 (previously known as eEF1A [eukaryotic translation elongation factor 1 alpha 1]) were identified as binding targets by biotinylated gamendazole (BT-GMZ) affinity purification from testis, Sertoli cells, and ID8 ovarian cancer cells; identification was confirmed by matrix-assisted laser desorption/ionization-time of flight mass spectrometry and Western blot analysis. BT-GMZ bound to purified yeast HSP82 (homologue to mammalian HSP90AB1) and EEF1A1, but not to TEF3 or HBS1, and was competed by unlabeled gamendazole. However, gamendazole did not inhibit nucleotide binding by EEF1A1. Gamendazole binding to purified Saccharomyces cerevisiae HSP82 inhibited luciferase refolding and was not competed by the HSP90 drugs geldanamycin or novobiocin analogue, KU-1. Gamendazole elicited degradation of the HSP90-dependent client proteins AKT1 and ERBB2 and had an antiproliferative effect in MCF-7 cells without inducing HSP90. These data suggest that gamendazole may represent a new class of selective HSP90AB1 and EEF1A1 inhibitors. Testis gene microarray analysis from gamendazole-treated rats showed a marked, rapid increase in three interleukin 1 genes and Nfkbia (NF-kappaB inhibitor alpha) 4 h after oral administration. A spike in II1a transcription was confirmed by RT-PCR in primary Sertoli cells 60 min after exposure to 100 nM gamendazole, demonstrating that Sertoli cells are a target. AKT1, NFKB, and interleukin 1 are known regulators of the Sertoli cell-spermatid junctional complexes. A current model for gamendazole action posits that this pathway links interaction with HSP90AB1 and EEF1A1 to the loss of spermatids and resulting infertility.


Asunto(s)
Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Indazoles/farmacología , Interleucina-1alfa/genética , Factor 1 de Elongación Peptídica/antagonistas & inhibidores , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Bloqueadores de Espermatogénesis/farmacología , Administración Oral , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Línea Celular Tumoral , Femenino , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Indazoles/administración & dosificación , Masculino , Modelos Biológicos , Datos de Secuencia Molecular , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Factor 1 de Elongación Peptídica/genética , Factor 1 de Elongación Peptídica/metabolismo , Ratas , Bloqueadores de Espermatogénesis/administración & dosificación , Testículo/efectos de los fármacos , Testículo/metabolismo , Transcripción Genética/efectos de los fármacos
20.
Biol Reprod ; 77(3): 466-75, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17567962

RESUMEN

Immunohistochemistry was used to examine GCNA1, a germ cell-specific protein, together with DMRT1 (Doublesex and Mab-3-related transcription factor-1), a transcription factor implicated in Sertoli cell and germ cell function, in order to resolve DMRT1's cellular profile during pre- and postnatal gonad development in the mouse. In the indifferent gonad (10.5-11.5 days postcoitus [dpc]), DMRT1 localized to somatic cells and GCNA1(+) germ cells and was indistinguishable in males and females. By 12.5 dpc, a clear sexual preference for DMRT1 in male somatic cells was observed, with male DMRT1 localized to testicular cords and more abundant in Sertoli cells than in germ cells and female DMRT1 diffusely labeled and markedly lower in somatic cells than in germ cells. A male somatic preference continued throughout development, with DMRT1 evident in Sertoli cells at all ages examined and absent in ovarian somatic cells from 13.5 dpc onward. In contrast, expression in primordial germ cells was not sexually distinct, and both sexes showed DMRT1 increasing through 13.5 dpc and absent by 15.5 dpc. Notably, sexual differences in germ cell DMRT1 were detected after birth, when it was detected only in spermatogonia of the testis. Colocalization of DMRT1 with proliferation markers KI67 and proliferating cell nuclear antigen (PCNA) and stem cell markers OCT4 (also known as POU5F1) and NGN3 indicated that, in postnatal testes, DMRT1 was present in both stem and proliferating spermatogonia. Together, the findings implicate opposite functions for DMRT1 in somatic and germ cells of the testis. In Sertoli cells, DMRT1 expression correlated with differentiation, whereas in germ cells, it suggested a role in expansion and maintenance of undifferentiated spermatogonia.


Asunto(s)
Desarrollo Embrionario/fisiología , Ovario/metabolismo , Testículo/metabolismo , Factores de Transcripción/biosíntesis , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Western Blotting , Proteínas de Unión al ADN/biosíntesis , Femenino , Inmunohistoquímica , Antígeno Ki-67/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/biosíntesis , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Ovario/embriología , Ovario/crecimiento & desarrollo , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Receptores Citoplasmáticos y Nucleares/biosíntesis , Diferenciación Sexual/fisiología , Testículo/embriología , Testículo/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda