Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros













Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 108(26): 10673-8, 2011 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-21636788

RESUMEN

The transcription factor steroidogenic factor 1 (SF-1) is exclusively expressed in the brain in the ventral medial hypothalamic nucleus (VMH) and is required for the development of this nucleus. However, the physiological importance of transcriptional programs regulated by SF-1 in the VMH is not well defined. To delineate the functional significance of SF-1 itself in the brain, we generated pre- and postnatal VMH-specific SF-1 KO mice. Both models of VMH-specific SF-1 KO were susceptible to high fat diet-induced obesity and displayed impaired thermogenesis after acute exposure to high fat diet. Furthermore, VMH-specific SF-1 KO mice showed significantly decreased LepR expression specifically in the VMH, leading to leptin resistance. Collectively, these results indicate that SF-1 directs transcriptional programs in the hypothalamus relevant to coordinated control of energy homeostasis, especially after excess caloric intake.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Ingestión de Energía , Leptina/fisiología , Factor Esteroidogénico 1/fisiología , Termogénesis , Núcleo Hipotalámico Ventromedial/fisiología , Animales , Homeostasis , Ratones , Ratones Noqueados , Factor Esteroidogénico 1/genética
2.
Endocrinology ; 152(1): 93-102, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21106871

RESUMEN

Glucose-6-phosphate (G6P) metabolism by the enzyme hexose-6-phosphate dehydrogenase (H6PDH) within the sarcoplasmic reticulum lumen generates nicotinamide adenine dinucleotide phosphate (reduced) to provide the redox potential for the enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) to activate glucocorticoid (GC). H6PDH knockout (KO) mice have a switch in 11ß-HSD1 activity, resulting in GC inactivation and hypothalamic-pituitary-adrenal axis activation. Importantly, H6PDHKO mice develop a type II fiber myopathy with abnormalities in glucose metabolism and activation of the unfolded protein response (UPR). GCs play important roles in muscle physiology, and therefore, we have examined the importance of 11ß-HSD1 and GC metabolism in mediating aspects of the H6PDHKO myopathy. To achieve this, we examined 11ß-HSD1/H6PDH double-KO (DKO) mice, in which 11ß-HSD1 mediated GC inactivation is negated. In contrast to H6PDHKO mice, DKO mice GC metabolism and hypothalamic-pituitary-adrenal axis set point is similar to that observed in 11ß-HSD1KO mice. Critically, in contrast to 11ß-HSD1KO mice, DKO mice phenocopy the salient features of the H6PDHKO, displaying reduced body mass, muscle atrophy, and vacuolation of type II fiber-rich muscle, fasting hypoglycemia, increased muscle glycogen deposition, and elevated expression of UPR genes. We propose that muscle G6P metabolism through H6PDH may be as important as changes in the redox environment when considering the mechanism underlying the activation of the UPR and the ensuing myopathy in H6PDHKO and DKO mice. These data are consistent with an 11ß-HSD1-independent function for H6PDH in which sarcoplasmic reticulum G6P metabolism and nicotinamide adenine dinucleotide phosphate-(oxidized)/nicotinamide adenine dinucleotide phosphate (reduced) redox status are important for maintaining muscle homeostasis.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Deshidrogenasas de Carbohidratos/metabolismo , Homeostasis/fisiología , Músculo Esquelético/fisiología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Animales , Glucemia , Deshidrogenasas de Carbohidratos/genética , Corticosterona/sangre , Regulación Enzimológica de la Expresión Génica , Insulina/sangre , Ratones , Ratones Noqueados , Enfermedades Musculares/enzimología , Enfermedades Musculares/genética
3.
Mol Endocrinol ; 24(6): 1240-50, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20339005

RESUMEN

The ventromedial hypothalamic nucleus (VMH) regulates a variety of homeostatic processes including female sexual behavior and reproduction. In the current study, we assessed the roles of steroidogenic factor 1 (SF-1) on reproductive function in the VMH using central nervous system-specific SF-1 knockout (SF-1 KO(nCre;F/-)) mice. Here we show that SF-1 KO(nCre;F/-) females exhibited marked impairment in female reproduction. Although male mice appeared to be normal in all aspects studied, including sexual behavior, SF-1 KO(nCre;F/-) females showed infertility or subfertility. Although adult SF-1 KO(nCre;F/-) females showed decreased or lacked corpora lutea, exogenous administration of gonadotropins induced the formation of multiple corpora lutea and induced normal ovulation, demonstrating that the ovaries are functionally intact. In addition, SF-1 KO(nCre;F/-) females stimulated with a synthetic GnRH agonist after priming exhibited markedly reduced LH secretion compared with wild-type littermates, arguing that disorganization in and around the VMH caused by SF-1 ablation interferes with the GnRH priming process or gonadotrope LH capacity. Furthermore, the SF-1 KO(nCre;F/-) females primed with estrogen benzoate and progesterone failed to induce steroid receptors around the VMH, consistent with impaired lordosis behavior in the SF-1 KO(nCre;F/-) females. Collectively, our results highlight that SF-1 in the VMH plays crucial roles in regulation of female reproductive function, presumably by organizing a precise neuronal connection and communication in and around the VMH.


Asunto(s)
Sistema Nervioso Central/metabolismo , Reproducción/fisiología , Factor Esteroidogénico 1/metabolismo , Animales , Estradiol/sangre , Ciclo Estral/metabolismo , Femenino , Fertilidad/fisiología , Hormona Folículo Estimulante/sangre , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Hipotálamo/patología , Integrasas/metabolismo , Hormona Luteinizante/sangre , Ratones , Ratones Noqueados , Especificidad de Órganos , Ovario/metabolismo , Ovario/patología , Ovulación/sangre , Progesterona/sangre , Conducta Sexual Animal
4.
Endocrinology ; 151(3): 1119-28, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20118198

RESUMEN

The adrenal capsule is postulated to harbor stem/progenitor cells, the progenies of which contribute to the growth of adrenocortex. We discovered that cells in the adrenal capsule are positive for Ptch1 and Gli1, genes indicative of responsiveness to the stimulation of Hedgehog (Hh) ligands. On the other hand, Sonic hedgehog (Shh), one of the mammalian Hh ligands, is expressed in the adrenocortex underneath the adrenal capsule, possibly acting upon the Hh-Responsive capsule. To investigate the functional significance of Shh in adrenal growth, we ablated Shh in an adrenocortex-specific manner using the Steroidogenic factor 1-Cre mouse. Loss of Shh in the adrenocortex led to reduced proliferation of capsular cells and a 50-75% reduction in adrenocortex thickness and adrenal size. The remaining adrenocortex underwent proper zonation and was able to synthesize steroids, indicating that Shh is dispensable for differentiation of adrenocortex. When these animals reached adulthood, their adrenocortex did not undergo compensatory growth in response to a high level of plasma ACTH, and the size of the adrenal remained significantly smaller than the control adrenal. Using a genetic lineage-tracing model, we further demonstrated that the Hh-responding cells in the adrenal capsule migrated centripetally into the adrenocortex. Our results not only provide the genetic evidence to support that the adrenal capsule contributes to the growth of adrenocortex in both fetal and adult life but also identify a novel role of Shh in this process.


Asunto(s)
Corteza Suprarrenal/crecimiento & desarrollo , Proteínas Hedgehog/metabolismo , Células Madre/fisiología , Factor Esteroidogénico 1/metabolismo , Corteza Suprarrenal/citología , Corteza Suprarrenal/embriología , Corteza Suprarrenal/enzimología , Corticoesteroides/biosíntesis , Animales , Diferenciación Celular , Linaje de la Célula , Femenino , Proteínas Hedgehog/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tamaño de los Órganos , Embarazo , Transducción de Señal
6.
Proc Natl Acad Sci U S A ; 106(52): 22323-8, 2009 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-20007774

RESUMEN

Dmrt1 (doublesex and mab-3 related transcription factor 1) is a conserved transcriptional regulator of male differentiation required for testicular development in vertebrates. Here, we show that in mice of the 129Sv strain, loss of Dmrt1 causes a high incidence of teratomas, whereas these tumors do not form in Dmrt1 mutant C57BL/6J mice. Conditional gene targeting indicates that Dmrt1 is required in fetal germ cells but not in Sertoli cells to prevent teratoma formation. Mutant 129Sv germ cells undergo apparently normal differentiation up to embryonic day 13.5 (E13.5), but some cells fail to arrest mitosis and ectopically express pluripotency markers. Expression analysis and chromatin immunoprecipitation identified DMRT1 target genes, whose missexpression may underlie teratoma formation. DMRT1 indirectly activates the GDNF coreceptor Ret, and it directly represses the pluripotency regulator Sox2. Analysis of human germ cell tumors reveals similar gene expression changes correlated to DMRT1 levels. Dmrt1 behaves genetically as a dose-sensitive tumor suppressor gene in 129Sv mice, and natural variation in Dmrt1 activity can confer teratoma susceptibility. This work reveals a genetic link between testicular dysgenesis, pluripotency regulation, and teratoma susceptibility that is highly sensitive to genetic background and to gene dosage.


Asunto(s)
Células Madre Fetales/citología , Células Madre Fetales/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Espermatogénesis/genética , Espermatogénesis/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Dosificación de Gen , Expresión Génica , Genes Supresores de Tumor , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias de Células Germinales y Embrionarias/metabolismo , Neoplasias de Células Germinales y Embrionarias/patología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Teratoma/genética , Teratoma/metabolismo , Teratoma/patología , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patología
7.
Mol Endocrinol ; 23(10): 1657-67, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19628584

RESUMEN

Deficiency of adrenal 4 binding protein/steroidogenic factor 1 (Ad4BP/SF-1; NR5A1) impairs adrenal development in a dose-dependent manner, whereas overexpression of Ad4BP/SF-1 is associated with adrenocortical tumorigenesis. Despite its essential roles in adrenal development, the mechanism(s) by which Ad4BP/SF-1 regulates this process remain incompletely understood. We previously identified a fetal adrenal enhancer (FAdE) that stimulates Ad4BP/SF-1 expression in the fetal adrenal gland by a two-step mechanism in which homeobox proteins initiate Ad4BP/SF-1 expression, which then maintains FAdE activity in an autoregulatory loop. In the present study, we examined the effect of transgenic expression of Ad4BP/SF-1 controlled by FAdE on adrenal development. When Ad4BP/SF-1 was overexpressed using a FAdE-Ad4BP/SF-1 transgene, FAdE activity expanded outside of its normal field, resulting in increased adrenal size and the formation of ectopic adrenal tissue in the thorax. The increased size of the adrenal gland did not result from a corresponding increase in cell proliferation, suggesting rather that the increased levels of Ad4BP/SF-1 may divert uncommitted precursors to the steroidogenic lineage. The effects of FAdE-controlled Ad4BP/SF-1 overexpression in mice provide a novel model of ectopic adrenal formation that further supports the critical role of Ad4BP/SF-1 in the determination of steroidogenic cell fate in vivo.


Asunto(s)
Glándulas Suprarrenales/embriología , Glándulas Suprarrenales/patología , Coristoma/metabolismo , Feto/citología , Regulación del Desarrollo de la Expresión Génica , Células Madre/metabolismo , Factor Esteroidogénico 1/genética , Corteza Suprarrenal/metabolismo , Corteza Suprarrenal/patología , Glándulas Suprarrenales/metabolismo , Envejecimiento/patología , Animales , Proliferación Celular , Coristoma/patología , Elementos de Facilitación Genéticos/genética , Ratones , Ratones Transgénicos , Células Madre/patología , Factor Esteroidogénico 1/metabolismo , Tórax/patología , Transgenes/genética , Sistema Urogenital/embriología , Sistema Urogenital/patología , beta-Galactosidasa/metabolismo
8.
Dev Biol ; 329(1): 96-103, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19268447

RESUMEN

Proper cell fate determination in mammalian gonads is critical for the establishment of sexual identity. The Hedgehog (Hh) pathway has been implicated in cell fate decision for various organs, including gonads. Desert Hedgehog (Dhh), one of the three mammalian Hh genes, has been implicated with other genes in the establishment of mouse fetal Leydig cells. To investigate whether Hh alone is sufficient to induce fetal Leydig cell differentiation, we ectopically activated the Hh pathway in Steroidogenic factor 1 (SF1)-positive somatic cell precursors of fetal ovaries. Hh activation transformed SF1-positive somatic ovarian cells into functional fetal Leydig cells. These ectopic fetal Leydig cells produced androgens and insulin-like growth factor 3 (INLS3) that cause virilization of female embryos and ovarian descent. However, the female reproductive system remained intact, indicating a typical example of female pseudohermaphroditism. The appearance of fetal Leydig cells was a direct consequence of Hh activation as evident by the absence of other testicular components in the affected ovary. This study provides not only insights into mechanisms of cell lineage specification in gonads, but also a model to understand defects in sexual differentiation.


Asunto(s)
Trastornos del Desarrollo Sexual/metabolismo , Feto , Proteínas Hedgehog/metabolismo , Células Intersticiales del Testículo/metabolismo , Ovario/metabolismo , Animales , Diferenciación Celular/genética , Trastornos del Desarrollo Sexual/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Feto/citología , Feto/metabolismo , Proteínas Hedgehog/genética , Inmunohistoquímica , Hibridación in Situ , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/fisiología , Masculino , Ratones , Ratones Transgénicos , Diferenciación Sexual , Transducción de Señal/genética
9.
Mol Cell Endocrinol ; 300(1-2): 132-6, 2009 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-18951950

RESUMEN

Steroidogenic factor 1 (SF-1) is a nuclear receptor that plays important roles in the hypothalamus-pituitary-steroidogenic organ axis. Global knockout studies in mice revealed the essential in vivo roles of SF-1 in the ventromedial hypothalamic (VMH) nucleus, adrenal glands, and gonads. One limitation of global SF-1 knockout mice is their early postnatal death from adrenocortical insufficiency. To overcome limitations of the global knockout mice and to delineate the roles of SF-1 in the brain, we used Cre/loxP recombination technology to genetically ablate SF-1 specifically in the central nervous system (CNS). Mice with CNS-specific knockout of SF-1 mediated by nestin-Cre showed increased anxiety-like behavior, revealing a crucial role of SF-1 in a complex behavioral phenotype. Our studies with CNS-specific SF-1 KO mice also defined roles of SF-1 in regulating the VMH expression of target genes implicated in anxiety and energy homeostasis. Therefore, this review will focus on our recent studies defining the functional roles of SF-1 in the VMH linked to anxiety and energy homeostasis.


Asunto(s)
Sistema Nervioso Central , Factor Esteroidogénico 1/metabolismo , Animales , Ansiedad/metabolismo , Conducta Animal/fisiología , Sistema Nervioso Central/anatomía & histología , Sistema Nervioso Central/metabolismo , Metabolismo Energético , Homeostasis , Ratones , Ratones Noqueados , Neuronas/citología , Neuronas/fisiología , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo , Factor Esteroidogénico 1/genética
10.
Mol Cell Biol ; 28(23): 7030-40, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18809574

RESUMEN

The nuclear receptor Ad4BP/SF-1 is essential for development of the adrenal cortex and the gonads, which derive from a common adrenogonadal primordium. The adrenal cortex subsequently forms morphologically distinct compartments: the inner (fetal) and outer (definitive or adult) zones. Despite considerable effort, the mechanisms that mediate the differential development of the adrenal and gonadal primordia and the fetal and adult adrenal cortices remain incompletely understood. We previously identified a fetal adrenal-specific enhancer (FAdE) in the Ad4BP/SF-1 locus that directs transgene expression to the fetal adrenal cortex and demonstrated that this enhancer is autoregulated by Ad4BP/SF-1. We now combine the FAdE with the Cre/loxP system to trace cell lineages in which the FAdE was active at some stage in development. These lineage-tracing studies establish definitively that the adult cortex derives from precursor cells in the fetal cortex in which the FAdE was activated before the organization into two distinct zones. The potential of these fetal adrenocortical cells to enter the pathway that eventuates in cells of the adult cortex disappeared by embryonic day 14.5. Thus, these studies demonstrate a direct link between the fetal and adult cortices involving a transition that must occur before a specific stage of development.


Asunto(s)
Corteza Suprarrenal/crecimiento & desarrollo , Linaje de la Célula , Animales , Feto , Integrasas , Ratones , Ratones Transgénicos , Células Madre , Factor Esteroidogénico 1/fisiología , Transgenes
11.
Biol Reprod ; 79(6): 1074-83, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18703422

RESUMEN

The nuclear receptor steroidogenic factor 1 (SF-1 [officially designated NR5A1]) is essential for fetal gonadal development, but its roles in postnatal ovarian function are less well defined. Herein, we have extended our analyses of knockout (KO) mice with markedly decreased SF-1 expression in granulosa cells. As described, these SF-1 KO mice had hypoplastic ovaries that contained a decreased number of follicles and lacked corpora lutea. In the present study, we showed that SF-1 KO mice exhibited abnormal estrous cycles, were infertile, and released significantly fewer oocytes in response to a standard superovulation regimen. Moreover, they had blunted induction of plasma estradiol in response to gonadotropins. The granulosa cell-specific SF-1 KO also significantly affected ovarian expression of putative SF-1 target genes. Consistent with their decreased follicle number, these mice had reduced ovarian expression of anti-müllerian hormone (Amh), which correlates with the reserve pool of ovarian follicles, as well as decreased gonadotropin-induced ovarian expression of aromatase (Cyp19a1) and cyclin D2 (Ccnd2). In contrast, perhaps because of their abnormal cyclicity, SF-1 KO ovaries had higher basal expression of inhibin-alpha. They also had decreased immunoreactivity for genes related to proliferation (Ccnd2 and Mki67 [also known as Ki67]) and increased expression of Cdkn1b, also known as p27, which inhibits cyclin-dependent kinases, arguing for a role of SF-1 in granulosa cell proliferation. These findings demonstrate that SF-1 has a key role in female reproduction via essential actions in granulosa cells.


Asunto(s)
Células de la Granulosa/fisiología , Infertilidad Femenina/genética , Folículo Ovárico/fisiología , Ovario/citología , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/fisiología , Alelos , Animales , Recuento de Células , Ciclo Estral/genética , Ciclo Estral/fisiología , Femenino , Fertilidad/genética , Fertilidad/fisiología , Regulación de la Expresión Génica/fisiología , Inmunohistoquímica , Ratones , Ratones Noqueados , Tamaño de los Órganos/fisiología , Embarazo , ARN/biosíntesis , ARN/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superovulación/fisiología , Útero/anatomía & histología , Útero/crecimiento & desarrollo
12.
Hum Mol Genet ; 17(19): 2949-55, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18617533

RESUMEN

During mammalian sex determination, expression of the Y-linked gene Sry shifts the bipotential gonad toward a testicular fate by upregulating a feed-forward loop between FGF9 and SOX9 to establish SOX9 expression in somatic cells. We previously proposed that these signals are mutually antagonistic with counteracting signals in XX gonads and that a shift in the balance of these factors leads to either male or female development. Evidence in mice and humans suggests that the male pathway is opposed by the expression of two signals, WNT4 and R-SPONDIN-1 (RSPO1), that promote the ovarian fate and block testis development. Both of these ligands can activate the canonical Wnt signaling pathway. Duplication of the distal portion of chromosome 1p, which includes both WNT4 and RSPO1, overrides the male program and causes male-to-female sex reversal in XY patients. To determine whether activation of beta-catenin is sufficient to block the testis pathway, we have ectopically expressed a stabilized form of beta-catenin in the somatic cells of XY gonads. Our results show that activation of beta-catenin in otherwise normal XY mice effectively disrupts the male program and results in male-to-female sex-reversal. The identification of beta-catenin as a key pro-ovarian and anti-testis signaling molecule will further our understanding of the mechanisms controlling sex determination and the molecular mechanisms that lead to sex-reversal.


Asunto(s)
Trastornos del Desarrollo Sexual , Regulación del Desarrollo de la Expresión Génica , Ovario/crecimiento & desarrollo , Testículo/crecimiento & desarrollo , beta Catenina/metabolismo , Animales , Femenino , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovario/metabolismo , Fenotipo , Diferenciación Sexual , Transducción de Señal , Testículo/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt4 , beta Catenina/genética
13.
Development ; 135(15): 2593-602, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18599507

RESUMEN

The nuclear receptor steroidogenic factor 1 (Sf1, Nr5a1) is essential for adrenal development and regulates genes that specify differentiated adrenocortical function. The transcriptional coactivator beta-catenin reportedly synergizes with Sf1 to regulate a subset of these target genes; moreover, Wnt family members, signaling via beta-catenin, are also implicated in adrenocortical development. To investigate the role of beta-catenin in the adrenal cortex, we used two Sf1/Cre transgenes to inactivate conditional beta-catenin alleles. Inactivation of beta-catenin mediated by Sf1/Cre(high), a transgene expressed at high levels, caused adrenal aplasia in newborn mice. Analysis of fetal adrenal development with Sf1/Cre(high)-mediated beta-catenin inactivation showed decreased proliferation in presumptive adrenocortical precursor cells. By contrast, the Sf1/Cre(low) transgene effected a lesser degree of beta-catenin inactivation that did not affect all adrenocortical cells, permitting adrenal survival to reveal age-dependent degeneration of the cortex. These results define crucial roles for beta-catenin--presumably as part of the Wnt canonical signaling pathway--in both embryonic development of the adrenal cortex and in maintenance of the adult organ.


Asunto(s)
Corteza Suprarrenal/metabolismo , Factor Esteroidogénico 1/metabolismo , beta Catenina/metabolismo , Corteza Suprarrenal/citología , Corteza Suprarrenal/embriología , Corteza Suprarrenal/crecimiento & desarrollo , Animales , Biomarcadores , Diferenciación Celular , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica , Integrasas/genética , Integrasas/metabolismo , Ratones , Ratones Noqueados , Transducción de Señal , Factor Esteroidogénico 1/genética , Proteínas Wnt/metabolismo , beta Catenina/deficiencia , beta Catenina/genética
14.
Mol Endocrinol ; 22(8): 1950-61, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18511494

RESUMEN

The nuclear receptor steroidogenic factor 1 (SF-1) plays essential roles in the development and function of the ventromedial hypothalamic nucleus (VMH). Considerable evidence links the VMH and SF-1 with the regulation of energy homeostasis. Here, we demonstrate that SF-1 colocalizes in VMH neurons with the cannabinoid receptor 1 (CB1R) and that a specific CB1R agonist modulates electrical activity of SF-1 neurons in hypothalamic slice preparations. We further show that SF-1 directly regulates CB1R gene expression via a SF-1-responsive element at -101 in its 5'-flanking region. Finally, we show that knockout mice with selective inactivation of SF-1 in the brain have decreased expression of CB1R in the region of the VMH and exhibit a blunted response to systemically administered CB1R agonists. These studies suggest that SF-1 directly regulates the expression of CB1R, which has been implicated in the regulation of energy homeostasis and anxiety-like behavior.


Asunto(s)
Regulación de la Expresión Génica , Receptor Cannabinoide CB1/genética , Factor Esteroidogénico 1/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Ácidos Araquidónicos/administración & dosificación , Ácidos Araquidónicos/farmacología , Peso Corporal/efectos de los fármacos , Línea Celular , Células Cultivadas , Oscuridad , Electrofisiología , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Conducta Alimentaria/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Técnicas In Vitro , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fosforilación/efectos de los fármacos , Receptor Cannabinoide CB1/metabolismo , Elementos de Respuesta , Núcleo Hipotalámico Ventromedial/efectos de los fármacos , Núcleo Hipotalámico Ventromedial/enzimología
15.
Mol Endocrinol ; 22(6): 1403-15, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18372344

RESUMEN

Steroidogenic factor 1 (SF-1) plays key roles in adrenal and gonadal development, expression of pituitary gonadotropins, and development of the ventromedial hypothalamic nucleus (VMH). If kept alive by adrenal transplants, global knockout (KO) mice lacking SF-1 exhibit delayed-onset obesity and decreased locomotor activity. To define specific roles of SF-1 in the VMH, we used the Cre-loxP system to inactivate SF-1 in a central nervous system (CNS)-specific manner. These mice largely recapitulated the VMH structural defect seen in mice lacking SF-1 in all tissues. In multiple behavioral tests, mice with CNS-specific KO of SF-1 had significantly more anxiety-like behavior than wild-type littermates. The CNS-specific SF-1 KO mice had diminished expression or altered distribution in the mediobasal hypothalamus of several genes whose expression has been linked to stress and anxiety-like behavior, including brain-derived neurotrophic factor, the type 2 receptor for CRH (Crhr2), and Ucn 3. Moreover, transfection and EMSAs support a direct role of SF-1 in Crhr2 regulation. These findings reveal important roles of SF-1 in the hypothalamic expression of key regulators of anxiety-like behavior, providing a plausible molecular basis for the behavioral effect of CNS-specific KO of this nuclear receptor.


Asunto(s)
Ansiedad/genética , Sistema Nervioso Central/metabolismo , Factor Esteroidogénico 1/genética , Animales , Animales Recién Nacidos , Conducta Animal/fisiología , Sitios de Unión , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Células COS , Chlorocebus aethiops , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos/genética , Regiones Promotoras Genéticas , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Factor Esteroidogénico 1/metabolismo , Factor Esteroidogénico 1/fisiología
16.
Endocrinology ; 149(5): 2138-48, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18258679

RESUMEN

Leptin, an adipocyte-derived hormone, has emerged as a critical regulator of energy homeostasis. The leptin receptor (Lepr) is expressed in discrete regions of the brain; among the sites of highest expression are several mediobasal hypothalamic nuclei known to play a role in energy homeostasis, including the arcuate nucleus, the ventromedial hypothalamic nucleus (VMH), and the dorsomedial hypothalamic nucleus. Although most studies have focused on leptin's actions in the arcuate nucleus, the role of Lepr in these other sites has received less attention. To explore the role of leptin signaling in the VMH, we used bacterial artificial chromosome transgenesis to target Cre recombinase to VMH neurons expressing steroidogenic factor 1, thereby inactivating a conditional Lepr allele specifically in steroidogenic factor 1 neurons of the VMH. These knockout (KO) mice, designated Lepr KO(VMH), exhibited obesity, particularly when challenged with a high-fat diet. On a low-fat diet, Lepr KO(VMH) mice exhibited significantly increased adipose mass even when their weights were comparable to wild-type littermates. Furthermore, these mice exhibited a metabolic syndrome including hepatic steatosis, dyslipidemia, and hyperleptinemia. Lepr KO(VMH) mice were hyperinsulinemic from the age of weaning and eventually developed overt glucose intolerance. These data define nonredundant roles of the Lepr in VMH neurons in energy homeostasis and provide a model system for studying other actions of leptin in the VMH.


Asunto(s)
Adiposidad/genética , Síndrome Metabólico/genética , Receptores de Leptina/genética , Núcleo Hipotalámico Ventromedial/metabolismo , Animales , Composición Corporal/genética , Peso Corporal/genética , Ingestión de Alimentos/genética , Sistema Endocrino/fisiología , Metabolismo Energético/genética , Femenino , Glucosa/metabolismo , Homeostasis/genética , Integrasas/genética , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Especificidad de Órganos , Receptores de Leptina/metabolismo
17.
Mol Endocrinol ; 22(4): 951-64, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18187601

RESUMEN

The steroidogenic acute regulatory protein (StAR) stimulates the regulated production of steroid hormones in the adrenal cortex and gonads by facilitating the delivery of cholesterol to the inner mitochondrial membrane. To explore key aspects of StAR function within bona fide steroidogenic cells, we used a transgenic mouse model to explore the function of StAR proteins in vivo. We first validated this transgenic bacterial artificial chromosome reconstitution system by targeting enhanced green fluorescent protein to steroidogenic cells of the adrenal cortex and gonads. Thereafter, we targeted expression of either wild-type StAR (WT-StAR) or a mutated StAR protein lacking the mitochondrial targeting signal (N47-StAR). In the context of mice homozygous for a StAR knockout allele (StAR-/-), all StAR activity derived from the StAR transgenes, allowing us to examine the function of the proteins that they encode. The WT-StAR transgene consistently restored viability and steroidogenic function to StAR-/- mice. Although the N47-StAR protein was reportedly active in transfected COS cells and mitochondrial reconstitution experiments, the N47-StAR transgene rescued viability in only 40% of StAR-/- mice. Analysis of lipid deposits in the primary steroidogenic tissues revealed a hierarchy of StAR function provided by N47-StAR: florid lipid deposits were seen in the adrenal cortex and ovarian theca region, with milder deposits in the Leydig cells. Our results confirm the ability of StAR lacking its mitochondrial targeting signal to perform some essential functions in vivo but also demonstrate important functional defects that differ from in vitro studies obtained in nonsteroidogenic cells.


Asunto(s)
Cromosomas Artificiales Bacterianos/genética , Mitocondrias/metabolismo , Fosfoproteínas/fisiología , Glándulas Suprarrenales/metabolismo , Hormona Adrenocorticotrópica/sangre , Animales , Southern Blotting , Corticosterona/sangre , Femenino , Técnicas de Transferencia de Gen , Gónadas/metabolismo , Immunoblotting , Masculino , Ratones , Ratones Transgénicos , Modelos Genéticos , Ovario/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transporte de Proteínas/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testículo/metabolismo
18.
J Biol Chem ; 283(13): 8453-61, 2008 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-18222920

RESUMEN

Hexose-6-phosphate dehydrogenase (H6PD) is the initial component of a pentose phosphate pathway inside the endoplasmic reticulum (ER) that generates NADPH for ER enzymes. In liver H6PD is required for the 11-oxoreductase activity of 11beta-hydroxysteroid dehydrogenase type 1, which converts inactive 11-oxo-glucocorticoids to their active 11-hydroxyl counterparts; consequently, H6PD null mice are relatively insensitive to glucocorticoids, exhibiting fasting hypoglycemia, increased insulin sensitivity despite elevated circulating levels of corticosterone, and increased basal and insulin-stimulated glucose uptake in muscles normally enriched in type II (fast) fibers, which have increased glycogen content. Here, we show that H6PD null mice develop a severe skeletal myopathy characterized by switching of type II to type I (slow) fibers. Running wheel activity and electrically stimulated force generation in isolated skeletal muscle are both markedly reduced. Affected muscles have normal sarcomeric structure at the electron microscopy level but contain large intrafibrillar membranous vacuoles and abnormal triads indicative of defects in structure and function of the sarcoplasmic reticulum (SR). SR proteins involved in calcium metabolism, including the sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA), calreticulin, and calsequestrin, show dysregulated expression. Microarray analysis and real-time PCR demonstrate overexpression of genes encoding proteins in the unfolded protein response pathway. We propose that the absence of H6PD induces a progressive myopathy by altering the SR redox state, thereby impairing protein folding and activating the unfolded protein response pathway. These studies thus define a novel metabolic pathway that links ER stress to skeletal muscle integrity and function.


Asunto(s)
Deshidrogenasas de Carbohidratos/deficiencia , Eliminación de Gen , Enfermedades Musculares/enzimología , Pliegue de Proteína , Transducción de Señal , Animales , Calcineurina/metabolismo , Deshidrogenasas de Carbohidratos/genética , Deshidrogenasas de Carbohidratos/metabolismo , Retículo Endoplásmico/enzimología , Activación Enzimática , Glucógeno/metabolismo , Ratones , Microscopía Electrónica , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Mutación/genética , NADP/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética
19.
Proc Natl Acad Sci U S A ; 104(42): 16558-63, 2007 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-17940049

RESUMEN

Targeted mutagenesis of Fgf9 in mice causes male-to-female sex reversal. Among the four FGF receptors, FGFR2 showed two highly specific patterns based on antibody staining, suggesting that it might be the receptor-mediating FGF9 signaling in the gonad. FGFR2 was detected at the plasma membrane in proliferating coelomic epithelial cells and in the nucleus in Sertoli progenitor cells. This expression pattern suggested that Fgfr2 might play more than one role in testis development. To test the hypothesis that Fgfr2 is required for male sex determination, we crossed mice carrying a floxed allele of Fgfr2 with two different Cre lines to induce a temporal or cell-specific deletion of this receptor. Results show that deletion of Fgfr2 in embryonic gonads phenocopies deletion of Fgf9 and leads to male-to-female sex reversal. Using these two Cre lines, we provide the first genetic evidence that Fgfr2 plays distinct roles in proliferation and Sertoli cell differentiation during testis development.


Asunto(s)
Diferenciación Celular/genética , Morfogénesis/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Células de Sertoli/citología , Procesos de Determinación del Sexo , Testículo/embriología , Animales , Proliferación Celular , Eliminación de Gen , Expresión Génica , Proteínas del Grupo de Alta Movilidad/genética , Integrasas/genética , Masculino , Ratones , Ratones Transgénicos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Factor de Transcripción SOX9 , Testículo/citología , Testículo/metabolismo , Factores de Transcripción/genética
20.
Eur J Endocrinol ; 157(2): 233-8, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17656604

RESUMEN

OBJECTIVE: Steroidogenic factor 1 (SF1), officially designated NR5A1, is a nuclear receptor that plays key roles in endocrine development and function. Previous reports of human SF1 mutations revealed a spectrum of phenotypes affecting adrenal function and/or gonadal development and sex differentiation. We present the clinical phenotype and functional effects of a novel SF1 mutation. PATIENT: The patient is a 22-year-old 46, XY Japanese patient who presented with dysgenetic testes, atrophic vasa deferentia and epididymides, lack of Müllerian structures, and clitoromegaly. Endocrine studies revealed normal adrenal function. RESULTS: Analysis of the SF1 gene revealed compound heterozygosity for a previously described p.G146A polymorphism and a novel missense mutation (p.R84C) in the accessory DNA-binding domain. The father carried the p.G146A polymorphism and the mother had the p.R84C mutation; both were clinically and reproductively normal. Functional studies demonstrated that the p.R84C SF1 had normal nuclear localization but decreased DNA-binding affinity and transcriptional activity compared with wild-type SF1; it did not exhibit any dominant negative activity. CONCLUSIONS: These results describe the human phenotype that results from compound heterozygosity of the p.G146A polymorphism and a novel p.R84C mutation of SF1, thereby extending the spectrum of human SF1 mutations that impair testis development and sex differentiation in a sex-limited manner while preserving normal adrenal function.


Asunto(s)
Proteínas de Unión al ADN/genética , Disgenesia Gonadal 46 XY/genética , Proteínas de Homeodominio/genética , Receptores Citoplasmáticos y Nucleares/genética , Factores de Transcripción/genética , Insuficiencia Suprarrenal/genética , Insuficiencia Suprarrenal/fisiopatología , Adulto , Células Cultivadas , Análisis Mutacional de ADN , Proteínas de Unión al ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Femenino , Técnica del Anticuerpo Fluorescente , Disgenesia Gonadal 46 XY/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Luciferasas/genética , Mutación/fisiología , Plásmidos/genética , Polimorfismo Genético/genética , Polimorfismo Genético/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Factor Esteroidogénico 1 , Receptores alfa de Hormona Tiroidea/genética , Factores de Transcripción/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA