Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Biol Reprod ; 88(6): 152, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23677985

RESUMEN

The type 1-cannabinoid receptor, CNR1, regulates differentiation of spermatids. Indeed, we have recently reported that the genetic inactivation of Cnr1 in mice influenced chromatin remodeling of spermatids, by reducing histone displacement and then sperm chromatin quality indices (chromatin condensation and DNA integrity). Herein, we have studied, at both central and testicular levels, the molecular signals potentially involved in histone displacement. In particular, investigation of the neuroendocrine axis involved in estrogen production demonstrated down-regulation of the axis supporting FSH/estrogen secretion in Cnr1-knockout male mice. Conversely, Cnr1-knockout male mice treated with 17beta-estradiol showed a weak increase of pituitary Fsh-beta subunit mRNA levels and a rescue of sperm chromatin quality indices demonstrating that estrogens, possibly in combination with FSH secretion, play an important role in regulating chromatin remodeling of spermatids.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Cromatina/metabolismo , Estradiol/sangre , Receptor Cannabinoide CB1/metabolismo , Espermátides/metabolismo , Animales , Aromatasa/genética , Aromatasa/metabolismo , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Estradiol/farmacología , Hormona Folículo Estimulante de Subunidad beta/genética , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Masculino , Ratones , Ratones Noqueados , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Receptor Cannabinoide CB1/genética , Receptores de HFE/genética , Receptores de HFE/metabolismo , Receptores LHRH/genética , Receptores LHRH/metabolismo , Espermátides/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Espermatogénesis/fisiología , Testículo/efectos de los fármacos , Testículo/metabolismo
2.
Biochim Biophys Acta ; 1800(6): 611-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20188144

RESUMEN

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is associated with obesity, insulin resistance and hepatic steatosis. Non-alcoholic steatohepatitis (NASH) is a serious consequence of NAFLD where chronic tissue damage and inflammation result in fibrosis which may progress to cirrhosis. Transforming growth factor beta1 (TGFbeta1), proinflammatory cytokines and oxidative stress are thought to play crucial roles in the pathogenesis of these conditions. The contributions of individual liver cell types to fibrogenesis remain controversial and the influence of selenium status has not been investigated. METHODS: In this study we have used a cell culture model of fat-loading using oleate-treated human hepatoblastoma (C3A) cells to investigate how fat-loading and selenium status might influence the production of collagen in response to TGFbeta1. The secretion of inflammatory cytokines was also investigated, together with the epithelial character of the treated cells. RESULTS: We found that in response to treatment with TGFbeta1, C3A cells produced mRNA encoding the pro-alphaI chain of procollagen I, secreted procollagen I peptide, up-regulated production of the proinflammatory cytokine interleukin-8 (IL-8) and the mesenchymal marker vimentin, and down-regulated albumin production. Most of these responses were considerably enhanced when cells were fat-loaded with oleate and were attenuated by selenium addition at a dose that optimised the expression of thioredoxin reductase and glutathione peroxidase. CONCLUSIONS: Our data establish that both fat-loading and suboptimal selenium status enhance collagen and IL-8 production by C3A hepatocytes in response to TGFbeta1, possibly as part of an epithelial to mesenchymal transition. GENERAL SIGNIFICANCE: These findings suggest that the hepatocyte may be an important contributor to the pathogenesis of fibrosis associated with NAFLD.


Asunto(s)
Grasas/análisis , Hepatoblastoma/metabolismo , Interleucina-8/biosíntesis , Neoplasias Hepáticas/metabolismo , Procolágeno/biosíntesis , Selenio/administración & dosificación , Factor de Crecimiento Transformador beta1/farmacología , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Células Epiteliales/citología , Hepatoblastoma/patología , Humanos , Neoplasias Hepáticas/patología , Mesodermo/citología , Reacción en Cadena de la Polimerasa , Selenio/farmacología
3.
Am J Physiol Gastrointest Liver Physiol ; 298(6): G884-95, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20338923

RESUMEN

Cyclin D1 is a cell cycle control protein that plays an important role in regenerating liver and many types of cancer. Previous reports have shown that cyclin D1 can directly enhance estrogen receptor activity and inhibit androgen receptor activity in a ligand-independent manner and thus may play an important role in hormone-responsive malignancies. In this study, we examine a distinct mechanism by which cyclin D1 regulates sex steroid signaling, via altered metabolism of these hormones at the tissue and cellular level. In male mouse liver, ectopic expression of cyclin D1 regulated genes involved in the synthesis and degradation of sex steroid hormones in a pattern that would predict increased estrogen and decreased androgen levels. Indeed, hepatic expression of cyclin D1 led to increased serum estradiol levels, increased estrogen-responsive gene expression, and decreased androgen-responsive gene expression. Cyclin D1 also regulated the activity of several key enzymatic reactions in the liver, including increased oxidation of testosterone to androstenedione and decreased conversion of estradiol to estrone. Similar findings were seen in the setting of physiological cyclin D1 expression in regenerating liver. Knockdown of cyclin D1 in HuH7 cells produced reciprocal changes in steroid metabolism genes compared with cyclin D1 overexpression in mouse liver. In conclusion, these studies establish a novel link between the cell cycle machinery and sex steroid metabolism and provide a distinct mechanism by which cyclin D1 may regulate hormone signaling. Furthermore, these results suggest that increased cyclin D1 expression, which occurs in liver regeneration and liver diseases, may contribute to the feminization seen in these settings.


Asunto(s)
Andrógenos/biosíntesis , Ciclina D1/metabolismo , Estrógenos/biosíntesis , Hígado/metabolismo , Animales , Inhibidores de la Aromatasa/farmacología , Línea Celular Tumoral , Ciclina D1/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Letrozol , Regeneración Hepática , Masculino , Ratones , Ratones Endogámicos BALB C , Nitrilos/farmacología , Triazoles/farmacología
4.
Endocr J ; 57(7): 651-6, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20467160

RESUMEN

Estrogen-secreting adrenal cancers are extremely rare, with feminizing symptoms attributed to aromatase expression in the adrenal tumor. We describe a case of hypogonadotropic hypogonadism as a consequence of aberrant aromatase expression in a patient with adrenocortical adenocarcinoma. A 54 year-old man presented with a two month history of gynecomastia and reduced libido. Endocrine biochemistry at presentation showed hypogonadotropic hypogonadism (LH 2.4 U/L, FSH <1.0 IU/L, testosterone 2.8 nmol/L) with increased serum estrone (E(1), 821 pmol/L) and estradiol (E(2), 797 pmol/L) and subclinical ACTH-independent hypercortisolism (serum cortisol post 1mg overnight dexamethasone suppression test, 291 nmol/L). A right adrenal mass was identified on CT scanning and the patient underwent an open adrenalectomy. Post-operative evaluation showed normalization of serum levels of E(1) (95 pmol/L), E(2 )(109 pmol/L), testosterone (11.4 nmol/L), LH (4.1 U/L) and FSH (5.9 IU/L), and of cortisol dynamics. Immunohistochemistry of the adrenal cancer confirmed aberrant expression of aromatase in most, although not all, carcinoma cells. Transcripts associated with utilization of promoters II, I.1 and I.3 were prominently represented in the tumor aromatase mRNA. This case highlights that clinical features of feminizing adrenocortical carcinomas can be secondary to estrogen production by aberrantly transcribed and translated aromatase within the tumor. Even in males, gonadotropin secretion is subject to predominantly estrogen-mediated feedback-inhibition. The diagnosis of adrenocortical adenocarcinoma should be considered in men presenting with low testosterone and gonadotropins, particularly in the presence of feminizing features.


Asunto(s)
Adenocarcinoma/genética , Neoplasias de la Corteza Suprarrenal/genética , Aromatasa/genética , Hipogonadismo/genética , Adenocarcinoma/complicaciones , Adenocarcinoma/metabolismo , Neoplasias de la Corteza Suprarrenal/complicaciones , Neoplasias de la Corteza Suprarrenal/metabolismo , Adulto , Edad de Inicio , Aromatasa/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Predisposición Genética a la Enfermedad , Humanos , Hipogonadismo/epidemiología , Hipogonadismo/etiología , Hipogonadismo/metabolismo , Masculino , Persona de Mediana Edad
5.
J Clin Invest ; 116(4): 953-60, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16585961

RESUMEN

In humans, sexual differentiation of the external genitalia is established at 7-12 weeks post conception (wpc). During this period, maintaining the appropriate intrauterine hormone environment is critical. In contrast to other species, this regulation extends to the human fetal adrenal cortex, as evidenced by the virilization that is associated with various forms of congenital adrenal hyperplasia. The mechanism underlying these clinical findings has remained elusive. Here we show that the human fetal adrenal cortex synthesized cortisol much earlier than previously documented, an effect associated with transient expression of the orphan nuclear receptor nerve growth factor IB-like (NGFI-B) and its regulatory target, the steroidogenic enzyme type 2 3beta-hydroxysteroid dehydrogenase (HSD3B2). This cortisol biosynthesis was maximal at 8-9 wpc under the regulation of ACTH. Negative feedback was apparent at the anterior pituitary corticotrophs. ACTH also stimulated the adrenal gland to secrete androstenedione and testosterone. In concert, these data promote a distinctive mechanism for normal human development whereby cortisol production, determined by transient NGFI-B and HSD3B2 expression, provides feedback at the anterior pituitary to modulate androgen biosynthesis and safeguard normal female sexual differentiation.


Asunto(s)
Hidrocortisona/biosíntesis , Diferenciación Sexual , Desarrollo Sexual , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Corteza Suprarrenal/embriología , Corteza Suprarrenal/metabolismo , Andrógenos/biosíntesis , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación de la Expresión Génica , Edad Gestacional , Humanos , Hidrocortisona/metabolismo , Modelos Biológicos , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Adenohipófisis/embriología , Adenohipófisis/crecimiento & desarrollo , Adenohipófisis/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Desarrollo Sexual/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Mol Hum Reprod ; 15(6): 379-92, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19414525

RESUMEN

The human ovarian surface epithelium (hOSE) is a squamous-to-cuboidal layer that surrounds the ovary. hOSE undergoes injury and repair cycles as a result of ovulation-induced inflammation, an event relevant to the development of epithelial ovarian cancer (EOC). Locally produced steroids mediate the response to inflammation. 3beta-Hydroxysteroid dehydrogenase (3beta-HSD) drives the intracrine generation of progestogens and androgens that potentially affect cell survival and proliferation. We therefore investigated the regulation of 3beta-HSD along with downstream steroid signalling in hOSE. Double immunofluorescence of cultured primary hOSE cells confirmed the expression of 3beta-HSD protein Interleukin (IL). IL-1alpha treatment of primary cells to mimic ovulation-associated inflammation suppressed 3beta-HSD1 expression and stimulated 3beta-HSD2 mRNA (P < 0.001), without affecting total 3beta-HSD protein and activity or androgen or progesterone receptor (PR) mRNA levels. Conversely, IL-4 as a proxy for a post-ovulatory healing cytokine increased both 3beta-HSD transcripts, total protein and activity (P < 0.01). IL-4 also suppressed androgen receptor expression (P < 0.01) without affecting that of the PR, thereby potentially sustaining both progesterone biosynthesis and its underlying signalling in the ovarian surface. 3beta-HSD protein was immunodetectable in primary ascites of women who were diagnosed with EOC but both mRNA transcripts were diminished relative to normal cells (P < 0.05). Notably, this difference was countered by IL-4 treatment (P < 0.01). We conclude that stimulation by IL-4 could be physiologically relevant to post-ovulatory ovarian healing and suggest a novel therapeutic strategy for the activation of progesterone-associated apoptosis in ovarian cancer. Also, our results suggest an attenuation of 3beta-HSD expression in EOC although further studies are required for confirmation.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/metabolismo , Citocinas/farmacología , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Ovario/efectos de los fármacos , Ovario/metabolismo , 17-Hidroxiesteroide Deshidrogenasas/genética , Adulto , Línea Celular , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Técnicas In Vitro , Interleucina-1alfa/farmacología , Interleucina-4/farmacología , Persona de Mediana Edad , Neoplasias Ováricas/genética , Ovario/citología , Ovario/patología , Adulto Joven
7.
Mol Cell Endocrinol ; 301(1-2): 65-73, 2009 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-18778748

RESUMEN

Ovulation-associated inflammation with accompanied cytokines and reproductive hormones impact upon the human ovarian surface epithelium (hOSE) and probably have a role in the aetiology of ovarian cancer. Progesterone and progestin-related events, i.e. pregnancy and oral contraception, protect from the disease. We have investigated the pre-receptor metabolism of progesterone in primary hOSE cells and an immortalised hOSE cell line, OSE-C2, focusing on transcriptional regulation of 3beta-hydroxysteroid dehydrogenase (3beta-HSD) by inflammatory, anti-inflammatory and apoptotic factors. In hOSE cells, we show that anti-inflammatory effects of IL-1alpha and IL-4 on 3beta-HSD2 mRNA involve a p38 MAPK signalling pathway, whereas pro-inflammatory response of IL-1alpha to 3beta-HSD1 mRNA involves a NF-kappaB inflammatory pathway. In OSE-C2 cells, retinoic acid and transforming growth factor-beta1 massively induce 3beta-HSD1 mRNA levels. In conclusion, we elaborate several mechanisms for intracrine formation of progesterone in hOSE that could contribute in the development of novel strategies to prevent, diagnose and/or treat ovarian cancer.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/metabolismo , Epitelio/enzimología , Ovario/enzimología , Receptores de Esteroides/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/genética , Adulto , Línea Celular , Epitelio/efectos de los fármacos , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/enzimología , Interleucina-1alfa/farmacología , Interleucina-4/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/metabolismo , Ovario/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Tretinoina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Mol Cell Endocrinol ; 299(2): 153-62, 2009 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-19059462

RESUMEN

Recent studies have shown that the adrenal cortex expresses high levels of farnesoid X receptor (FXR), but its function remains unknown. Herein, using microarray technology, we tried to identify candidate FXR targeting genes in the adrenal glands, and showed that FXR regulated 3beta-hydroxysteroid dehydrogenase type 2 (HSD3B2) expression in human adrenocortical cells. We further demonstrated that FXR stimulated HSD3B2 promoter activity and have defined the cis-element responsible for FXR regulation of HSD3B2 transcription. Transfection of H295R adrenocortical cells with FXR expression vector effectively increased FXR expression levels and additional treatment with chenodeoxycholic acid (CDCA) caused a 25-fold increase in the mRNA for organic solute transporter alpha (OSTalpha), a known FXR target gene. HSD3B2 mRNA levels also increased following CDCA treatment in a concentration-dependent manner. Cells transfected with a HSD3B2 promoter construct and FXR expression vector responded to CDCA with a 20-fold increase in reporter activity compared to control. Analysis of constructs containing sequential deletions of the HSD3B2 promoter suggested a putative regulatory element between -166 and -101. Mutation of an inverted repeat between -137 and -124 completely blocked CDCA/FXR induced reporter activity. Chromatin immunoprecipitation assays further confirmed the presence of a FXR response element in the HSD3B2 promoter. In view of the emerging role of FXR agonists as therapeutic treatment of diabetes and certain liver diseases, the effects of such agonists on other FXR expressing tissues should be considered. Our findings suggest that in human adrenal cells, FXR increases transcription and expression of HSD3B2. Alterations in this enzyme would influence the capacity of the adrenal gland to produce corticosteroids.


Asunto(s)
Glándulas Suprarrenales/citología , Glándulas Suprarrenales/enzimología , Proteínas de Unión al ADN/metabolismo , Progesterona Reductasa/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Secuencia de Bases , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Inmunohistoquímica , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Datos de Secuencia Molecular , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Elementos de Respuesta/genética , Esteroides/biosíntesis , Factores de Transcripción/genética
9.
Mol Cell Endocrinol ; 300(1-2): 115-20, 2009 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-19026713

RESUMEN

Adrenocortical carcinoma is an uncommon malignancy and feminizing symptoms secondary to adrenal estrogen-secretion are extremely rare. The direct secretion of estradiol by adrenocortical tumors requires, in addition to the expression of aromatase (CYP19), the expression of one or more of the reductive 17beta-hydroxysteroid dehydrogenases. The expression of CYP19 transcripts and protein were markedly induced in the H295 adrenocortical carcinoma cell line after treatment with either forskolin or vasoactive intestinal peptide (VIP). Western immunoblotting demonstrated a marked induction of the CYP19 protein of characteristic size after only a short (6h) treatment period with VIP or forskolin. The CYP19 mRNA transcripts were derived from both promoters PII (Ic) and I.3 (Id) after treatment with both agents. The reductive type 5 17beta-hydroxysteroid dehydrogenase (AKR1C3) was also constitutively expressed in the H295 cells but neither its mRNA transcript nor protein levels were altered after forskolin or VIP treatment. Western immunoblotting of an estrogen-secreting adrenal carcinoma revealed notable levels of both aromatase and AKR1C3 expression while an aldosterone-producing adrenal adenoma lacked aromatase expression and showed a reduced level of AKR1C3 expression. Immunohistochemistry of the carcinoma-bearing adrenal revealed localization of AKR1C3 not only in the tumor but also principally in the zona reticularis of the normal adrenal tissue. Adrenal aromatase and AKR1C3 expression therefore appear to be features of adrenocortical malignancies that are associated with biosynthesis of active estrogen.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/metabolismo , Línea Celular Tumoral , Estrógenos/biosíntesis , 17-Hidroxiesteroide Deshidrogenasas/genética , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , 3-Hidroxiesteroide Deshidrogenasas , Neoplasias de la Corteza Suprarrenal/patología , Adulto , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas , Aromatasa/genética , Aromatasa/metabolismo , Colforsina/metabolismo , Citocromo P-450 CYP11B2/genética , Citocromo P-450 CYP11B2/metabolismo , Femenino , Humanos , Hidroxiprostaglandina Deshidrogenasas , Progesterona Reductasa/genética , Progesterona Reductasa/metabolismo , Regiones Promotoras Genéticas , Esteroide 11-beta-Hidroxilasa/genética , Esteroide 11-beta-Hidroxilasa/metabolismo , Esteroide 17-alfa-Hidroxilasa/genética , Esteroide 17-alfa-Hidroxilasa/metabolismo , Péptido Intestinal Vasoactivo/metabolismo
10.
Reprod Fertil Dev ; 21(7): 827-39, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19698287

RESUMEN

The objective of the present study was to characterise the expression and tissue distribution of steroid receptors (oestrogen receptor-alpha and -beta (ERalpha, ERbeta), androgen receptor (AR) and progesterone receptor (PR)) and steroidogenic enzymes (P450 aromatase (P450arom), 3beta-hydroxysteroid dehydrogenase (3beta-HSD) and steroidogenic acute regulatory protein (StAR)) in letrozole-induced polycystic ovaries of rats. Changes in serum hormone levels, protein expression in whole ovaries by western blot analysis and protein localisation by immunohistochemistry were determined in female rats treated with the aromatase inhibitor letrozole and compared with controls in proestrous and diestrous rats. Increases in the serum LH, FSH and testosterone concentrations were observed in letrozole-treated rats whereas serum oestradiol and progesterone levels were reduced. Protein expression as analysed by western immunoblot was consistent with the immunohistochemical data. Letrozole treatment induced an increase in the expression of AR, StAR and 3beta-HSD and a decrease in ERbeta. ERalpha, PR and P450arom showed partial changes in relation to some cycle stages. These results indicate that cystogenesis in this experimental model is characterised by changes in steroid receptors and steroidogenic enzyme expression that may be essential to proper ovarian functioning and are in agreement with similar changes observed in women with PCOS.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/metabolismo , Aromatasa/metabolismo , Ovario/enzimología , Fosfoproteínas/metabolismo , Síndrome del Ovario Poliquístico/enzimología , Receptores de Esteroides/metabolismo , Animales , Western Blotting , Diestro , Modelos Animales de Enfermedad , Estradiol/sangre , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Hormona Folículo Estimulante/sangre , Inmunohistoquímica , Letrozol , Hormona Luteinizante/sangre , Nitrilos , Ovario/fisiopatología , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/fisiopatología , Proestro , Progesterona/sangre , Ratas , Ratas Wistar , Receptores Androgénicos/metabolismo , Receptores de Progesterona/metabolismo , Testosterona/sangre , Triazoles
11.
Sci Rep ; 9(1): 3250, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30824750

RESUMEN

The hypothalamic-pituitary-adrenal (HPA) axis regulates responses to internal and external stressors. Many patients diagnosed with conditions such as depression or anxiety also have hyperactivity of the HPA axis. Hyper-stimulation of the HPA axis results in sustained elevated levels of glucocorticoids which impair neuronal function and can ultimately result in a psychiatric disorder. Studies investigating Glucocorticoid Receptor (GR/NR3C1) in the brain have primarily focused on the forebrain, however in recent years, the hindbrain has become a region of interest for research into the development of anxiety and depression, though the role of GR signalling in the hindbrain remains poorly characterised. To determine the role of glucocorticoid signalling in the hindbrain we have developed a novel mouse model that specifically ablates hindbrain GR to ascertain its role in behaviour, HPA-axis regulation and adrenal structure. Our study highlights that ablation of GR in the hindbrain results in excessive barbering, obsessive compulsive digging and lack of cage exploration. These mice also develop kyphosis, elevated circulating corticosterone and severe adrenal cortex disruption. Together, this data demonstrates a role for hindbrain GR signalling in regulating stress-related behaviour and identifies a novel mouse model to allow further investigation into the pathways impacting stress and anxiety.


Asunto(s)
Receptores de Glucocorticoides/metabolismo , Rombencéfalo/metabolismo , Estrés Psicológico/metabolismo , Corteza Suprarrenal/patología , Animales , Conducta Animal , Peso Corporal , Modelos Animales de Enfermedad , Femenino , Cifosis/complicaciones , Cifosis/diagnóstico por imagen , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Tomografía de Emisión de Positrones , Recombinación Genética/genética , Rombencéfalo/diagnóstico por imagen , Estrés Psicológico/diagnóstico por imagen , Tomografía Computarizada por Rayos X
12.
Sci Rep ; 9(1): 10457, 2019 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-31320667

RESUMEN

Androgens are known to be an essential regulator of male health. Androgen receptor (AR) is widely expressed throughout the adrenal cortex, yet the wider role for androgen signalling in the adrenal remains underexplored. To investigate AR-dependent and AR-independent androgen signalling in the adrenal, we used a novel mouse model with a specific ablation of androgen receptor in the adrenal cortex with or without reduction of circulating androgen levels by castration. Our results describe AR expression in the human and mouse adrenal and highlight that the mouse is a viable model to investigate androgen signalling in the adrenal cortex. We show androgen signalling via AR is required for X-zone regression during puberty. Furthermore, cortex measurements define differences in X-zone morphology depending on whether circulating androgens or AR have been removed. We show androgens promote both cortical cell differentiation and apoptosis but are dispensable for the formation of the definitive cortex. Additionally, investigation of aged mice with AR ablation reveals severe cortex disruption, spindle cell hyperplasia and X-zone expansion. The data described herein demonstrates AR-signalling is required to facilitate X-zone regression, cell clearance and to protect against adrenal degeneration during ageing.


Asunto(s)
Corteza Suprarrenal/citología , Envejecimiento/fisiología , Andrógenos/farmacología , Sustancias Protectoras/farmacología , Receptores Androgénicos/fisiología , Corteza Suprarrenal/efectos de los fármacos , Corteza Suprarrenal/metabolismo , Envejecimiento/efectos de los fármacos , Animales , Apoptosis , Castración , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
13.
Int J Androl ; 31(2): 103-11, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17877717

RESUMEN

A testicular dysgenesis-like syndrome is induced in rats by fetal exposure to di(n-butyl) phthalate (DBP). A key feature of this is the formation of focal dysgenetic areas comprising malformed seminiferous cords/tubules and intratubular Leydig cells (ITLC), but how and why these arise remains unclear. The present study has used combinations of cell-specific markers and immunohistochemistry to address this. The results show that focal dysgenetic areas and ITLC first appear postnatally at 4-10 days of age, but this only occurs in treatment groups in which formation of fetal Leydig cell aggregation is induced between e17.5 and e21.5. Extreme variability in the formation and size of the Leydig cell aggregates probably accounts for the equally extreme variation in occurrence and size of focal dysgenetic areas postnatally. DBP-induced fetal Leydig cell aggregation traps Sertoli and other cells within the aggregates, but it is unclear why this happens nor why cords fail to form prenatally in these cell mixtures but do elsewhere in the fetal testis. The present studies show that differentiation of the fetal Leydig cells is drastically delayed at e15.5 after DBP exposure, which may be indicative of a wider delay in testis cell development and organisation, and this might account for some of the unexplained findings.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades Testiculares/patología , Testículo/crecimiento & desarrollo , Animales , Inmunohistoquímica , Masculino , Ratas , Ratas Wistar
14.
Neuromolecular Med ; 9(3): 230-48, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17914181

RESUMEN

The primary function of gonadotropin-releasing hormone (GnRH) is the regulation of pituitary gonadotropin hormone gene transcription, biosynthesis and release. These effects are mediated through intracellular mobilization of Ca2+ and activation of PKC isoforms and MAP kinases. We show here that DAN (differential screening-selected gene aberrative in neuroblastoma) which is a secreted bone morphogenic protein (BMP) antagonist belonging to the TGFbeta protein superfamily, is controlled by GnRH in murine gonadotrope cells. Acute GnRH stimulation induced a rapid, 27-fold, elevation of DAN mRNA, accompanied by an approximate 3-fold increase in the amount of mature DAN glycoprotein in the cell cytoplasm and in DAN secretion into the culture medium. Incubation of L beta T2 cells in DAN-containing medium altered the levels of a number of cellular proteins. Two of these were identified as the steroidogenic acute regulatory protein (StAR) and the actin-related protein 2/3 complex subunits 2 (p34-ARC) which are primarily involved in steroidogenesis and cytoskeleton remodelling, respectively. DAN caused an approximate 2-fold specific elevation in the cytoplasmic levels of both these proteins in L beta T2 cells. We further tested the effects of DAN on classical GnRH effects viz. gonadotropin and GnRH receptor gene expression. Co-transfection of L beta T2 cells with DAN and gonadotropin subunit promoter luciferase reporter genes had no effect on GnRH stimulation of alpha GSU and LH beta or on the additive GnRH and activin induction of FSH beta subunit transcription. However, co-transfection of DAN markedly inhibited the synergistic activation of GnRH and activin on GnRH receptor gene expression thus implicating DAN as a novel autocrine/paracrine factor that modulates GnRH function in pituitary gonadotropes.


Asunto(s)
Hormona Liberadora de Gonadotropina/fisiología , Proteínas/metabolismo , Receptores LHRH/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/biosíntesis , Activinas/metabolismo , Secuencia de Aminoácidos , Animales , Comunicación Autocrina , Células COS , Proteínas de Ciclo Celular , Línea Celular , Chlorocebus aethiops , Citoplasma/metabolismo , Regulación de la Expresión Génica , Hormona Liberadora de Gonadotropina/farmacología , Ratones , Datos de Secuencia Molecular , Comunicación Paracrina , Fosfoproteínas/biosíntesis , Regiones Promotoras Genéticas , Subunidades de Proteína/biosíntesis , ARN Mensajero/metabolismo , Receptores LHRH/genética , Transcripción Genética
15.
J Endocrinol ; 192(2): 405-19, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17283241

RESUMEN

Transforming growth factor (TGF) beta1 facilitates FSH-induced differentiation of rat ovarian granulosa cells. The signaling crosstalk between follicle stimulating hormone (FSH) and TGFbeta receptors remains unclear. This study was to investigate the interplay of cAMP/protein kinase A (PKA) and phosphatidylinositol-3-kinase (PI3K) signaling including mammalian target of rapamycin (mTOR)C1 dependence in FSH- and TGFbeta1-stimulated steroidogenesis in rat granulosa cells. To achieve this aim, inhibitors of PKA (PKAI), PI3K (wortmannin), and mTORC1 (rapamycin) were employed. PKAI and wortmannin suppressions of the FSH-increased progesterone production were partly attributed to decreased level of 3beta-HSD, and their suppression of the FSH plus TGFbeta1 effect was attributed to the reduction of all the three key players, steroidogenic acute regulatory (StAR) protein, P450scc, and 3beta-HSD. Further, FSH activated the PI3K pathway including increased integrin-linked kinase (ILK) activity and phosphorylation of Akt(S473), mTOR(S2481), S6K(T389), and transcription factors particularly FoxO1(S256) and FoxO3a(S253), which were reduced by wortmannin treatment but not by PKAI. Interestingly, PKAI suppression of FSH-induced phosphorylation of cAMP regulatory element-binding protein (CREB(S133)) disappeared in the presence of wortmannin, suggesting that wortmannin may affect intracellular compartmentalization of signaling molecule(s). In addition, TGFbeta1 had no effect on FSH-activated CREB and PI3K signaling mediators. We further found that rapamycin reduced the TGFbeta1-enhancing effect of FSH-stimulated steroidogenesis, yet it exhibited no effect on FSH action. Surprisingly, rapamycin displayed a suppressive effect at concentrations that had no effect on mTORC1 activity. Together, this study demonstrates a delicate interplay between cAMP/PKA and PI3K signaling in FSH and TGFbeta1 regulation of steroidogenesis in rat granulosa cells. Furthermore, we demonstrate for the first time that TGFbeta1 acts in a rapamycin-hypersensitive and mTORC1-independent manner in augmenting FSH-stimulated steroidogenesis in rat granulosa cells.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Células de la Granulosa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Progesterona/biosíntesis , Factor de Crecimiento Transformador beta1/farmacología , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Androstadienos/farmacología , Animales , Células Cultivadas , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Hormona Folículo Estimulante/farmacología , Células de la Granulosa/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosfoproteínas/metabolismo , Fosforilación , Progesterona/análisis , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Sirolimus/metabolismo , Sirolimus/farmacología , Estimulación Química , Serina-Treonina Quinasas TOR , Factor de Crecimiento Transformador beta1/metabolismo , Wortmanina
16.
J Biotechnol ; 129(4): 635-44, 2007 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-17386955

RESUMEN

Human cytochrome P45017alpha (CYP17), present in mammalian adrenal and gonadal tissues, catalyses both steroid 17-hydroxylation and C17,20 lyase reactions, producing intermediates for the glucocorticoid and androgenic pathways, respectively. The characterisation of this complex enzyme was initially hampered due to low level in vivo expression of CYP17. Heterologous expression systems have contributed greatly to our current knowledge of CYP17's dual catalytic activity. However, due to the hydrophobic nature of this membrane-bound protein, primarily truncated and modified forms of CYP17 are currently being expressed heterologously. Although the N-terminally modified enzyme has been well characterised, protein structure and function studies still necessitate the expression of unmodified, wild-type CYP17. We report here the expression of a catalytically active, unmodified human CYP17 in the industrial methylotrophic yeast, Pichia pastoris. A typical P450 carbon monoxide difference spectrum, with an absorption maximum at 448nm and a substrate-induced type I spectrum were recorded using a detergent-solubilised cellular fraction containing CYP17. The expressed enzyme catalysed the conversion of progesterone to 17-hydroxyprogesterone as well as 16-hydroxyprogesterone, a product unique to human and chimpanzee CYP17. This is the first report showing the heterologous expression of a fully functional human steroidogenic cytochrome P450 enzyme in P. pastoris.


Asunto(s)
Pichia/enzimología , Esteroide 17-alfa-Hidroxilasa/genética , Humanos , Cinética , Progesterona/metabolismo , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Solubilidad , Esteroide 17-alfa-Hidroxilasa/aislamiento & purificación , Esteroide 17-alfa-Hidroxilasa/metabolismo , Transfección
17.
PLoS One ; 12(8): e0183013, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28800626

RESUMEN

BACKGROUND: Abdominal surgery and disease cause persistent abdominal adhesions, pelvic pain, infertility and occasionally, bowel obstruction. Current treatments are ineffective and the aetiology is unclear, although excessive collagen deposition is a consistent feature. Lysyl oxidase (Lox) is a key enzyme required for crosslinking and deposition of insoluble collagen, so we investigated whether targeting Lox might be an approach to reduce abdominal adhesions. METHODS: Female C57Bl/6 mice were treated intraperitoneally with multiwalled carbon nanotubes (NT) to induce fibrosis, together with chemical (ß-aminoproprionitrile-BAPN) or miRNA Lox inhibitors, progesterone or dexamethasone. Fibrotic lesions on the diaphragm, and expression of fibrosis-related genes in abdominal wall peritoneal mesothelial cells (PMC) were measured. Effects of BAPN and dexamethasone on collagen fibre alignment were observed by TEM. Isolated PMC were cultured with interleukin-1 alpha (IL-1α) and progesterone to determine effects on Lox mRNA in vitro. RESULTS: NT-induced fibrosis and collagen deposition on the diaphragm was ameliorated by BAPN, Lox miRNA, or steroids. BAPN and dexamethasone disrupted collagen fibres. NT increased PMC Lox, Col1a1, Col3a1 and Bmp1 mRNA, which was inhibited by steroids. Progesterone significantly inhibited IL-1α induced Lox expression by PMC in vitro. CONCLUSION: Our results provide proof-of-concept that targeting peritoneal Lox could be an effective approach in ameliorating fibrosis and adhesion development.


Asunto(s)
Aminopropionitrilo/farmacología , Colágeno/antagonistas & inhibidores , Dexametasona/farmacología , Proteínas de la Matriz Extracelular/antagonistas & inhibidores , Terapia Molecular Dirigida , Fibrosis Peritoneal/prevención & control , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Adherencias Tisulares/prevención & control , Cavidad Abdominal/cirugía , Animales , Colágeno/genética , Colágeno/metabolismo , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Expresión Génica , Humanos , Interleucina-1alfa/farmacología , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Nanotubos de Carbono/toxicidad , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/genética , Fibrosis Peritoneal/patología , Cultivo Primario de Células , Progesterona/farmacología , Proteína-Lisina 6-Oxidasa/genética , Proteína-Lisina 6-Oxidasa/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Adherencias Tisulares/inducido químicamente , Adherencias Tisulares/genética , Adherencias Tisulares/patología
18.
Mol Cell Endocrinol ; 248(1-2): 72-8, 2006 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-16406280

RESUMEN

Key reproductive events, such as menstruation and implantation, are considered to be inflammatory processes and glucocorticoids act as anti-inflammatory agents. The balance of expression of types 1 and 2 11beta-hydroxysteroid dehydrogenases (11betaHSD) controls the availability of cortisol to bind to the glucocorticoid receptor (GR) and mineralocorticoid receptor (MR). Expression profiles of glucocorticoid-metabolising enzymes and their cognate receptors have been characterized in the reproductive tract. We propose that factors that peripherally promote glucocorticoid action are part of an anti-inflammatory response to tissue remodelling in human endometrium. Protein and mRNA expression in endometrium were investigated using immunohistochemistry and quantitative real-time PCR. There was up-regulated expression of 11betaHSD-1 at menstruation and in first trimester decidua. 11BetaHSD-2 and GR were expressed across the cycle. The MR expression pattern across the cycle and in decidua implies progesterone may also play a regulatory role. The precise roles and interactions of these proteins require further investigation.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Endometrio/enzimología , Menstruación/metabolismo , Primer Trimestre del Embarazo/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/análisis , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/análisis , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , Femenino , Humanos , Menstruación/genética , Embarazo , Primer Trimestre del Embarazo/genética , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Regulación hacia Arriba
19.
Endocrinology ; 146(9): 4117-26, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15919750

RESUMEN

It is established that androgens and unidentified Sertoli cell (SC)-derived factors can influence the development of adult Leydig cells (LC) in rodents, but the mechanisms are unclear. We evaluated adult LC development and function in SC-selective androgen receptor (AR) knockout (SCARKO) and complete AR knockout (ARKO) mice. In controls, LC number increased 26-fold and LC size increased by approximately 2-fold between 12 and 140 d of age. LC number in SCARKOs was normal on d 12, but was reduced by more than 40% at later ages, although LC were larger and contained more lipid droplets and mitochondria than control LC by adulthood. ARKO LC number was reduced by up to 83% at all ages compared with controls, and LC size did not increase beyond d 12. Serum LH and testosterone levels and seminal vesicle weights were comparable in adult SCARKOs and controls, whereas LH levels were elevated 8-fold in ARKOs, although testosterone levels appeared normal. Immunohistochemistry and quantitative PCR for LC-specific markers indicated steroidogenic function per LC was probably increased in SCARKOs and reduced in ARKOs. In SCARKOs, insulin-like factor-3 and estrogen sulfotransferase (EST) mRNA expression were unchanged and increased 3-fold, respectively, compared with controls, whereas the expression of both was reduced more than 90% in ARKOs. Changes in EST expression, coupled with reduced platelet-derived growth factor-A expression, are potential causes of altered LC number and function in SCARKOs. These results show that loss of androgen action on SC has major consequences for LC development, and this could be mediated indirectly via platelet-derived growth factor-A and/or estrogens/EST.


Asunto(s)
Células Intersticiales del Testículo/fisiología , Células Intersticiales del Testículo/ultraestructura , Receptores Androgénicos/genética , Células de Sertoli/citología , Células de Sertoli/fisiología , Factores de Edad , Andrógenos/fisiología , Animales , Recuento de Células , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Testículo/citología , Testículo/fisiología
20.
Trends Endocrinol Metab ; 13(6): 234-9, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12128283

RESUMEN

The human adrenal cortex produces aldosterone, cortisol and the so-called adrenal androgens, dehydroepiandrosterone (DHEA) and DHEA sulfate (DHEAS). Within the adult adrenal, the zona glomerulosa produces aldosterone, the zona fasciculata cortisol and the zona reticularis both DHEA and DHEAS. The processes regulating aldosterone and cortisol synthesis are well defined; however, the mechanisms regulating the production of DHEA(S) remain elusive. The emphasis of this review is based on increasing evidence that cytochrome b(5), DHEA sulfotransferase and 3 beta-hydroxysteroid dehydrogenase play crucial roles in regulating production of DHEA(S). Insight into the mechanisms that regulate the synthesis of these key components of DHEA(S) synthesis should provide important clues to the regulation of adrenal androgen biosynthesis.


Asunto(s)
Corteza Suprarrenal/metabolismo , Andrógenos/biosíntesis , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Citocromos b5/metabolismo , Deshidroepiandrosterona/biosíntesis , Sulfato de Deshidroepiandrosterona/metabolismo , Humanos , Esteroide 17-alfa-Hidroxilasa/metabolismo , Sulfotransferasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA