Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Steroid Biochem Mol Biol ; 170: 39-48, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27353462

RESUMEN

We investigated the effects of estrogens on glucose homeostasis using the Aromatase Knockout (ArKO) mouse, which is unable to convert androgens into estrogens. The ArKO mouse is a model of total estrogen ablation which develops symptoms of metabolic syndrome. To determine the development and progression of whole body state of insulin resistance of ArKO mice, comprehensive whole body tolerance tests were performed on WT, ArKO and estrogen administrated mice at 3 and 12 months of age. The absence of estrogens in the male ArKO mice leads to hepatic insulin resistance, glucose and pyruvate intolerance from 3 to 12 months with consistent improvement upon estrogen treatment. Estrogen absence in the female ArKO mice leads to glucose intolerance without pyruvate intolerance or insulin resistance. The replacement of estrogens in the female WT and ArKO mice exhibited both insulin sensitizing and resistance effects depending on age and dosage. In conclusion, this study presents information on the sexually dimorphic roles of estrogens on glucose homeostasis regulation.


Asunto(s)
Aromatasa/deficiencia , Aromatasa/genética , Estrógenos/metabolismo , Glucosa/metabolismo , Homeostasis , Animales , Aromatasa/metabolismo , Índice de Masa Corporal , Femenino , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Caracteres Sexuales
2.
PLoS One ; 10(8): e0136143, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26317527

RESUMEN

The maintenance of glucose homeostasis within the body is crucial for constant and precise performance of energy balance and is sustained by a number of peripheral organs. Estrogens are known to play a role in the maintenance of glucose homeostasis. Aromatase knockout (ArKO) mice are estrogen-deficient and display symptoms of dysregulated glucose metabolism. We aim to investigate the effects of estrogen ablation and exogenous estrogen administration on glucose homeostasis regulation. Six month-old female wildtype, ArKO, and 17ß-estradiol (E2) treated ArKO mice were subjected to whole body tolerance tests, serum examination of estrogen, glucose and insulin, ex-vivo muscle glucose uptake, and insulin signaling pathway analyses. Female ArKO mice display increased body weight, gonadal (omental) adiposity, hyperinsulinemia, and liver triglycerides, which were ameliorated upon estrogen treatment. Tolerance tests revealed that estrogen-deficient ArKO mice were pyruvate intolerant hence reflecting dysregulated hepatic gluconeogenesis. Analyses of skeletal muscle, liver, and adipose tissues supported a hepatic-based glucose dysregulation, with a down-regulation of Akt phosphorylation (a key insulin signaling pathway molecule) in the ArKO liver, which was improved with E2 treatment. Concurrently, estrogen treatment lowered ArKO serum leptin and adiponectin levels and increased inflammatory adipokines such as tumour necrosis factor alpha (TNFα) and interleukin 6 (IL6). Furthermore, estrogen deficiency resulted in the infiltration of CD45 macrophages into gonadal adipose tissues, which cannot be reversed by E2 treatment. This study describes the effects of estrogens on glucose homeostasis in female ArKO mice and highlights a primary phenotype of hepatic glucose dysregulation and a parallel estrogen modified adipokine profile.


Asunto(s)
Adipoquinas/sangre , Aromatasa/genética , Estradiol/sangre , Estrógenos/sangre , Gluconeogénesis , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Interleucina-6/sangre , Leptina/sangre , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Triglicéridos/metabolismo , Factor de Necrosis Tumoral alfa/sangre
3.
PLoS One ; 9(2): e87230, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24520329

RESUMEN

Estrogens are known to play a role in modulating metabolic processes within the body. The Aromatase knockout (ArKO) mice have been shown to harbor factors of Metabolic syndrome with central adiposity, hyperinsulinemia and male-specific hepatic steatosis. To determine the effects of estrogen ablation and subsequent replacement in males on whole body glucose metabolism, three- and six-month-old male ArKO mice were subjected to whole body glucose, insulin and pyruvate tolerance tests and analyzed for ensuing metabolic changes in liver, adipose tissue, and skeletal muscle. Estrogen-deficient male ArKO mice showed increased gonadal adiposity which was significantly reduced upon 17ß-estradiol (E2) treatment. Concurrently, elevated ArKO serum leptin levels were significantly reduced upon E2 treatment and lowered serum adiponectin levels were restored to wild type levels. Three-month-old male ArKO mice were hyperglycemic, and both glucose and pyruvate intolerant. These phenotypes continued through to 6 months of age, highlighting a loss of glycemic control. ArKO livers displayed changes in gluconeogenic enzyme expression, and in insulin signaling pathways upon E2 treatment. Liver triglycerides were increased in the ArKO males only after 6 months of age, which could be reversed by E2 treatment. No differences were observed in insulin-stimulated ex vivo muscle glucose uptake nor changes in ArKO adipose tissue and muscle insulin signaling pathways. Therefore, we conclude that male ArKO mice develop hepatic glucose intolerance by the age of 3 months which precedes the sex-specific development of hepatic steatosis. This can be reversed upon the administration of exogenous E2.


Asunto(s)
Aromatasa/deficiencia , Aromatasa/metabolismo , Intolerancia a la Glucosa/enzimología , Hígado/metabolismo , Hígado/patología , Adiponectina/sangre , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Estrógenos/farmacología , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/genética , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/patología , Insulina/sangre , Resistencia a la Insulina , Leptina/sangre , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculos/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ácido Pirúvico/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Triglicéridos/metabolismo
4.
J Endocrinol ; 210(3): 323-34, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21705395

RESUMEN

Male aromatase knockout mice (ArKO; an estrogen-deficient model) present with male-specific hepatic steatosis that is reversible upon 17ß-estradiol replacement. This study aims to elucidate which estrogen receptor (ER) subtype, ERα or ERß, is involved in the regulation of triglyceride (TG) homeostasis in the liver. Nine-month-old male ArKO mice were treated with vehicle, ERα- or ERß-specific agonists via s.c. injection, daily for 6 weeks. Male ArKO mice treated with ERα agonist had normal liver histology and TG contents compared with vehicle-treated ArKO; omental (gonadal) and infra-renal (visceral) fat pad weights were normalized to those of vehicle-treated wild-type (WT). In contrast, ERß agonist treatment did not result in the similar reversal of these ArKO phenotypes. In vehicle-treated ArKO mice, hepatic transcript expression of fatty acid synthase (Fasn) and stearoyl-coenzyme A desaturase 1 (key enzymes in de novo FA synthesis) were significantly elevated compared with vehicle-treated WT, but only Fasn expression was lowered to WT level after ERα agonist treatment. There were no significant changes in the transcript levels of carnitine palmitoyl transferase 1 (required for transfer of FA residues into the mitochondria for ß-oxidation) and sterol regulatory element-binding factor 1c (the upstream regulator of de novo FA synthesis). We also confirmed by RT-PCR that only ERα is expressed in the mouse liver. There were no changes in hepatic androgen receptor transcript level across all treatment groups. Our data suggest that estrogens act via ERα to regulate TG homeostasis in the ArKO liver. Since the liver, adipose tissue and arcuate nucleus express mainly ERα, estrogens could regulate hepatic functions via peripheral and central pathways.


Asunto(s)
Aromatasa/deficiencia , Receptor alfa de Estrógeno/agonistas , Hígado Graso/tratamiento farmacológico , Hígado Graso/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Animales , Aromatasa/genética , Secuencia de Bases , Cartilla de ADN/genética , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/agonistas , Receptor beta de Estrógeno/genética , Ácidos Grasos/biosíntesis , Hígado Graso/genética , Hígado Graso/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico , ARN/genética , ARN/metabolismo , Transducción de Señal , Triglicéridos/metabolismo
5.
Protein Cell ; 2(4): 333-46, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21574023

RESUMEN

Estrogen is implicated as playing an important role in aging and tumorigenesis of estrogen responsive tissues; however the mechanisms underlying the mitogenic actions of estrogen are not fully understood. Here we report that estrogen deficiency in mice caused by targeted disruption of the aromatase gene results in a significant inhibition of telomerase maintenance of telomeres in mouse ovaries in a tissue-specific manner. The inhibition entails a significant shortening of telomeres and compromised proliferation in the follicular granulosa cell compartment of ovary. Gene expression analysis showed decreased levels of proto-oncogene c-Myc and the telomerase catalytic subunit, telomerase reverse transcriptase (TERT), in response to estrogen deficiency. Estrogen replacement therapy led to increases in TERT gene expression, telomerase activity, telomere length and ovarian tissue growth, thereby reinstating ovary development to normal in four weeks. Our data demonstrate for the first time that telomere maintenance is the primary mechanism mediating the mitogenic effect of estrogen on ovarian granulosa cell proliferation by upregulating the genes of c-Myc and TERT in vivo. Estrogen deficiency or over-activity may cause ovarian tissue aging or tumorigenesis, respectively, through estrogen regulation of telomere remodeling.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX/genética , Envejecimiento/genética , Aromatasa/genética , Estrógenos , Células de la Granulosa/metabolismo , Ginecomastia/genética , Infertilidad Masculina/genética , Errores Innatos del Metabolismo/genética , Telomerasa/metabolismo , Telómero/química , Trastornos del Desarrollo Sexual 46, XX/tratamiento farmacológico , Trastornos del Desarrollo Sexual 46, XX/metabolismo , Envejecimiento/metabolismo , Animales , Aromatasa/deficiencia , Aromatasa/metabolismo , Proliferación Celular/efectos de los fármacos , Terapia de Reemplazo de Estrógeno , Estrógenos/deficiencia , Estrógenos/farmacología , Femenino , Expresión Génica , Genes myc/genética , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/patología , Ginecomastia/tratamiento farmacológico , Ginecomastia/metabolismo , Humanos , Infertilidad Masculina/tratamiento farmacológico , Infertilidad Masculina/metabolismo , Errores Innatos del Metabolismo/tratamiento farmacológico , Errores Innatos del Metabolismo/metabolismo , Ratones , Ratones Noqueados , Proto-Oncogenes Mas , Telomerasa/genética , Telómero/metabolismo , Telómero/patología
6.
Mol Cell Endocrinol ; 327(1-2): 56-64, 2010 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-20546829

RESUMEN

It remains to be established as to whether the absence of estrogen (direct) or the elevated levels of gonadotrophins and androgens (indirect) are responsible for the ArKO (aromatase knockout) ovarian phenotype. The aim of this study was to determine the effects of E(2) (17beta-estradiol) replacement, acyline (GnRH antagonist) and flutamide (anti-androgen) treatment on the ovarian phenotype of ArKO mice. E(2) replacement and acyline treatment but not flutamide treatment, reduced serum gonadotrophin levels of ArKO mice to within normal ranges. E(2) replacement improved uterine and ovarian follicular phenotypes and reduced the number of Sertoli-like filled cords by 62%. Acyline treatment reduced the number of hemorrhagic cysts and the number of Sertoli-like filled cords within ArKO ovaries. The data indicate that the absence of estrogen in concert with elevated levels of circulating gonadotrophins, principally LH, is responsible for the abnormal reproductive phenotype of the female ArKO mouse.


Asunto(s)
Aromatasa/deficiencia , Estrógenos/deficiencia , Hormona Luteinizante/metabolismo , Animales , Aromatasa/metabolismo , Peso Corporal/efectos de los fármacos , Cuerpo Lúteo/efectos de los fármacos , Cuerpo Lúteo/metabolismo , Cuerpo Lúteo/patología , Estradiol/farmacología , Estrógenos/metabolismo , Ciclo Estral/efectos de los fármacos , Femenino , Flutamida/farmacología , Gonadotropinas/sangre , Inmunohistoquímica , Ratones , Ratones Noqueados , Oligopéptidos/farmacología , Tamaño de los Órganos/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo , Folículo Ovárico/patología , Fenotipo , Placebos , Transporte de Proteínas/efectos de los fármacos , Factor de Transcripción SOX9/metabolismo , Esteroides/sangre
7.
Psychopharmacology (Berl) ; 206(2): 267-79, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19597801

RESUMEN

RATIONALE AND OBJECTIVES: The aim of the present study was to investigate the possible role of oestrogen in schizophrenia by comparing aromatase knockout (ArKO) mice, which are unable to produce oestrogen, with wild-type controls using two behavioural animal models with relevance to the illness, psychotropic drug-induced locomotor hyperactivity and prepulse inhibition (PPI). RESULTS: Baseline PPI was not different between ArKO and controls. Treatment with apomorphine, MK-801 and amphetamine caused disruption of PPI in all groups. However, in female but not male ArKO mice, the effect of both apomorphine and amphetamine was reduced. In female ArKO mice, amphetamine-induced hyperlocomotion was markedly reduced, but in male mice, the genotype difference was far smaller. Female but not male ArKO mice also showed a reduction of phencyclidine-induced locomotor hyperactivity. The density of dopamine transporters, but not D1 and D2 receptors, was significantly increased in the caudate putamen of male but not female ArKO mice compared to wild-type mice. This could represent a compensatory dopaminergic upregulation in male ArKO mice. CONCLUSION: Because of their lack of oestrogen production, it was anticipated that ArKO mice would display enhanced effects of amphetamine on locomotor activity and PPI. Instead, in these animals, aromatase knockout appeared to be 'protective'. This may represent limitations in the ability to model a complex illness such as schizophrenia in a constitutive knockout model, such as ArKO mice. Moreover, the current results may point at the involvement of other sex steroids, which are also altered in ArKO mice, in dopaminergic control of behaviour.


Asunto(s)
Aromatasa/deficiencia , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/fisiología , Hipercinesia/inducido químicamente , Inhibición Neural/efectos de los fármacos , Psicotrópicos/farmacología , Receptores de Dopamina D1/fisiología , Receptores de Dopamina D2/fisiología , Estimulación Acústica/métodos , Animales , Apomorfina/farmacología , Autorradiografía/métodos , Conducta Animal/efectos de los fármacos , Núcleo Caudado/efectos de los fármacos , Núcleo Caudado/metabolismo , Estudios Cruzados , Maleato de Dizocilpina/farmacología , Agonistas de Dopamina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Humanos , Hipercinesia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Fenciclidina/farmacología , Unión Proteica/efectos de los fármacos , Psicoacústica , Distribución Aleatoria , Reflejo de Sobresalto/efectos de los fármacos , Factores Sexuales , Factores de Tiempo
8.
Menopause ; 16(3): 582-8, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19182696

RESUMEN

OBJECTIVE: To explore the effects of tibolone on adiposity in the absence of aromatase and determine which of the hormonal properties of tibolone are exerting these effects. METHODS: In this study, vehicle; tibolone; estrogenic (ethinyl estradiol [EE]), progestogenic (ORG2058), or androgenic (dihydrotestosterone) compounds; or a combination of ORG2058 + EE was administered to 6-month-old ovariectomized aromatase knockout (ArKO) mice for a period of 6 weeks. RESULTS: In response to tibolone or EE-alone treatments, omental adipose tissue and infrarenal adipose tissue weights were significantly reduced (P = 0.004 and P = 0.01; P = 0.009 and P = 0.014, respectively) compared with those in ovariectomized and vehicle-treated ArKO mice. In contrast, adipose tissue weight tended to increase after ORG2058-alone treatment. Furthermore, EE in the presence of ORG2058 (ORG2058 + EE group) results in little effect on adiposity when compared with that in ovariectomized and vehicle-treated ArKO mice, showing that ORG2058 can negate the effect of EE. Dihydrotestosterone treatment did not have an impact on adipose tissue mass. Adipocyte volume and numbers followed the same treatment trends. CONCLUSIONS: In summary, our study in the ArKO mouse has confirmed the efficacy of tibolone as a hormone therapy to reduce adipose tissue accumulation after menopause and also shows that aromatization of tibolone is not required to elicit these estrogenic effects.


Asunto(s)
Adiposidad/efectos de los fármacos , Anabolizantes/farmacología , Aromatasa/metabolismo , Norpregnenos/farmacología , Ovariectomía , Adiposidad/fisiología , Animales , Femenino , Ratones , Ratones Noqueados
9.
Mol Cell Neurosci ; 41(1): 1-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19185610

RESUMEN

The aromatase knockout (ArKO) mouse is completely estrogen deficient. We previously detected apoptosis in the hypothalamus of 1 year-old male ArKO mice. This study shows that 12 week-old female ArKO mice display spontaneous apoptosis of pyramidal neurons in the frontal cortex while wild-type (WT) littermates show no signs of apoptosis. Concomitantly, bcl-2 related anti-apoptotic genes are down-regulated whereas the pro-apoptotic gene TRADD is up-regulated in the female ArKO frontal cortex. This phenotype can be rescued by 3-week replacement of 17beta-estradiol. Furthermore, the apoptosis phenotype is exacerbated in 12-15 month-old female ArKO mice, which have 30% less neurons in the frontal cortex and lower brain weights than WT counterparts. These data show that estrogens are essential for the survival of female cortical neurons even in the absence of pathological conditions or external assaults. Our observations also demonstrate the sexually dimorphic susceptibility of neurons to estrogen deficiency.


Asunto(s)
Apoptosis/fisiología , Aromatasa , Estrógenos/deficiencia , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Animales , Aromatasa/genética , Aromatasa/metabolismo , Caspasa 3/metabolismo , Supervivencia Celular , Proteínas de Unión al ADN , Estradiol/administración & dosificación , Femenino , Lóbulo Frontal/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Neuronas/patología , Proteínas Nucleares/metabolismo , Tamaño de los Órganos
10.
Ann N Y Acad Sci ; 1114: 48-55, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17986576

RESUMEN

Estrogen regulates tissue development, function, and aging by regulating expression of critical genes. Recent studies suggest that estrogen promotes cell proliferation by stimulating telomerase activity, an enzyme that plays a key role in extending cell life span. In some estrogen-responsive tissues, estrogen appears important in regulating telomerase activity in vivo. Further characterization will provide a greater understanding of the link between estrogen, telomerase, and aging.


Asunto(s)
Envejecimiento/metabolismo , Estrógenos/fisiología , Telomerasa/metabolismo , Envejecimiento/fisiología , Animales , Humanos
11.
Nat Clin Pract Endocrinol Metab ; 3(5): 414-21, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17452968

RESUMEN

Aromatase deficiency is rare in humans. Affected individuals cannot synthesize endogenous estrogens. Aromatase is the enzyme that catalyzes conversion of androgens into estrogens, and if aromatase is nonfunctional because of an inactivating mutation, estrogen synthesis cannot occur. If the fetus lacks aromatase activity, dehydroepiandrosterone sulfate produced by the fetal adrenal glands cannot be converted to estrogen by the placenta, so is converted to testosterone peripherally and results in virilization of both fetus and mother. Virilization manifests as pseudohermaphroditism in female infants, with hirsutism and acne in the mother; the maternal indicators resolve following delivery. To date, only seven males and seven females with aromatase deficiency have been reported. Affected females are typically diagnosed at birth because of the pseudohermaphroditism. Cystic ovaries and delayed bone maturation can occur during childhood and adolescence in these girls, who present at puberty with primary amenorrhea, failure of breast development, virilization, and hypergonadotrophic hypogonadism. Affected males, on the other hand, do not present with obvious defects at birth, so are diagnosed much later in life, presenting with clinical symptoms, which include tall stature, delayed skeletal maturation, delayed epiphyseal closure, bone pain, eunuchoid body proportions and excess adiposity. Estrogen replacement therapy reverses the symptoms in male and female patients.


Asunto(s)
Aromatasa/deficiencia , Adiposidad/fisiología , Adolescente , Adulto , Aromatasa/genética , Huesos/metabolismo , Terapia de Reemplazo de Estrógeno , Estrógenos/uso terapéutico , Femenino , Hormonas/uso terapéutico , Humanos , Infertilidad Masculina/metabolismo , Resistencia a la Insulina/fisiología , Masculino , Errores Innatos del Metabolismo/diagnóstico , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/terapia , Mutación , Embarazo , Factores Sexuales
12.
Biol Psychiatry ; 61(3): 359-66, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16566897

RESUMEN

BACKGROUND: Aromatase converts androgen to estrogen. Thus, the aromatase knockout (ArKO) mouse is estrogen deficient. We investigated the compulsive behaviors of these animals and the protein levels of catechol-O-methyltransferase (COMT) in frontal cortex, hypothalamus and liver. METHODS: Grooming was analyzed during the 20-min period immediately following a water-mist spray. Running wheel activity over two consecutive nights and barbering were analyzed. COMT protein levels were measured by Western analysis. RESULTS: Six-month old male but not female ArKO mice develop compulsive behaviors such as excessive barbering, grooming and wheel-running. Excessive activities were reversed by 3 weeks of 17beta-estradiol replacement. Interestingly, the presentation of compulsive behaviors is accompanied by concomitant decreases (p < .05) in hypothalamic COMT protein levels in male ArKO mice. These values returned to normal upon 17beta-estradiol treatment. In contrast, hepatic and frontal cortex COMT levels were not affected by the estrogen status, indicating region- and tissue-specific regulation of COMT levels by estrogen. No differences in COMT levels were detectable between female animals of both genotypes. CONCLUSIONS: This study describes the novel observation of a possible link between estrogen, COMT and development of compulsive behaviors in male animals which may have therapeutic implications in obsessive compulsive disorder (OCD) patients.


Asunto(s)
Conducta Compulsiva/fisiopatología , Conducta Compulsiva/psicología , Estrógenos/deficiencia , Animales , Aromatasa/genética , Aromatasa/fisiología , Western Blotting , Encéfalo/enzimología , Catecol O-Metiltransferasa/genética , Catecol O-Metiltransferasa/metabolismo , Conducta Compulsiva/genética , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Aseo Animal/fisiología , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/fisiología
13.
Endocrinology ; 147(12): 5907-13, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16990341

RESUMEN

Menopause is associated with an accumulation of visceral fat. An emerging concept suggests that relatively elevated levels of circulating androgens, compared with estrogens in postmenopausal women, underlie this shift in body fat distribution. In this study we administered dihydrotestosterone (DHT) to ovariectomized mice to examine the effect of relative androgen excess on adipose tissue distribution and function in estrogen-deficient mice. Compared with controls, DHT-treated mice exhibited increased body weight and visceral fat mass associated with triglyceride accumulation. Phosphorylation of AMP-activated protein kinase (AMPK) and acetyl CoA carboxylase was significantly decreased by DHT in visceral fat. In 3T3-L1 cells, DHT decreased phosphorylation of AMPK in a dose-dependent manner. In addition, DHT increased the expression of lipogenic genes (fatty acid synthase, sterol regulatory element binding protein-2, and lipoprotein lipase) in visceral fat. These data provide the first in vivo evidence that an increased androgen to estrogen ratio can promote visceral fat accumulation by inhibiting AMPK activation and stimulating lipogenesis.


Asunto(s)
Andrógenos/farmacología , Estrógenos/deficiencia , Grasa Intraabdominal/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Complejos Multienzimáticos/metabolismo , Obesidad/etiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP , Acetil-CoA Carboxilasa/metabolismo , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Tejido Adiposo/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Dihidrotestosterona/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ovariectomía/efectos adversos , Fosforilación/efectos de los fármacos , Triglicéridos/biosíntesis
14.
Psychopharmacology (Berl) ; 188(1): 100-10, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16896955

RESUMEN

OBJECTIVE: The aim of this study was to investigate the interaction of sex steroid hormones, particularly oestrogen, in the regulation of prepulse inhibition (PPI) by serotonin-1A (5-HT1A) receptors. MATERIALS AND METHODS: We studied aromatase knockout (ArKO) mice, which are unable to produce oestrogen but have high levels of testosterone, and the effects of castration. RESULTS AND DISCUSSION: Treatment of male ArKO mice with the 5-HT1A receptor agonist, 8-hydroxy-dipropyl-aminotetralin (8-OH-DPAT), caused an increase in PPI that was significantly greater than in male wild-type controls. Castration of male mice caused a significant enhancement of the effect of 8-OH-DPAT in control mice; however, there was no change in the effect of this drug in ArKO mice. There was no significant effect of 8-OH-DPAT on PPI in either female ArKO or wild-type controls. In all experiments, the effects of 8-OH-DPAT on startle were not different between the groups. [3H]8-OH-DPAT autoradiography showed no differences in 5-HT1A receptor binding densities in areas of the forebrain, hippocampus or raphe region that could explain the PPI results. These data show that the absence of oestrogen in male ArKO mice leads to a greater effect of 5-HT1A receptor stimulation on PPI. This effect can be mimicked in male control mice by castration. The differential involvement of oestrogen and testosterone in these animal models is discussed.


Asunto(s)
8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Aromatasa/deficiencia , Castración , Estrógenos/metabolismo , Reflejo de Sobresalto/efectos de los fármacos , Agonistas de Receptores de Serotonina/farmacología , Animales , Aromatasa/genética , Sitios de Unión , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Interacciones Farmacológicas , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Receptor de Serotonina 5-HT1A/metabolismo
15.
Trends Endocrinol Metab ; 17(2): 55-64, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16480891

RESUMEN

We are rapidly becoming aware of the importance of estrogen in maintaining virtually all facets of male health. In order for estrogens to be synthesized endogenously, the enzyme responsible for their synthesis from androgens, aromatase, must be functional. The seven known men in whom aromatase is nonfunctional all have a mutation in either exon V or IX of the CYP19 gene, which encodes aromatase. Collectively, these men are reported to have undetectable estrogen; normal to high levels of testosterone and gonadotropins; tall stature with delayed skeletal maturation and epiphyseal closure; osteoporosis; impaired lipid and insulin metabolism; and impaired reproductive function. The aromatase knockout mouse presents with a phenotype that is similar in many aspects and provides a valuable tool with which to examine and manipulate the actions of estrogen. By studying the naturally occurring aromatase-deficient humans, together with studies of the aromatase-knockout mouse, we are expanding our understanding of the essential role of estrogen in male physiology.


Asunto(s)
Aromatasa/deficiencia , Andrógenos/fisiología , Animales , Aromatasa/fisiología , Conducta/fisiología , Desarrollo Óseo/fisiología , Fenómenos Fisiológicos Cardiovasculares , Metabolismo Energético/fisiología , Estrógenos/deficiencia , Estrógenos/fisiología , Humanos , Sistema Inmunológico/fisiología , Síndrome Metabólico/fisiopatología , Ratones , Fenotipo
16.
Brain Res Mol Brain Res ; 140(1-2): 127-32, 2005 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-16083992

RESUMEN

The aromatase knockout (ArKO) mouse is estrogen deficient. Using reverse-transcription and real-time PCR, we showed that transcript levels of the N-methyl-d-aspartate (NMDA) receptor subunit NR2B are significantly higher in the hippocampus of female ArKO mice compared to wild-type (WT) littermates. Expression levels of NR1, NR2A, but not NR2C, also tended to be higher in ArKO mice. In the Morris watermaze test for spatial memory, both genotypes displayed equal significant improvement in the latency in locating the invisible platform over the 5-day training period. These findings show that selective loss of estrogen synthesis is associated with changes in NMDA receptor subunit expression in the hippocampus but little change in spatial learning ability.


Asunto(s)
Estrógenos/deficiencia , Hipocampo/fisiología , Aprendizaje por Laberinto/fisiología , Receptores de N-Metil-D-Aspartato/genética , Animales , Secuencia de Bases , Cartilla de ADN , Femenino , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa
17.
Neuroendocrinology ; 81(3): 158-66, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16015026

RESUMEN

The mechanism of regulation of growth hormone (GH) secretion by oestrogens and androgens is still controversial. Available data on the action of oestrogens on GH expression and secretion in somatotropes is poorly understood. We previously reported that the aromatase knockout (ArKO) mouse with oestrogen deficiency and excessive androgen levels had dysfunctional somatotropes. In order to clarify the influence of androgen and oestrogen, we investigated the in vivo treatment of ovariectomised (OVX) ArKO mice with exogenous oestradiol (E2) on the mRNA expression of GH, GH-secretagogue receptor (GHS-R), GH-releasing hormone receptor (GHRH-R), pituitary-specific transcription factor (Pit-1), and somatostatin receptors (sst1-5) in pituitary glands. Circulating plasma GH levels were also evaluated. The results showed that ArKO/OVX mice have a low expression of pituitary GH, GHRH-R, GHS-R and Pit-1, and significantly reduced GH levels. Treatment of female ArKO/OVX (E2-deficient without excessive androgen) mice with E2 for 21 days enhanced expression of pituitary GHRH-R and Pit-1 to 151 and 168%, respectively, of that in mice without treatment. E2 treatment increased GH expression and plasma levels in ArKO/OVX mice to levels comparable with those in wild-type female mice. We conclude therefore that long-term E2 replacement rescues the dysfunction of somatotropes in ArKO/OVX mice through increases in expression of GH, GHRH-R, and Pit-1 in the pituitary somatotropes, whereas the level of androgen in this oestrogen-deficient female mouse does not significantly influence the function of somatotropes.


Asunto(s)
Aromatasa/fisiología , Estradiol/fisiología , Hormona del Crecimiento/metabolismo , Hipófisis/metabolismo , Andrógenos/fisiología , Animales , Aromatasa/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Estradiol/deficiencia , Terapia de Reemplazo de Estrógeno , Femenino , Hormona del Crecimiento/genética , Ratones , Ratones Noqueados , Tamaño de los Órganos , Ovariectomía , Hipófisis/citología , ARN Mensajero/análisis , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Ghrelina , Receptores de Neuropéptido/genética , Receptores de Neuropéptido/metabolismo , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Factor de Transcripción Pit-1 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Mol Cell Neurosci ; 27(4): 466-76, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15555924

RESUMEN

The aromatase knockout (ArKO) mouse is unable to synthesize estrogens. Immunohistochemical studies on active caspase-3 and tyrosine hydroxylase (TH) revealed apoptosis of dopaminergic neurons in the medial preoptic area (MPO) and arcuate nucleus (Arc) of the hypothalamus of 1-year-old (1yo) male ArKO mice while no active caspase-3 was detected in wild type (WT). Furthermore, the number of TH-positive cells in the MPO and caudal Arc was significantly decreased in 1yo ArKO compared to WT. RNase protection assays support the presence of apoptosis in 1yo ArKO hypothalamus, revealing an up-regulation of pro-apoptotic genes: FASL, FADD, and caspase-8. Concomitantly, the ratio of bcl-2-related anti-apoptotic genes to pro-apoptotic genes in the hypothalamus of 1yo ArKO mice was significantly down-regulated. Previously, we have reported that no such changes were observed in the hypothalamus of female ArKO mice. Thus, we have provided direct evidence that estrogen is required to maintain the survival and functional integrity of dopaminergic neurons in the MPO and Arc of male, but not female mice.


Asunto(s)
Apoptosis/genética , Núcleo Arqueado del Hipotálamo/fisiopatología , Dopamina/metabolismo , Estrógenos/deficiencia , Degeneración Nerviosa/metabolismo , Área Preóptica/fisiopatología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Núcleo Arqueado del Hipotálamo/patología , Aromatasa/deficiencia , Aromatasa/genética , Caspasa 3 , Caspasa 8 , Caspasas/genética , Caspasas/metabolismo , Recuento de Células , Supervivencia Celular/genética , Regulación hacia Abajo/genética , Estrógenos/biosíntesis , Proteína Ligando Fas , Proteína de Dominio de Muerte Asociada a Fas , Genes bcl-2/genética , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Degeneración Nerviosa/genética , Área Preóptica/patología , ARN Mensajero/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Regulación hacia Arriba/genética
19.
Proc Natl Acad Sci U S A ; 101(34): 12628-33, 2004 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-15314222

RESUMEN

Sjögren's syndrome (SS) is an incurable, autoimmune exocrinopathy that predominantly affects females and whose pathogenesis remains unknown. Like rheumatoid arthritis, its severity increases after menopause, and estrogen deficiency has been implicated. We have reported that estrogen receptor-alpha and -beta-knockout mice develop autoimmune nephritis and myeloid leukemia, respectively, but neither develops SS. One model of estrogen deficiency in rodents is the aromatase-knockout (ArKO) mouse. In these animals, there is elevated B lymphopoiesis in bone marrow. We now report that ArKO mice develop severe autoimmune exocrinopathy resembling SS. By 1 year of age, there is B cell hyperplasia in the bone marrow, spleen, and blood of ArKO mice and spontaneous autoimmune manifestations such as proteinuria and severe leukocyte infiltration in the salivary glands and kidney. Also, as is typically found in human SS, there were proteolytic fragments of alpha-fodrin in the salivary glands and anti-alpha-fodrin antibodies in the serum of both female and male ArKO mice. When mice were raised on a phytoestrogen-free diet, there was a mild but significant incidence of infiltration of B lymphocytes in WT mice and severe destructive autoimmune lesions in ArKO mice. In age-matched WT mice fed a diet containing normal levels of phytoestrogen, there were no autoimmune lesions. These results reveal that estrogen deficiency results in a lymphoproliferative autoimmune disease resembling SS and suggest that estrogen might have clinical value in the prevention or treatment of this disease.


Asunto(s)
Aromatasa/metabolismo , Linfocitos B/metabolismo , Estrógenos/deficiencia , Trastornos Linfoproliferativos/inmunología , Síndrome de Sjögren/inmunología , Animales , Aromatasa/genética , Linfocitos B/citología , Linfocitos B/patología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Femenino , Humanos , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Trastornos Linfoproliferativos/metabolismo , Trastornos Linfoproliferativos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Proteinuria , Síndrome de Sjögren/metabolismo , Síndrome de Sjögren/patología , Bazo/metabolismo , Bazo/patología
20.
Endocrinology ; 145(9): 4286-91, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15178650

RESUMEN

Aromatase-deficient (ArKO) mice are deficient in estrogens due to deletion of the aromatase gene. We hypothesized that there may be changes in the cardiovascular system of ArKO mice because of evidence linking estrogens with improved cardiovascular outcomes and the induction of the glucocorticoid-metabolizing enzyme, 11beta-hydroxysteroid dehydrogenase type 2 (11betaHSD2), gene in the kidney, which is important for the regulation of blood pressure (BP). BP and baroreflex sensitivity (BRS) in female conscious ArKO mice were compared with those in age- and weight-matched wild-type (WT) mice. Power spectral analysis was used to determine cardiovascular variability and BRS. Although systolic BP was similar in the two groups, diastolic and mean BPs were lower in the ArKO mice (-6.3 +/- 1.9 and -4.6 +/- 2.1 mm Hg, respectively). Heart rate (HR) was greater in the ArKO mice (+36 +/- 6 beats/min). The mean BP in WT mice was 105 mm Hg, and the HR was 481 beats/min. In the autonomic frequency range, BP variability was 74% greater, and HR variability was only 26% that in WT mice. The BRS of ArKO mice was 46% of the value observed in WT mice. 11betaHSD2 levels were unaltered in ArKO mice, except in the kidney, where they were only 10% of WT levels. Estradiol administration to ArKO mice restored renal 11betaHSD2 to WT levels. The results show that ArKO mice have lower diastolic BP, but increased BP variability, perhaps due to an impaired BRS. Thus, aromatase activity is critical for normal autonomic control of the heart and, hence, for reducing the deleterious effects of high BP variability.


Asunto(s)
Aromatasa/genética , Aromatasa/metabolismo , Barorreflejo/fisiología , Presión Sanguínea/fisiología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Alimentación Animal , Animales , Estrógenos/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA