Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 38(7): 1519-1527, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29853568

RESUMEN

OBJECTIVE: Androgen deprivation therapy has been associated with increased cardiovascular risk in men. Experimental studies support that testosterone protects against atherosclerosis, but the target cell remains unclear. T cells are important modulators of atherosclerosis, and deficiency of testosterone or its receptor, the AR (androgen receptor), induces a prominent increase in thymus size. Here, we tested the hypothesis that atherosclerosis induced by testosterone deficiency in male mice is T-cell dependent. Further, given the important role of the thymic epithelium for T-cell homeostasis and development, we hypothesized that depletion of the AR in thymic epithelial cells will result in increased atherosclerosis. APPROACH AND RESULTS: Prepubertal castration of male atherosclerosis-prone apoE-/- mice increased atherosclerotic lesion area. Depletion of T cells using an anti-CD3 antibody abolished castration-induced atherogenesis, demonstrating a role of T cells. Male mice with depletion of the AR specifically in epithelial cells (E-ARKO [epithelial cell-specific AR knockout] mice) showed increased thymus weight, comparable with that of castrated mice. E-ARKO mice on an apoE-/- background displayed significantly increased atherosclerosis and increased infiltration of T cells in the vascular adventitia, supporting a T-cell-driven mechanism. Consistent with a role of the thymus, E-ARKO apoE-/- males subjected to prepubertal thymectomy showed no atherosclerosis phenotype. CONCLUSIONS: We show that atherogenesis induced by testosterone/AR deficiency is thymus- and T-cell dependent in male mice and that the thymic epithelial cell is a likely target cell for the antiatherogenic actions of testosterone. These insights may pave the way for new therapeutic strategies for safer endocrine treatment of prostate cancer.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Células Epiteliales/metabolismo , Linfocitos T/metabolismo , Testosterona/metabolismo , Timo/metabolismo , Animales , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Modelos Animales de Enfermedad , Células Epiteliales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Orquiectomía , Receptores Androgénicos/deficiencia , Receptores Androgénicos/genética , Testosterona/deficiencia , Timectomía , Timo/patología , Timo/cirugía
2.
Nat Commun ; 9(1): 2067, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29802242

RESUMEN

Testosterone deficiency in men is associated with increased risk for autoimmunity and increased B cell numbers through unknown mechanisms. Here we show that testosterone regulates the cytokine BAFF, an essential survival factor for B cells. Male mice lacking the androgen receptor have increased splenic B cell numbers, serum BAFF levels and splenic Baff mRNA. Testosterone deficiency by castration causes expansion of BAFF-producing fibroblastic reticular cells (FRCs) in spleen, which may be coupled to lower splenic noradrenaline levels in castrated males, as an α-adrenergic agonist decreases splenic FRC number in vitro. Antibody-mediated blockade of the BAFF receptor or treatment with the neurotoxin 6-hydroxydopamine revert the increased splenic B cell numbers induced by castration. Among healthy men, serum BAFF levels are higher in men with low testosterone. Our study uncovers a previously unrecognized regulation of BAFF by testosterone and raises important questions about BAFF in testosterone-mediated protection against autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Factor Activador de Células B/inmunología , Factor Activador de Células B/metabolismo , Linfocitos B/inmunología , Testosterona/metabolismo , Agonistas alfa-Adrenérgicos/farmacología , Animales , Enfermedades Autoinmunes/inmunología , Factor Activador de Células B/sangre , Receptor del Factor Activador de Células B/antagonistas & inhibidores , Receptor del Factor Activador de Células B/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Castración , Humanos , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Norepinefrina/metabolismo , Oxidopamina/farmacología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología , Testosterona/sangre , Testosterona/deficiencia , Testosterona/inmunología
3.
Cancer Cell ; 32(6): 824-839.e8, 2017 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-29153842

RESUMEN

Despite expression of oncogenic KRAS, premalignant pancreatic intraepithelial neoplasia 1 (PanIN1) lesions rarely become fully malignant pancreatic ductal adenocarcinoma (PDAC). The molecular mechanisms through which established risk factors, such as chronic pancreatitis, acinar cell damage, and/or defective autophagy increase the likelihood of PDAC development are poorly understood. We show that accumulation of the autophagy substrate p62/SQSTM1 in stressed KrasG12D acinar cells is associated with PDAC development and maintenance of malignancy in human cells and mice. p62 accumulation promotes neoplastic progression by controlling the NRF2-mediated induction of MDM2, which acts through p53-dependent and -independent mechanisms to abrogate checkpoints that prevent conversion of differentiated acinar cells to proliferative ductal progenitors. MDM2 targeting may be useful for preventing PDAC development in high-risk individuals.


Asunto(s)
Adenocarcinoma in Situ/patología , Carcinoma Ductal Pancreático/patología , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Células Acinares/metabolismo , Células Acinares/patología , Adenocarcinoma in Situ/metabolismo , Animales , Carcinoma Ductal Pancreático/metabolismo , Progresión de la Enfermedad , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Pancreáticas/metabolismo , Transducción de Señal/fisiología
4.
Oncotarget ; 7(51): 84326-84337, 2016 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-27741517

RESUMEN

Nf1 mutations or deletions are suggested to underlie the tumor predisposition of NF1 (neurofibromatosis type 1) and few treatments are available for treating NF1 patients with advanced malignant tumors. Aberrant activation of Ras in Nf1-deficient conditions is responsible for the promotion of tumorigenesis in NF1. PKC is proven to be an important factor in supporting the viability of Nf1-defected cells, but the molecular mechanisms are not fully understood. In this study, we demonstrate that the inhibition of protein kinase C (PKC) by 1-O-Hexadecyl-2-O-methyl-rac-glycerol (HMG, a PKC inhibitor) preferentially sensitizes Nf1-defected cells to apoptosis, via triggering a persistent mitotic arrest. In this process, Ral A is activated. Subsequently, Chk1 is phosphorylated and translocated to the nucleus. Silencing Ral A significantly blocks Chk1 nuclear translocation and releases HMG-treated Nf1-deficient cells from mitotic arrest, resulting in the reduction of the magnitude of apoptosis. Thus, our study reveals that PKC is able to maintain the homeostasis or viability of Nf1-defected cells and may serve as a potential target for developing new therapeutic strategies.


Asunto(s)
Apoptosis , Puntos de Control de la Fase M del Ciclo Celular , Neurofibromina 1/metabolismo , Proteína Quinasa C/metabolismo , Proteínas de Unión al GTP ral/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Éteres de Glicerilo/farmacología , Humanos , Ratones Endogámicos BALB C , Neoplasias de la Vaina del Nervio/tratamiento farmacológico , Neoplasias de la Vaina del Nervio/genética , Neoplasias de la Vaina del Nervio/metabolismo , Neurofibromina 1/genética , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Interferencia de ARN , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas de Unión al GTP ral/genética
5.
Endocrinology ; 157(11): 4200-4205, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27631553

RESUMEN

2-Methoxyestradiol (2ME2), a metabolite of 17ß-estradiol (E2), exerts bone sparing effects in animal models. We hypothesized that the underlying mechanism is back conversion of 2ME2 to E2, which subsequently acts via estrogen receptor (ER)α. We measured serum E2 levels in orchidectomized wild-type (WT) mice treated with 2ME2 66.6 µg/d or placebo. In placebo-treated animals, E2 was below the detection limit. In 2ME2-treated mice, the serum E2 level was 4.97 ± 0.68 pg/mL. This corresponds to the level found in diesterus in cycling female mice. Next, we investigated bone parameters in orchidectomized WT and ERα knockout mice treated with 2ME2 or placebo for 35 days. 2ME2 (6.66 µg/d) preserved trabecular and cortical bone in WT mice. Trabecular volumetric-bone mineral density was 64 ± 20%, and trabecular bone volume/total volume was 60 ± 20% higher in the metaphyseal region of the femur in the 2ME2 group, compared with placebo (P < .01). Both trabecular number and trabecular thickness were increased (P < .01). Cortical bone mineral content in the diaphyseal region of the femur was 31 ± 3% higher in the 2ME2 group, compared with placebo (P < .001). This was due to larger cortical area (P < .001). Three-point bending showed an increased bone strength in WT 2ME2-treated animals compared with placebo (maximum load [Fmax] +19±5% in the 2ME2 group, P < .05). Importantly, no bone parameter was affected by 2ME2 treatment in ERα knockout mice. In conclusion, 2ME2 treatment of orchidectomized mice results in increased serum E2. ERα mediates the bone sparing effects of 2ME2. The likely mediator of this effect is E2 resulting from back conversion of 2ME2.


Asunto(s)
Huesos/efectos de los fármacos , Huesos/metabolismo , Estradiol/análogos & derivados , Receptores de Estrógenos/metabolismo , 2-Metoxiestradiol , Animales , Peso Corporal/efectos de los fármacos , Densidad Ósea/efectos de los fármacos , Hueso Esponjoso/efectos de los fármacos , Hueso Esponjoso/metabolismo , Hueso Cortical/efectos de los fármacos , Hueso Cortical/metabolismo , Estradiol/sangre , Estradiol/metabolismo , Estradiol/farmacología , Femenino , Masculino , Ratones , Ratones Noqueados , Orquiectomía , Tamaño de los Órganos/efectos de los fármacos , Receptores de Estrógenos/genética , Testosterona/sangre , Tomografía Computarizada por Rayos X , Microtomografía por Rayos X
6.
Endocrinology ; 157(10): 3915-3923, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27533884

RESUMEN

Intimal hyperplasia is a vascular pathological process involved in the pathogenesis of atherosclerosis. Data suggest that T, the most important sex steroid hormone in males, protects men from atherosclerotic cardiovascular disease. T mainly acts via the androgen receptor (AR), and in this study we evaluated formation of intimal hyperplasia in male AR knockout (ARKO) mice using a vascular injury model. Two weeks after ligation of the carotid artery, male ARKO mice showed increased intimal area and intimal thickness compared with controls. After endothelial denudation by an in vivo scraping injury, there was no difference in the reendothelialization in ARKO compared with control mice. Ex vivo, we observed increased outgrowth of vascular smooth muscle cells from ARKO compared with control aortic tissue explants; the number of outgrown cells was almost doubled in ARKO. In vitro, stimulation of human aortic vascular smooth muscle cells with a physiological T concentration inhibited both migration and proliferation of the cells. Analyzing the expression of central regulators of cell proliferation and migration, we found that mRNA and protein levels of p27 were lower in uninjured arteries from ARKO mice and that T replacement to castrated male mice increased p27 mRNA in an AR-dependent manner. In conclusion, AR deficiency in male mice increases intimal hyperplasia in response to vascular injury, potentially related to the effects of androgens/AR to inhibit proliferation and migration of smooth muscle cells.


Asunto(s)
Traumatismos de las Arterias Carótidas/complicaciones , Neointima/etiología , Receptores Androgénicos/metabolismo , Testosterona/metabolismo , Animales , Traumatismos de las Arterias Carótidas/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Masculino , Ratones Noqueados , Miocitos del Músculo Liso/fisiología , Neointima/metabolismo
7.
Proc Natl Acad Sci U S A ; 112(45): E6166-74, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26512112

RESUMEN

Pancreatic acinar cells possess very high protein synthetic rates as they need to produce and secrete large amounts of digestive enzymes. Acinar cell damage and dysfunction cause malnutrition and pancreatitis, and inflammation of the exocrine pancreas that promotes development of pancreatic ductal adenocarcinoma (PDAC), a deadly pancreatic neoplasm. The cellular and molecular mechanisms that maintain acinar cell function and whose dysregulation can lead to tissue damage and chronic pancreatitis are poorly understood. It was suggested that autophagy, the principal cellular degradative pathway, is impaired in pancreatitis, but it is unknown whether impaired autophagy is a cause or a consequence of pancreatitis. To address this question, we generated Atg7(Δpan) mice that lack the essential autophagy-related protein 7 (ATG7) in pancreatic epithelial cells. Atg7(Δpan) mice exhibit severe acinar cell degeneration, leading to pancreatic inflammation and extensive fibrosis. Whereas ATG7 loss leads to the expected decrease in autophagic flux, it also results in endoplasmic reticulum (ER) stress, accumulation of dysfunctional mitochondria, oxidative stress, activation of AMPK, and a marked decrease in protein synthetic capacity that is accompanied by loss of rough ER. Atg7(Δpan) mice also exhibit spontaneous activation of regenerative mechanisms that initiate acinar-to-ductal metaplasia (ADM), a process that replaces damaged acinar cells with duct-like structures.


Asunto(s)
Células Acinares/fisiología , Autofagia/fisiología , Estrés del Retículo Endoplásmico/fisiología , Homeostasis/fisiología , Proteínas Asociadas a Microtúbulos/deficiencia , Páncreas/citología , Biosíntesis de Proteínas/fisiología , Animales , Proteína 7 Relacionada con la Autofagia , Técnica del Anticuerpo Fluorescente , Immunoblotting , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Microscopía Electrónica de Transmisión
8.
FASEB J ; 29(4): 1540-50, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25550469

RESUMEN

Androgens have important cardiometabolic actions in males, but their metabolic role in females is unclear. To determine the physiologic androgen receptor (AR)-dependent actions of androgens on atherogenesis in female mice, we generated female AR-knockout (ARKO) mice on an atherosclerosis-prone apolipoprotein E (apoE)-deficient background. After 8 weeks on a high-fat diet, but not on a normal chow diet, atherosclerosis in aorta was increased in ARKO females (+59% vs. control apoE-deficient mice with intact AR gene). They also displayed increased body weight (+18%), body fat percentage (+62%), and hepatic triglyceride levels, reduced insulin sensitivity, and a marked atherogenic dyslipidemia (serum cholesterol, +52%). Differences in atherosclerosis, body weight, and lipid levels between ARKO and control mice were abolished in mice that were ovariectomized before puberty, consistent with a protective action of ovarian androgens mediated via the AR. Furthermore, the AR agonist dihydrotestosterone reduced atherosclerosis (-41%; thoracic aorta), subcutaneous fat mass (-44%), and cholesterol levels (-35%) in ovariectomized mice, reduced hepatocyte lipid accumulation in hepatoma cells in vitro, and regulated mRNA expression of hepatic genes pivotal for lipid homeostasis. In conclusion, we demonstrate that the AR protects against diet-induced atherosclerosis in female mice and propose that this is mediated by modulation of body composition and lipid metabolism.


Asunto(s)
Aterosclerosis/prevención & control , Dislipidemias/prevención & control , Obesidad/prevención & control , Receptores Androgénicos/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Colesterol/metabolismo , Dieta/efectos adversos , Dihidrotestosterona/farmacología , Dislipidemias/etiología , Dislipidemias/metabolismo , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso/prevención & control , Femenino , Resistencia a la Insulina , Metabolismo de los Lípidos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Obesidad/metabolismo , Orquiectomía , Ovariectomía , Receptores Androgénicos/deficiencia , Receptores Androgénicos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...