Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Drug Dev Res ; 79(7): 324-331, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30267584

RESUMEN

Shenmai injection (SMI) is increasingly used in tumor combination therapy, devoting to enhancing anti-tumor effects and reducing the toxicity of chemotherapy drugs. This study aimed to explore the role of SMI in papillary thyroid carcinoma (PTC) treatment. Flow cytometry was used to examine Treg cells percentage in CD4 + T cells. The expression of RNA and protein was analyzed by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot, respectively. Inducers were used to stimulate CD4 + T cells to differentiate into Treg cells. The interaction between miR-103 and G protein-coupled estrogen receptor 1 (GPER1) was confirmed with the dual luciferase assays. Cell transfection and recombinant plasmids were used to achieve endogenous expression. Compared with patients not treated with 131 I, the Treg cells percentage and Foxp3 expression were clearly increased in patients with 131 I radiotherapy, just the opposite in SMI combination therapy. SMI inhibited the differentiation of CD4 + T cells into Treg cells. Aberrant expression of miR-103 and GPER1 induced by 131 I was reversed by SMI and 131 I combination therapy. GPER1 was negatively regulated by miR-103 and SMI inhibits the differentiation of CD4 + T cells into Treg cells via miR-103/GPER1 axis, which improves the postoperative immunological function of PTC patients with 131 I radiotherapy.


Asunto(s)
Diferenciación Celular/fisiología , Medicamentos Herbarios Chinos/administración & dosificación , Inmunidad Celular/fisiología , MicroARNs/biosíntesis , Receptores de Estrógenos/biosíntesis , Receptores Acoplados a Proteínas G/biosíntesis , Linfocitos T Reguladores/metabolismo , Cáncer Papilar Tiroideo/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Combinación de Medicamentos , Humanos , Inmunidad Celular/efectos de los fármacos , Inyecciones , MicroARNs/inmunología , Cuidados Posoperatorios/métodos , Receptores de Estrógenos/inmunología , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Cáncer Papilar Tiroideo/tratamiento farmacológico , Cáncer Papilar Tiroideo/inmunología
2.
Stress ; 18(4): 367-80, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26176590

RESUMEN

The last decade has witnessed profound growth in studies examining the role of fundamental neuroimmune processes as key mechanisms that might form a natural bridge between normal physiology and pathological outcomes. Rooted in core concepts from psychoneuroimmunology, this review utilizes a succinct, exemplar-driven approach of several model systems that contribute significantly to our knowledge of the mechanisms by which neuroimmune processes interact with stress physiology. Specifically, we review recent evidence showing that (i) stress challenges produce time-dependent and stressor-specific patterns of cytokine/chemokine expression in the CNS; (ii) inflammation-related genes exhibit unique expression profiles in males and females depending upon individual, cooperative or antagonistic interactions between steroid hormone receptors (estrogen and glucocorticoid receptors); (iii) adverse social experiences incurred through repeated social defeat engage a dynamic process of immune cell migration from the bone marrow to brain and prime neuroimmune function and (iv) early developmental exposure to an inflammatory stimulus (carageenin injection into the hindpaw) has a lasting influence on stress reactivity across the lifespan. As such, the present review provides a theoretical framework for understanding the role that neuroimmune mechanisms might play in stress plasticity and pathological outcomes, while at the same time pointing toward features of the individual (sex, developmental experience, stress history) that might ultimately be used for the development of personalized strategies for therapeutic intervention in stress-related pathologies.


Asunto(s)
Encéfalo/inmunología , Citocinas/inmunología , Regulación de la Expresión Génica , Neuroinmunomodulación/inmunología , Receptores de Estrógenos/inmunología , Receptores de Glucocorticoides/inmunología , Estrés Psicológico/inmunología , Adultos Sobrevivientes de Eventos Adversos Infantiles , Animales , Encéfalo/crecimiento & desarrollo , Quimiocinas/inmunología , Femenino , Humanos , Inflamación , Masculino , Plasticidad Neuronal , Factores Sexuales
3.
J Immunol ; 194(6): 2810-8, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25681337

RESUMEN

Macrophages play a major role in joint inflammation. Estrogen is involved in rheumatoid arthritis and temporomandibular disorders. However, the underlying mechanism is still unclear. This study was done to verify and test how estrogen affects M1/M2-like macrophage polarization and then contributes to joint inflammation. Female rats were ovariectomized and treated with increasing doses of 17ß-estradiol for 10 d and then intra-articularly injected with CFA to induce temporomandibular joint (TMJ) inflammation. The polarization of macrophages and expression of cadherin-11 was evaluated at 24 h after the induction of TMJ inflammation and after blocking cadherin-11 or estrogen receptors. NR8383 macrophages were treated with estradiol and TNF-α, with or without blocking cadherin-11 or estrogen receptors, to evaluate the expression of the M1/M2-like macrophage-associated genes. We found that estradiol increased the infiltration of macrophages with a proinflammatory M1-like predominant profile in the synovium of inflamed TMJ. In addition, estradiol dose-dependently upregulated the expressions of the M1-associated proinflammatory factor inducible NO synthase (iNOS) but repressed the expressions of the M2-associated genes IL-10 and arginase in NR8383 macrophages. Furthermore, estradiol mainly promoted cadherin-11 expression in M1-like macrophages of inflamed TMJ. By contrast, blockage of cadherin-11 concurrently reversed estradiol-potentiated M1-like macrophage activation and TMJ inflammation, as well as reversed TNF-α-induced induction of inducible NO synthase and NO in NR8383 macrophages. The blocking of estrogen receptors reversed estradiol-potentiated M1-like macrophage activation and cadherin-11 expression. These results suggested that estradiol could promote M1-like macrophage activation through cadherin-11 to aggravate the acute inflammation of TMJs.


Asunto(s)
Cadherinas/inmunología , Estradiol/inmunología , Inflamación/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Articulación Temporomandibular/inmunología , Animales , Arginasa/genética , Arginasa/inmunología , Arginasa/metabolismo , Artritis/genética , Artritis/inmunología , Artritis/metabolismo , Western Blotting , Cadherinas/genética , Cadherinas/metabolismo , Estradiol/análogos & derivados , Estradiol/farmacología , Antagonistas del Receptor de Estrógeno/farmacología , Estrógenos/inmunología , Estrógenos/farmacología , Femenino , Fulvestrant , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Inflamación/genética , Inflamación/metabolismo , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-10/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Microscopía Confocal , Óxido Nítrico/inmunología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ovariectomía , Ratas Sprague-Dawley , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/inmunología , Receptores de Estrógenos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Articulación Temporomandibular/efectos de los fármacos , Articulación Temporomandibular/patología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/farmacología
4.
Clinics (Sao Paulo) ; 61(5): 479-88, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17072448

RESUMEN

Several clinical and experimental studies have demonstrated gender dimorphism in immune and organ responsiveness and in the susceptibility to and morbidity from shock, trauma, and sepsis. In this respect, cell-mediated immune responses have been shown to be depressed in males following trauma-hemorrhage, whereas they were aintained/enhanced in proestrus females. Furthermore, sex hormones have been shown to be responsible for this gender-specific immune response following adverse circulatory conditions. More specifically, studies indicate that androgens produce immunodepression following trauma-hemorrhage in males. In contrast, female sex steroids appear to exhibit immunoprotective properties following trauma and severe blood loss. With regard to the underlying mechanisms, receptors for sex hormones have been identified on various immune cells suggesting direct effects of these hormones on the immune cells. Alternatively, indirect effects of sex hormones, ie, modulation of cardiovascular responses or androgen- and estrogen-synthesizing enzymes, might contribute to gender-specific immune responses. Recent studies indicate that sex hormones, eg, dehydroepiandrosterone (DHEA), also modulate the function of peripheral blood mononuclear cells in surgical patients. Thus, the immunomodulatory properties of sex hormones/receptor antagonists/sex steroid synthesizing enzymes following trauma-hemorrhage suggests novel therapeutic strategies for the treatment of immunodepression in surgical patients.


Asunto(s)
Hormonas Esteroides Gonadales/inmunología , Sepsis/inmunología , Factores Sexuales , Choque Hemorrágico/inmunología , Heridas y Lesiones/inmunología , Adyuvantes Inmunológicos/uso terapéutico , Antagonistas de Receptores Androgénicos , Andrógenos/inmunología , Circulación Sanguínea , Deshidroepiandrosterona/inmunología , Deshidroepiandrosterona/uso terapéutico , Susceptibilidad a Enfermedades , Estrógenos/inmunología , Femenino , Humanos , Inmunocompetencia , Masculino , Receptores Androgénicos/inmunología , Receptores Androgénicos/uso terapéutico , Receptores de Estrógenos/inmunología , Sepsis/tratamiento farmacológico , Sepsis/fisiopatología , Choque Hemorrágico/tratamiento farmacológico , Choque Hemorrágico/fisiopatología , Índices de Gravedad del Trauma , Heridas y Lesiones/tratamiento farmacológico , Heridas y Lesiones/fisiopatología
5.
Clinics ; 61(5): 479-488, Oct. 2006. ilus
Artículo en Inglés | LILACS | ID: lil-436774

RESUMEN

Several clinical and experimental studies have demonstrated gender dimorphism in immune and organ responsiveness and in the susceptibility to and morbidity from shock, trauma, and sepsis. In this respect, cell-mediated immune responses have been shown to be depressed in males following trauma-hemorrhage, whereas they were aintained/enhanced in proestrus females. Furthermore, sex hormones have been shown to be responsible for this gender-specific immune response following adverse circulatory conditions. More specifically, studies indicate that androgens produce immunodepression following trauma-hemorrhage in males. In contrast, female sex steroids appear to exhibit immunoprotective properties following trauma and severe blood loss. With regard to the underlying mechanisms, receptors for sex hormones have been identified on various immune cells suggesting direct effects of these hormones on the immune cells. Alternatively, indirect effects of sex hormones, ie, modulation of cardiovascular responses or androgen- and estrogen-synthesizing enzymes, might contribute to gender-specific immune responses. Recent studies indicate that sex hormones, eg, dehydroepiandrosterone (DHEA), also modulate the function of peripheral blood mononuclear cells in surgical patients. Thus, the immunomodulatory properties of sex hormones/receptor antagonists/sex steroid synthesizing enzymes following trauma-hemorrhage suggests novel therapeutic strategies for the treatment of immunodepression in surgical patients.


Uma série de estudos clínicos e experimentais demonstram a existência de dimorfismo sexual das respostas imunológicas e orgânicas, bem como da suscetibilidade e morbidade em relação ao choque, ao trauma e à sepse. Respostas imunes celularmente mediadas apresentam-se deprimidas em machos em resposta ao binômio trauma-hemorragia, mas conservados/enaltecidos em fêmeas em proestro. Adicionalmente demonstra-se que os hormônios sexuais são responsáveis por esta dicomotomia de resposta sexualmente específica, em condições cardiovasculares adversas. Estudos específicos indicam que os andrógenos produzem imunodepressão pós-trauma hemorragia em machos. Em contraste, esteróides sexuais femininos parecem exibir propriedades imunoprotetoras após episódios de trauma com ou sem perda importante de sangue. No terreno dos mecanismos subjacentes, foram identificados receptores para hormônios sexuais em várias células do sistema imunológico, sugerindo a existência de efeitos diretos destes hormônios sobre tais células. Alternativamente, observam efeitos indiretos de hormônios sexuais tais como modulação das respostas cardiovasculares das enzimas sintetizadores de andrógeno e estrógeno, que podem contribuir para as estas respostas sexualmente diferenciadas. Estudos recentes indicam que os hormônios sexuais, como por exemplo a dehidroepiandrosterona também modulam a função de células mononucleares da série branca em pacientes cirúrgicos. Assim, as propriedades imunomodulatórias de hormônios sexuais/antagonistas de receptores/enzimas sintetizadores de esteróides após a ocorrência de trauma ou de hemorragia sugerem o caminho para novas estratégias terapêuticas para o tratamento de imunodepressão em pacientes cirúrgicos.


Asunto(s)
Humanos , Masculino , Femenino , Hormonas Esteroides Gonadales/inmunología , Caracteres Sexuales , Sepsis/inmunología , Choque Hemorrágico/inmunología , Heridas y Lesiones/inmunología , Adyuvantes Inmunológicos/uso terapéutico , Andrógenos/inmunología , Circulación Sanguínea , Susceptibilidad a Enfermedades , Deshidroepiandrosterona/inmunología , Deshidroepiandrosterona/uso terapéutico , Estrógenos/inmunología , Inmunocompetencia , Receptores Androgénicos/antagonistas & inhibidores , Receptores Androgénicos/inmunología , Receptores Androgénicos/uso terapéutico , Receptores de Estrógenos/inmunología , Sepsis/tratamiento farmacológico , Sepsis/fisiopatología , Choque Hemorrágico/tratamiento farmacológico , Choque Hemorrágico/fisiopatología , Índices de Gravedad del Trauma , Heridas y Lesiones/tratamiento farmacológico , Heridas y Lesiones/fisiopatología
6.
J Nutr ; 136(3): 704-8, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16484547

RESUMEN

Estrogen regulates thymic development and immune function. Despite the critical role of estrogens in inducing thymic involution and modulating immune responses, the mechanism of this effect is unclear. Similarly, humans and animals are exposed to increasing amounts of the estrogenic soy isoflavone genistein in the diet, but whether genistein can induce immune changes has not been definitively established. We reported previously that genistein induces thymic atrophy in mice, and decreases both humoral and cell-mediated immunity. These thymic effects of genistein occur via estrogen receptor (ER)-mediated and non-ER-mediated pathways. Genistein injections produced the most pronounced effects, but dietary administration to mice that produced serum genistein concentrations similar to those reported in human infants consuming soy formula also had demonstrable effects. Microarray analysis of the effects of estradiol and genistein on neonatal thymus indicated that estradiol affected genes involved in transcription, apoptosis, cell cycle, and thymic development and function; genistein had similar effects on many estradiol target genes, but also had unique actions not replicated by estradiol. Despite extensive work showing inhibitory effects of genistein on immunity, other rodent studies reported that genistein or other phytoestrogens stimulate various aspects of immune function. Although the present data strongly indicate that genistein can regulate immune function, possibly at physiologic concentrations, further work is required to definitively establish overall thymic and immune effects of genistein and soy, which may vary with age, species, and specific end point.


Asunto(s)
Genisteína/farmacología , Inmunidad , Fitoestrógenos/farmacología , Receptores de Estrógenos/inmunología , Animales , Dieta , Cobayas , Humanos , Ratones , Modelos Animales , Ratas , Receptores de Estrógenos/efectos de los fármacos , Glycine max
7.
Crit Care Med ; 33(8): 1779-86, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16096456

RESUMEN

OBJECTIVE: Peripheral blood mononuclear cell (PBMC) dysfunction occurs following major abdominal surgery and correlates with an increased rate of septic complications. Studies have shown that dehydroepiandrosterone (DHEA) restores cell-mediated immune responses after trauma-hemorrhage in mice. Nonetheless, it remains unknown whether DHEA has any salutary effects on depressed PBMC function in surgical patients. DESIGN: Laboratory experiment. SETTING: University laboratory. PATIENTS: Fifteen patients undergoing major abdominal surgery. INTERVENTIONS: Blood samples were obtained preoperatively and 2 hrs postoperatively. MEASUREMENTS AND MAIN RESULTS: PBMCs were cultured with 33% plasma in the presence or absence of DHEA (10(-10) M, 10(-8) M physiologic concentration, 10(-6) M, 10(-5) M). In an additional set of samples, the estrogen receptor antagonist tamoxifen (10(-6) M) was added. The release of proinflammatory cytokines (interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha) was measured in the supernatants by enzyme-linked immunosorbent assay. Abdominal surgery resulted in depressed interleukin-1beta and tumor necrosis factor-alpha release by PBMC. Addition of DHEA to the culture medium, however, significantly improved the release of interleukin-1beta and tumor necrosis factor-alpha and stimulated the interleukin-6 release capacity of PBMC. This effect was most pronounced for a concentration of 10(-5)M DHEA. The immunomodulatory effect of DHEA on PBMC cytokine release was completely blocked by tamoxifen. In contrast, the modulatory effect of DHEA was enhanced by the addition of postoperative plasma. CONCLUSIONS: DHEA stimulates proinflammatory cytokine release capacities of human PBMCs following major abdominal surgery. The estrogen receptor appears to be involved in mediating the immunomodulatory effect of DHEA. Thus, DHEA might be a useful adjunct for preventing immunosuppression in surgical patients.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Deshidroepiandrosterona/farmacología , Huésped Inmunocomprometido/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Complicaciones Posoperatorias/inmunología , Receptores de Estrógenos/inmunología , Abdomen/cirugía , Adulto , Anciano , Células Cultivadas , Citocinas/sangre , Antagonistas de Estrógenos/farmacología , Femenino , Humanos , Inmunidad Celular , Huésped Inmunocomprometido/inmunología , Leucocitos Mononucleares/metabolismo , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias/prevención & control , Estudios Prospectivos , Sepsis/inmunología , Sepsis/prevención & control , Tamoxifeno/farmacología
8.
Clin Exp Immunol ; 135(2): 219-25, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14738448

RESUMEN

Diets rich in soy phytoestrogens have many potential health benefits but isoflavones such as genistein may suppress cell mediated immune function. The effect of dietary phytoestrogens on the host response to infection has not been extensively examined. Mice were fed a diet containing soy phytoestrogens and infected with Mycobacterium avium to establish a chronic infection and inflammatory response. As phytoestrogens may act through classical oestrogen receptors (ER), mice deficient in ERalpha signalling and wild type mice were evaluated for a panel of Type 1-associated cytokines (IFNgamma, IL-12 and IL-18) in the spleen. IFNgamma production in the spleen was increased approximately 4-fold in ERalpha-deficient mice fed a casein-based diet over wild type mice fed a casein-based diet (P < 0.05), suggesting a role for ERalpha in suppressing IFNgamma production. IL-18 levels in spleens of wild type mice were decreased compared to ERalpha-deficient mice on a casein diet. Splenic IL-12 and IL-18 levels were not affected in wild type and ERalpha-deficient mice on the phytoestrogen containing diets, with the exception that whole soy increased IL-12 levels in the tissues of ERalpha deficient mice. We conclude that ERalpha and dietary phytoestrogens can influence production of key regulatory cytokines in response to chronic bacterial infection.


Asunto(s)
Glycine max/efectos adversos , Inmunosupresores/administración & dosificación , Interferón gamma/biosíntesis , Isoflavonas/administración & dosificación , Mycobacterium avium/inmunología , Preparaciones de Plantas/administración & dosificación , Receptores de Estrógenos/inmunología , Tuberculosis/inmunología , Animales , Caseínas/administración & dosificación , Cromatografía Líquida de Alta Presión/métodos , Recuento de Colonia Microbiana , Suplementos Dietéticos/efectos adversos , Inhibidores Enzimáticos/sangre , Genisteína/administración & dosificación , Genisteína/sangre , Inmunidad Celular/inmunología , Inmunosupresores/efectos adversos , Interleucina-12/análisis , Interleucina-18/análisis , Isoflavonas/efectos adversos , Ratones , Ratones Endogámicos C57BL , Fitoestrógenos , Preparaciones de Plantas/efectos adversos , Transducción de Señal/inmunología , Bazo/inmunología
9.
Biol Reprod ; 69(4): 1318-24, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12773414

RESUMEN

Pituitary gland growth hormone (GH) secretion is influenced by two hypothalamic neuropeptides: growth hormone-releasing hormone (GHRH) and somatostatin. Recent data also suggest that estrogen modulates GH release, particularly at the time of the preovulatory luteinizing hormone surge, when a coincident surge of GH is observed in sheep. The GHRH neurons do not possess estrogen receptor alpha (ERalpha), suggesting that estrogen does not act directly on GHRH neurons. Similarly, few somatotropes express ERalpha, suggesting a weak pituitary effect of estradiol on GH. It was hypothesized, therefore, that estradiol may affect somatostatin neurons to modulate GH release from the pituitary. Using immunocytochemical approaches, the present study revealed that although somatostatin neurons were located in several hypothalamic sites, only those in the arcuate nucleus (13% +/- 2%) and ventromedial nucleus (VMN; 29% +/- 1%) expressed ERalpha. In addition, we found that all neurons immunoreactive for somatostatin-14 were also immunoreactive for somatostatin-28(1-12). To determine whether increased GH secretion in response to estradiol is through modulation of GHRH and/or somatostatin neuronal activity, a final study investigated whether c-fos expression increased in somatostatin- and GHRH-immunoreactive cells at the time of the estradiol-induced LH surge in intact anestrous ewes. Estradiol significantly (P < 0.05) increased the percentage of GHRH (estradiol, 75% +/- 3%; no estradiol, 19% +/- 2%) neurons expressing c-fos in the hypothalamus. The percentage of somatostatin-immunoreactive neurons coexpressing c-fos in the estradiol-treated animals was significantly (P < 0.05) higher (periventricular, 44% +/- 3%; arcuate, 72% +/- 5%; VMN, 81% +/- 5%) than in the control animals (periventricular, 22% +/- 1%; arcuate, 29% +/- 3%; VMN, 31% +/- 3%). The present study suggests that estradiol modulates the activity of GHRH and somatostatin neurons but that this effect is most likely mediated through an indirect interneuronal pathway.


Asunto(s)
Estradiol/farmacología , Hipotálamo/metabolismo , Neuronas/metabolismo , Receptores de Estrógenos/metabolismo , Somatostatina/metabolismo , Animales , Receptor alfa de Estrógeno , Femenino , Hormona del Crecimiento/metabolismo , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/inmunología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/inmunología , Ovinos , Somatostatina/efectos de los fármacos , Somatostatina/inmunología , Somatostatina-28
10.
Brain Res Dev Brain Res ; 91(2): 300-3, 1996 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-8852382

RESUMEN

Developmental expression of the estrogen receptor (ER) in rat hypothalamus was examined using immunohistochemistry. In the medial preoptic nucleus and ventromedial nucleus ER-immunoreactivity was detected as early as E17, whereas ER protein expression in the periventricular preoptic nucleus and arcuate nucleus was delayed until E19. These results show that following a region specific onset of the ER protein expression sex differences in ER levels are already detectable during the perinatal period.


Asunto(s)
Hipotálamo/embriología , Hipotálamo/crecimiento & desarrollo , Receptores de Estrógenos/inmunología , Animales , Especificidad de Anticuerpos , Femenino , Hipotálamo/química , Inmunohistoquímica , Masculino , Embarazo , Ratas , Ratas Wistar , Receptores de Estrógenos/análisis , Caracteres Sexuales
11.
Neuroendocrinology ; 59(6): 552-60, 1994 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8084379

RESUMEN

The H222 and ER-715 anti-estrogen receptor (ER) antibodies were used to examine the distribution of ER immunoreactive (ERir) neurons in hypothalamic and limbic sites of: (i) castrated male rats; (ii) castrated males implanted s.c. with silastic capsules containing testosterone (T), and (iii) castrated males receiving T together with 0.25 mg/kg/day of the nonsteroidal aromatase inhibitor, fadrozole (CIBA-Geigy CGS 16949A), delivered s.c. by means of implanted osmotic minipumps. Because labeling of ERir neurons in rat brain with H222 anti-ER antibody is reported to decrease when estrogen is present, it was used here to determine whether or not estrogen derived from the aromatization of T would affect ERir neuronal labeling. Castrated males showed H222 ERir-positive neurons in the lateral septum, medial preoptic area, several subdivisions of the hypothalamus, amygdala, and bed nucleus of stria terminalis. In contrast, in T-treated castrates, H222 ERir labeling was either eliminated or greatly reduced in all brain areas with the exception of the lateral septum. In castrated male rats given T together with fadrozole, H222 ERir labeling was restored in all brain areas where it had been reduced by T treatment. The ER-715 antibody effectively labeled neurons in all brain regions independently of the treatment condition, indicating that ER was present in the brains of animals in all treatment groups. These findings point to functional differences in ER dynamics in brain areas implicated in the control of sexual behavior by male rats.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Inhibidores de la Aromatasa , Encéfalo/metabolismo , Neuronas/metabolismo , Receptores de Estrógenos/metabolismo , Testosterona/farmacología , Animales , Anticuerpos , Autorradiografía , Encéfalo/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Estradiol/farmacología , Fadrozol/farmacología , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inmunohistoquímica , Masculino , Neuronas/efectos de los fármacos , Orquiectomía , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/inmunología
12.
J Neuroendocrinol ; 6(1): 79-87, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8025570

RESUMEN

Estrogen is known to play an important role in regulating reproductive function in female rats through actions exerted at the preoptic area, a part of the brain that is markedly sexually dimorphic and which contains abundant estrogen receptors. A critical question to our understanding of estrogen's action on the brain is to identify the types of neurons that contain estrogen receptors (ER). Previous studies have shown that atrial natriuretic peptide (ANP) is in abundance in the preoptic area, and that ANP and other natriuretic peptides are capable of regulating gonadotropin secretion. In an effort to determine whether ERs are present in natriuretic peptide-immunoreactive (NP-ir) neurons in the preoptic area of the rat, double label immunocytochemistry was performed. Since ER-ir, as demonstrated with antibody H222 is known to be localized predominantly in cell nuclei, while NP-ir is present in the cytoplasm, single cells can be double labeled. Diaminobenzidine tetrahydrochloride was used for localization of NP-ir neurons, while nickel-enhanced diaminobenzidine tetrahydrochloride was used for localization of ER-ir. The results revealed that many nuclei throughout the preoptic area contained neurons that were ER-ir or NP-ir and that a substantial number were double labeled. Cell counts in selected preoptic nuclei and components, including the anteroventral periventricular nucleus, periventricular preoptic nucleus, medial part of the medial preoptic nucleus, and central part of the medial preoptic nucleus revealed that 13.6%, 11.1%, 13.5%, and 24.4%, respectively, of the NP-ir neurons in these nuclei also contained ER-ir. Collectively, a total of 14.9% of the NP-ir neurons in these nuclei also contained ER-ir.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Área Preóptica/metabolismo , Receptores del Factor Natriurético Atrial/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Factor Natriurético Atrial/inmunología , Femenino , Hipotálamo/citología , Hipotálamo/metabolismo , Inmunohistoquímica , Área Preóptica/citología , Ratas , Ratas Sprague-Dawley , Receptores del Factor Natriurético Atrial/inmunología , Receptores de Estrógenos/inmunología , Proteínas Recombinantes/inmunología
13.
Brain Res ; 631(2): 304-12, 1993 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-8131058

RESUMEN

The distribution of estrogen-receptor containing cells in the preoptic area, hypothalamus and amygdala of female Syrian hamster brain was studied by immunocytochemical methods. Dense populations of estrogen-receptor immunoreactive (ER-IR) cells were found in the medial preoptic area, the bed nucleus of the stria terminalis, amygdala, ventral and lateral parts of the hypothalamus, and the arcuate nucleus. Injection of estradiol caused a decrease in estrogen-receptor immunoreactivity (ERIR) containing cells within one hour, a decrease that may reflect a change in the ability of the occupied estrogen receptor to bind the particular antibody (H222) used rather than down-regulation of the estrogen receptor. Our findings on the distribution of estrogen-receptor containing cells in these areas using an immunocytochemical technique are consistent with and extend the findings of others using autoradiographic and in vitro binding techniques to study estrogen receptor distribution in hamster brain.


Asunto(s)
Encéfalo/metabolismo , Receptores de Estrógenos/metabolismo , Amígdala del Cerebelo/anatomía & histología , Amígdala del Cerebelo/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Encéfalo/anatomía & histología , Cricetinae , Estradiol/farmacología , Femenino , Hipotálamo/anatomía & histología , Hipotálamo/metabolismo , Inmunohistoquímica , Mesocricetus , Área Preóptica/anatomía & histología , Área Preóptica/metabolismo , Receptores de Estrógenos/inmunología
14.
J Neurobiol ; 22(2): 143-57, 1991 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-2030339

RESUMEN

The relative distributions of aromatase and of estrogen receptors were studied in the brain of the Japanese quail by a double-label immunocytochemical technique. Aromatase immunoreactive cells (ARO-ir) were found in the medial preoptic nucleus, in the septal region, and in a large cell cluster extending from the dorso-lateral aspect of the ventromedial nucleus of the hypothalamus to the tuber at the level of the nucleus inferioris hypothalami. Immunoreactive estrogen receptors (ER) were also found in each of these brain areas but their distribution was much broader and included larger parts of the preoptic, septal, and tuberal regions. In the ventromedial and tuberal hypothalamus, the majority of the ARO-ir cells (over 75%) also contained immunoreactive ER. By contrast, very few of the ARO-ir cells were double-labeled in the preoptic area and in the septum. More than 80% of the aromatase-containing cells contained no ER in these regions. This suggests that the estrogens, which are formed centrally by aromatization of testosterone, might not exert their biological effects through binding with the classical nuclear ER. The fact that significant amounts of aromatase activity are found in synaptosomes purified by differential centrifugation and that aromatase immunoreactivity is observed at the electron microscope level in synaptic boutons suggests that aromatase might produce estrogens that act at the synaptic level as neurohormones or neuromodulators.


Asunto(s)
Aromatasa/metabolismo , Encéfalo/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Aromatasa/inmunología , Encéfalo/anatomía & histología , Coturnix , Hipotálamo/anatomía & histología , Hipotálamo/metabolismo , Hipotálamo/ultraestructura , Inmunohistoquímica , Masculino , Microscopía Electrónica , Área Preóptica/anatomía & histología , Área Preóptica/metabolismo , Área Preóptica/ultraestructura , Receptores de Estrógenos/análisis , Receptores de Estrógenos/inmunología , Núcleo Hipotalámico Ventromedial/anatomía & histología , Núcleo Hipotalámico Ventromedial/metabolismo , Núcleo Hipotalámico Ventromedial/ultraestructura
15.
Histochemistry ; 96(2): 157-62, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1917572

RESUMEN

Application of Sternberger's unlabeled antibody enzyme method for detection of the estrogen receptor (ER) using a rat primary antibody with rat tissues has been discouraged, presumably because nonspecific staining of endogenous IgG was expected with the required anti-rat IgG bridging antibody. Because the blood-brain barrier greatly reduces immunoglobulin infiltration into the brain, we hypothesized that rat brain tissue could be specifically immunostained using rat IgG primary antibodies. A rat monoclonal anti-ER antibody (H222) specifically stained ERs in the brains of ovariectomized but not in ovariectomized estrogen-treated rats. In contrast, the uterus, a well-perfused target organ stained intensely in a nonspecific fashion. Dense populations of estrogen receptors were observed in the medial preoptic area, the bed nucleus of the stria terminalis, and the arcuate and ventromedial nuclei of the hypothalamus. A monoclonal rat IgG directed against alpha-tubulin labeled primarily cortical dendrites quite distinct from the neuronal nuclei that are the primary antigenic sites for the estrogen receptor antibody. These results confirm that the sensitive unlabeled antibody method can be applied to rat brain tissues, even when the primary antibody is rat IgG and that labeling of endogenous IgG may be used as a simple method to evaluate the integrity of the blood brain barrier.


Asunto(s)
Encéfalo/metabolismo , Inmunohistoquímica/métodos , Receptores de Estrógenos/análisis , Animales , Anticuerpos Monoclonales , Química Encefálica , Femenino , Hipotálamo/metabolismo , Ratas , Ratas Endogámicas , Receptores de Estrógenos/inmunología , Tubulina (Proteína)/análisis , Tubulina (Proteína)/inmunología , Útero/metabolismo
16.
Scand J Rheumatol ; 18(2): 69-76, 1989.
Artículo en Inglés | MEDLINE | ID: mdl-2786646

RESUMEN

It has now been recognized that there are complex interactions between the gonadal endocrine and the immune systems. The action of sex hormones on the immune system has important physiological and pathological consequences. The preponderance in women of autoimmune diseases in humans and in experimental animals has a basis in sex hormones. Hypoandrogenic/hyperestrogenic states are thought to contribute to the disease process. This article presents evidence for the action of sex hormones in various experimental animal models of autoimmune diseases and discusses several mechanisms of sex hormone action on the immune system. These mechanisms remain complex and it is to be hoped that the recent advances in immunology, endocrinology, pharmacology, and molecular biology will enable the description and clarification of these mechanisms of action.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Estrógenos/inmunología , Receptores Androgénicos , Testosterona/inmunología , Animales , Autoanticuerpos/análisis , Linfocitos B/inmunología , Bromelaínas/inmunología , Eritrocitos/inmunología , Femenino , Masculino , Ratones , Orquiectomía , Receptores de Estrógenos/inmunología , Receptores de Esteroides/inmunología , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA