Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Neurosci ; 40(41): 7965-7979, 2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-32887744

RESUMEN

Microglia, a type of CNS immune cell, have been shown to contribute to ethanol-activated neuronal death of the stress regulatory proopiomelanocortin (POMC) neuron-producing ß-endorphin peptides in the hypothalamus in a postnatal rat model of fetal alcohol spectrum disorders. We determined whether the microglial extracellular vesicle exosome is involved in the ethanol-induced neuronal death of the ß-endorphin neuron. Extracellular vesicles were prepared from hypothalamic tissues collected from postnatal rats (both males and females) fed daily with 2.5 mg/kg ethanol or control milk formula for 5 d or from hypothalamic microglia cells obtained from postnatal rats, grown in cultures for several days, and then challenged with ethanol or vehicle for 24 h. Nanoparticle tracking analysis and transmission electron microscopy indicated that these vesicles had the size range and shape of exosomes. Ethanol treatments increased the number and the ß-endorphin neuronal killing activity of microglial exosomes both in vivo and in vitro Proteomics analyses of exosomes of cultured microglial cells identified a large number of proteins, including various complements, which were elevated following ethanol treatment. Proteomics data involving complements were reconfirmed using quantitative protein assays. Ethanol treatments also increased deposition of the complement protein C1q in ß-endorphin neuronal cells in both in vitro and in vivo systems. Recombinant C1q protein increased while C1q blockers reduced ethanol-induced C3a/b, C4, and membrane attack complex/C5b9 formations; ROS production; and ultimately cellular death of ß-endorphin neurons. These data suggest that the complement system involving C1q-C3-C4-membrane attack complex and ROS regulates exosome-mediated, ethanol-induced ß-endorphin neuronal death.SIGNIFICANCE STATEMENT Neurotoxic action of alcohol during the developmental period is recognized for its involvement in fetal alcohol spectrum disorders, but the lack of clear understanding of the mechanism of alcohol action has delayed the progress in therapeutic intervention of this disease. Proopiomelanocortin neurons known to regulate stress, energy homeostasis, and immune functions are reported to be killed by developmental alcohol exposure because of activation of microglial immune cells in the brain. While microglia are known to use extracellular vesicles to communicate with neurons for maintaining homeostasis, we show here that ethanol exposure during the developmental period hijacks this system to spread apoptotic factors, including complement protein C1q, to induce the membrane attack complex and reactive super-oxygen species for proopiomelanocortin neuronal killing.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Complemento C1q/farmacología , Etanol/farmacología , Exosomas/efectos de los fármacos , Trastornos del Espectro Alcohólico Fetal/patología , Microglía/efectos de los fármacos , Proopiomelanocortina/genética , Animales , Animales Recién Nacidos , Muerte Celular/efectos de los fármacos , Células Cultivadas , Femenino , Trastornos del Espectro Alcohólico Fetal/metabolismo , Hipotálamo/metabolismo , Hipotálamo/patología , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Embarazo , Proteómica , Ratas , Ratas Sprague-Dawley , betaendorfina/metabolismo
2.
J Neuroinflammation ; 14(1): 83, 2017 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-28407740

RESUMEN

BACKGROUND: Opioid receptors are known to control neurotransmission of various peptidergic neurons, but their potential role in regulation of microglia and neuronal cell communications is unknown. We investigated the role of mu-opioid receptors (MOR) and delta-opioid receptors (DOR) on microglia in the regulation of apoptosis in proopiomelanocortin (POMC) neurons induced by neonatal ethanol in the hypothalamus. METHODS: Neonatal rat pups were fed a milk formula containing ethanol or control diets between postnatal days 2-6. Some of the alcohol-fed rats additionally received pretreatment of a microglia activation blocker minocycline. Two hours after the last feeding, some of the pups were sacrificed and processed for histochemical detection of microglial cell functions or confocal microscopy for detection of cellular physical interaction or used for gene and protein expression analysis. The rest of the pups were dissected for microglia separation by differential gradient centrifugation and characterization by measuring production of various activation markers and cytokines. In addition, primary cultures of microglial cells were prepared using hypothalamic tissues of neonatal rats and used for determination of cytokine production/secretion and apoptotic activity of neurons. RESULTS: In the hypothalamus, neonatal alcohol feeding elevated cytokine receptor levels, increased the number of microglial cells with amoeboid-type circularity, enhanced POMC and microglial cell physical interaction, and decreased POMC cell numbers. Minocycline reversed these cellular effects of alcohol. Alcohol feeding also increased levels of microglia MOR protein and pro-inflammatory signaling molecules in the hypothalamus, and MOR receptor antagonist naltrexone prevented these effects of alcohol. In primary cultures of hypothalamic microglia, both MOR agonist [D-Ala 2, N-MePhe 4, Gly-ol]-enkephalin (DAMGO) and ethanol increased microglial cellular levels and secretion of pro-inflammatory cell signaling proteins. However, a DOR agonist [D-Pen2,5]enkephalin (DPDPE) increased microglial secretion of anti-inflammatory cytokines and suppressed ethanol's ability to increase microglial production of inflammatory signaling proteins and secretion of pro-inflammatory cytokines. In addition, MOR-activated inflammation promoted while DOR-suppressed inflammation inhibited the apoptotic effect of ethanol on POMC neurons. CONCLUSIONS: These results suggest that ethanol's neurotoxic action on POMC neurons results from MOR-activated neuroinflammatory signaling. Additionally, these results identify a protective effect of a DOR agonist against the pro-inflammatory and neurotoxic action of ethanol.


Asunto(s)
Etanol/toxicidad , Microglía/metabolismo , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Receptores Opioides delta/fisiología , Receptores Opioides mu/fisiología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Células Cultivadas , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hipotálamo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas
3.
Cancer Prev Res (Phila) ; 8(1): 56-67, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25403848

RESUMEN

ß-Endorphin (BEP)-producing neuron in the hypothalamus plays a key role in bringing the stress axis to a state of homeostasis and maintaining body immune defense system. Long-term delivery of BEP to obtain beneficial effect on chemoprevention is challenging, as the peptides rapidly develop tolerance. Using rats as animal models, we show here that transplantation of BEP neurons into the hypothalamus suppressed carcinogens- and hormone-induced cancers in various tissues and prevented growth and metastasis of established tumors via activation of innate immune functions. In addition, we show that intracerebroventricular administration of nanosphere-attached dibutyryl cyclic adenosine monophosphate (dbcAMP) increased the number of BEP neurons in the hypothalamus, reduced the stress response, enhanced the innate immune function, and prevented tumor cell growth, progression, and metastasis. BEP neuronal supplementation did not produce any deleterious effects on general health but was beneficial in suppressing age-induced alterations in physical activity, metabolic, and immune functions. We conclude that the neuroimmune system has significant control over cancer growth and progression, and that activation of the neuroimmune system via BEP neuronal supplementation/induction may have therapeutic value for cancer prevention and improvement of general health.


Asunto(s)
Anticarcinógenos/uso terapéutico , Neoplasias/prevención & control , Neuronas/trasplante , betaendorfina/metabolismo , Animales , Bucladesina/química , Carcinógenos/química , Diferenciación Celular , Modelos Animales de Enfermedad , Femenino , Prueba de Tolerancia a la Glucosa , Hipotálamo/metabolismo , Sistema Inmunológico , Inmunohistoquímica , Células Asesinas Naturales/metabolismo , Masculino , Metástasis de la Neoplasia , Ratas , Ratas Endogámicas F344 , Ratas Desnudas , Ratas Sprague-Dawley
4.
Alcohol Clin Exp Res ; 38(12): 2988-97, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25581653

RESUMEN

BACKGROUND: Recently, retrograde tracing has provided evidence for an influence of hypothalamic ß-endorphin (BEP) neurons on the liver, but functions of these neurons are not known. We evaluated the effect of BEP neuronal activation on alcohol-induced liver injury and hepatocellular cancer. METHODS: Male rats received either BEP neuron transplants or control transplants in the hypothalamus and were randomly assigned to feeding alcohol-containing liquid diet or control liquid diet for 8 weeks or to treatment of a carcinogen diethylnitrosamine (DEN). Liver tissues of these animals were analyzed histochemically and biochemically for tissue injuries or cancer. RESULTS: Alcohol feeding increased liver weight and induced several histopathological changes such as prominent microvesicular steatosis and hepatic fibrosis. Alcohol feeding also increased the levels of triglyceride, hepatic stellate cell (HSC) activation factors, and catecholamines in the liver and endotoxin levels in the plasma. However, these effects of alcohol on the liver were reduced in animals with BEP neuron transplants. BEP neuron transplants also suppressed carcinogen-induced liver histopathologies such as extensive fibrosis, large focus of inflammatory infiltration, hepatocellular carcinoma (HCC), collagen deposition, numbers of preneoplastic foci, levels of HSC activation factors and catecholamines, as well as inflammatory milieu and increased the levels of natural killer cell cytotoxic factors in the liver. CONCLUSIONS: These findings are the first evidence for a role of hypothalamic BEP neurons in influencing liver functions. Additionally, the data identify that BEP neuron transplantation prevents hepatocellular injury and HCC formation possibly via influencing the immune function.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Modelos Animales de Enfermedad , Etanol/toxicidad , Hipotálamo/trasplante , Neoplasias Hepáticas/prevención & control , Neuronas/trasplante , betaendorfina , Animales , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Etanol/administración & dosificación , Femenino , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/patología , Masculino , Embarazo , Distribución Aleatoria , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley
5.
Alcohol Clin Exp Res ; 37(8): 1370-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23550806

RESUMEN

BACKGROUND: We have previously shown that ethanol (EtOH) increases cellular apoptosis to developing neurons via the effects on oxidative stress of neurons directly and via increasing production of microglia-derived factors. To study further the mechanism of EtOH action on neuronal apoptosis, we determined the effects of 2 well-known PKA activators, dibutyryl cAMP (dbcAMP) and brain-derived neurotrophic factor (BDNF), on EtOH-activated oxidative stress and apoptotic processes in the hypothalamic neurons in the presence and absence of microglial cells' influence. METHODS: In enriched neuronal cells from fetal rat hypothalami treated with EtOH or with conditioned medium from EtOH-treated microglia, we measured cellular apoptosis by the free nucleosome assay and the levels of cAMP, BDNF, O²â», reactive oxygen species (ROS), nitrite, glutathione (GSH), and catalase following treatment with EtOH or EtOH-treated microglial culture conditioned medium. Additionally, we tested the effectiveness of dbcAMP and BDNF in preventing EtOH or EtOH-treated microglial conditioned medium on cellular apoptosis and oxidative stress in enriched hypothalamic neuronal cell in primary cultures. RESULTS: Neuronal cell cultures following treatment with EtOH or EtOH-activated microglial conditioned medium showed decreased production levels of cAMP and BDNF. EtOH also increased apoptotic death as well as oxidative status, as demonstrated by higher cellular levels of oxidants but lower levels of antioxidants, in neuronal cells. These effects of EtOH on oxidative stress and cell death were enhanced by the presence of microglia. Treatment with BDNF or dbcAMP decreased EtOH or EtOH-activated microglial conditioned medium-induced changes in the levels of intracellular free radicals, ROS and O²â», nitrite, GSH, and catalase. CONCLUSIONS: These data support the possibility that EtOH by acting directly and via increasing the production of microglial-derived factors reduces cellular levels of cAMP and BDNF to increase cellular oxidative status and apoptosis in hypothalamic neuronal cells in primary cultures.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , AMP Cíclico/fisiología , Etanol/metabolismo , Hipotálamo/metabolismo , Microglía/fisiología , Animales , Antioxidantes/metabolismo , Apoptosis/inmunología , Células Cultivadas , Depresores del Sistema Nervioso Central/efectos adversos , Etanol/efectos adversos , Femenino , Trastornos del Espectro Alcohólico Fetal/etiología , Hipotálamo/efectos de los fármacos , Microglía/efectos de los fármacos , Estrés Oxidativo/inmunología , Embarazo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/fisiología
6.
Alcohol Clin Exp Res ; 37(2): 252-62, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22823548

RESUMEN

BACKGROUND: Animals exposed to alcohol during the developmental period develop many physiological and behavioral problems because of neuronal loss in various brain areas including the hypothalamus. Because alcohol exposure is known to induce oxidative stress in developing neurons, we tested whether hypothalamic cells from the fetal brain exposed to ethanol (EtOH) may alter the cell-cell communication between neurons and microglia, thereby leading to increased oxidative stress and the activation of apoptotic processes in the neuronal population in the hypothalamus. METHODS: Using enriched neuronal and microglial cells from fetal rat hypothalami, we measured cellular levels of various oxidants (O2 -, reactive oxygen species, nitrite), antioxidants (glutathione [GSH]), antioxidative enzymes (glutathione peroxidase [GSH-Px], catalase, superoxide dismutase) and apoptotic death in neurons in the presence and absence of EtOH or EtOH-treated microglial culture medium. Additionally, we tested the effectiveness of antioxidative agents in preventing EtOH or EtOH-treated microglial conditioned medium actions on oxidative stress and apoptosis in neuronal cell cultures. RESULTS: Neuronal cell cultures showed increased oxidative stress, as demonstrated by higher cellular levels of oxidants but lower levels of antioxidant and antioxidative enzymes, as well as, increased apoptotic death following treatment with EtOH. These effects of EtOH on oxidative stress and cell death were enhanced by the presence of microglia. Antioxidative agents protected developing hypothalamic neurons from oxidative stress and cellular apoptosis which is caused by EtOH or EtOH-treated microglial culture medium. CONCLUSIONS: These data suggest that exposure of developing hypothalamic neurons to EtOH increases cellular apoptosis via the effects on oxidative stress of neurons directly and via increasing production of microglial-derived factor(s).


Asunto(s)
Apoptosis/efectos de los fármacos , Etanol/efectos adversos , Feto/efectos de los fármacos , Hipotálamo/metabolismo , Microglía/metabolismo , Estrés Oxidativo/efectos de los fármacos , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Cromanos/farmacología , Medios de Cultivo Condicionados/farmacología , Relación Dosis-Respuesta a Droga , Etanol/antagonistas & inhibidores , Femenino , Feto/metabolismo , Hipotálamo/fisiopatología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Compuestos Organometálicos/farmacología , Embarazo , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Salicilatos/farmacología
7.
Cancer Res ; 71(19): 6282-91, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21835894

RESUMEN

Neurobehavioral stress has been shown to promote tumor growth and progression and dampen the immune system. In this study, we investigated whether inhibiting stress hormone production could inhibit the development of mammary carcinoma and metastasis in a rat model of breast carcinogenesis. To enhance ß-endorphin (BEP), the endogenous opioid polypeptide that boosts immune activity and decreases stress, we generated BEP neurons by in vitro differentiation from fetal neuronal stem cells and transplanted them into the hypothalami of rats subjected to breast carcinogenesis. BEP-transplanted rats displayed a reduction in mammary tumor incidence, growth, malignancy rate, and metastasis compared with cortical cells-transplanted rats. BEP neuron transplants also reduced inflammation and epithelial to mesenchymal transition in the tumor tissues. In addition, BEP neuron transplants increased peripheral natural killer (NK) cell and macrophage activities, elevated plasma levels of antiinflammatory cytokines, and reduced plasma levels of inflammatory cytokines. Antimetastatic effects along with stimulation of NK cells and macrophages could be reversed by treatment with the opiate antagonist naloxone, the ß-receptor agonist metaproterenol, or the nicotine acetylcholine receptor antagonist methyllycaconitine. Together, our findings establish a protective role for BEP against the growth and metastasis of mammary tumor cells by altering autonomic nervous system activities that enhance innate immune function.


Asunto(s)
Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/prevención & control , Neuronas/trasplante , betaendorfina/metabolismo , Corteza Suprarrenal/citología , Animales , Sistema Nervioso Autónomo , Diferenciación Celular , Células Cultivadas , Transición Epitelial-Mesenquimal , Femenino , Hipotálamo/inmunología , Hipotálamo/metabolismo , Hipotálamo/patología , Inmunidad Innata , Células Asesinas Naturales/inmunología , Activación de Macrófagos , Masculino , Neoplasias Mamarias Experimentales/patología , Clasificación del Tumor , Metástasis de la Neoplasia , Células-Madre Neurales/citología , Neuronas/inmunología , Neuronas/metabolismo , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley
8.
Alcohol Clin Exp Res ; 34(9): 1613-8, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20586752

RESUMEN

BACKGROUND: Ethanol exposure during early life has been shown to permanently alter the circadian expression of clock regulatory genes and the beta-endorphin precursor proopiomelanocortin (POMC) gene in the hypothalamus. Ethanol also alters the stress- and immune-regulatory functions of beta-endorphin neurons in laboratory rodents. Our aim was to determine whether the circadian clock regulatory Per2 gene modulates the action of ethanol on beta-endorphin neurons in mice. METHODS: Per2 mutant (mPer2(Brdml)) and wild type (C57BL/6J) mice were used to determine the effect of Per2 mutation on ethanol-regulated beta-endorphin neuronal activity during neonatal period using an in vitro mediobasal hypothalamic (MBH) cell culture model and an in vivo milk formula feeding animal model. The beta-endorphin neuronal activity following acute and chronic ethanol treatments was evaluated by measuring the peptide released from cultured cells or peptide levels in the MBH tissues, using enzyme-linked immunosorbent assay (ELISA). RESULTS: Per2 mutant mice showed a higher basal level of beta-endorphin release from cultured MBH cells and a moderate increase in the peptide content in the MBH in comparison with control mice. However, unlike wild type mice, Per2 mutant mice showed no stimulatory or inhibitory beta-endorphin-secretory responses to acute and chronic ethanol challenges in vitro. Furthermore, Per2 mutant mice, but not wild type mice, failed to show the stimulatory and inhibitory responses of MBH beta-endorphin levels to acute and chronic ethanol challenges in vivo. CONCLUSIONS: These results suggest for the first time that the Per2 gene may be critically involved in regulating beta-endorphin neuronal function. Furthermore, the data revealed an involvement of the Per2 gene in regulating beta-endorphin neuronal responses to ethanol.


Asunto(s)
Etanol/farmacología , Eliminación de Gen , Hipotálamo/metabolismo , Neuronas/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/fisiología , betaendorfina/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Femenino , Hipotálamo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL
9.
Proc Natl Acad Sci U S A ; 105(26): 9105-10, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18562281

RESUMEN

Pituitary adenylate cyclase-activating peptide (PACAP), a cAMP-activating agent, is highly expressed in the hypothalamus during the period when many neuroendocrine cells become differentiated from the neural stem cells (NSCs). Activation of the cAMP system in rat hypothalamic NSCs differentiated these cells into beta-endorphin (BEP)-producing neurons in culture. When these in vitro differentiated neurons were transplanted into the paraventricular nucleus (PVN) of the hypothalamus of an adult rat, they integrated well with the surrounding cells and produced BEP and its precursor gene product, proopiomelanocortin (POMC). Animals with BEP cell transplants demonstrated remarkable protection against carcinogen induction of prostate cancer. Unlike carcinogen-treated animals with control cell transplants, rats with BEP cell transplants showed rare development of glandular hyperplasia, prostatic intraepithelial neoplasia (PIN), or well differentiated adenocarcinoma with invasion after N-methyl-N-nitrosourea (MNU) and testosterone treatments. Rats with the BEP neuron transplants showed increased natural killer (NK) cell cytolytic function in the spleens and peripheral blood mononuclear cells (PBMCs), elevated levels of antiinflammatory cytokine IFN-gamma, and decreased levels of inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) in plasma. These results identified a critical role for cAMP in the differentiation of BEP neurons and revealed a previously undescribed role of these neurons in combating the growth and progression of neoplastic conditions like prostate cancer, possibly by increasing the innate immune function and reducing the inflammatory milieu.


Asunto(s)
Diferenciación Celular , AMP Cíclico/metabolismo , Neuronas/citología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , betaendorfina/metabolismo , Animales , Muerte Celular , Proliferación Celular , Células Cultivadas , Femenino , Hipotálamo/citología , Interferón gamma/biosíntesis , Células Asesinas Naturales/citología , Masculino , Metilnitrosourea , Neuronas/trasplante , Neoplasias de la Próstata/inducido químicamente , Ratas , Ratas Sprague-Dawley , Células Madre/citología , Testosterona , Factor de Necrosis Tumoral alfa/biosíntesis
10.
Endocrinology ; 148(6): 2828-34, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17347308

RESUMEN

Proopiomelanocortin-producing neurons in the arcuate nucleus of the hypothalamus secrete beta-endorphin (beta-EP), which controls varieties of body functions including the feedback regulation of the CRH neuronal activity in the paraventricular nucleus of the hypothalamus. Whether ethanol exposure in developing rats induces beta-EP neuronal death and alters their influence on CRH neurons in vivo has not been determined. We report here that binge-like ethanol exposures in newborn rats increased the number of apoptotic beta-EP neurons in the arcuate nucleus of the hypothalamus. We also found that immediately after ethanol treatments there was a significant reduction in the expression of proopiomelanocortin and adenylyl cyclases mRNA and an increased expression of several TGF-beta1-linked apoptotic genes in beta-EP neurons isolated by laser-captured microdissection from arcuate nuclei of young rats. Several weeks after the ethanol treatment, we detected a reduction in the number of beta-EP neuronal perikarya in arcuate nuclei and in the number of beta-EP neuronal terminals in paraventricular nuclei of the hypothalamus in the treated rats. Additionally, these rats showed increased response of the hypothalamic CRH mRNA to the lipopolysaccharide challenge. The ethanol-treated animals also showed incompetent ability to respond to exogenous beta-EP to alter the lipopolysaccharide-induced CRH mRNA levels. These data suggest that ethanol exposure during the developmental period causes beta-EP neuronal death by cellular mechanisms involving the suppression of cyclic AMP production and activation of TGF-beta1-linked apoptotic signaling and produces long-term structural and functional deficiency of beta-EP neurons in the hypothalamus.


Asunto(s)
Apoptosis/efectos de los fármacos , Etanol/toxicidad , Hipotálamo/efectos de los fármacos , Hipotálamo/crecimiento & desarrollo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Fisiológico/fisiopatología , betaendorfina/metabolismo , Animales , Animales Recién Nacidos , Proteínas Reguladoras de la Apoptosis/genética , Muerte Celular/efectos de los fármacos , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , AMP Cíclico/metabolismo , Femenino , Hipotálamo/metabolismo , Neuronas/fisiología , Terminales Presinápticos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/metabolismo , betaendorfina/fisiología
11.
Alcohol Clin Exp Res ; 30(11): 1925-32, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17067358

RESUMEN

BACKGROUND: Immune signals activate a network of cytokines in the central nervous system (CNS) that in turn causes release of neurotransmitters and hormones to modulate immune cell functions. We have recently shown that hypothalamic beta-endorphin neurons, via inhibition of the sympathetic neuronal activity, activate natural killer (NK) cell function in the spleen, and this communication is disrupted following chronic ethanol administration. Beta-endorphin neuronal function is known to be regulated by various proinflammatory and anti-inflammatory cytokines. The effects of ethanol on the proinflammatory and anti-inflammatory cytokines known to control beta-endorphin neuronal and NK cell functions during immune challenges have not been determined. METHODS: In the present study, we evaluated the effects of chronic ethanol consumption on the basal and lipopolysaccharide (LPS)-activated NK cells' functions in the spleen, the beta-endorphin peptide precursor proopiomelanocortin (POMC) gene expression in the arcuate nucleus (ARC) of the hypothalamus, and mRNA levels of proinflammatory cytokines interleukin-1beta (IL-1beta), tumor necrosis factor alpha (TNF-alpha), and anti-inflammatory cytokines IL-6 and IL-10 in the spleen and in the ARC. Male rats were ad libitum fed rat chow (ad lib-fed), pair-fed an isocaloric liquid diet, or fed an ethanol-containing liquid diet, and each was treated with LPS (100 microg/kg body weight). After 2 hours, splenocytes and ARC tissues were isolated and used for this study. Splenocytes were used to determine mRNA levels of IL-1beta, TNF-alpha, IL-6, IL-10, granzyme B, and perforin using the real-time RT-PCR assays. Splenocytes were also used to determine the cytolytic activity using a standard 4-hour (51)Cr release assay against YAC-1 lymphoma target cells. Arcuate nuclei were used to determine IL-1beta, TNF-alpha, IL-6, IL-10, and POMC mRNA levels using real-time RT-PCR assays. RESULTS: The results demonstrate that ethanol feeding via a liquid diet for 2 weeks suppressed both basal and LPS-stimulated NK cell cytolytic functions and the levels of cytotoxicity-regulatory perforin and granzyme B mRNAs in the spleen. Ethanol feeding reduced the basal and LPS-stimulated levels of POMC mRNA in the ARC. Ethanol also impaired LPS-induced levels of IL-1beta and TNF-alpha mRNAs both in the spleen and in the ARC. In contrast, ethanol feeding did not cause any significant changes in basal and the LPS-stimulated expression of IL-6 and IL-10 mRNAs in the spleen and of IL-6 mRNA levels in the ARC. These results indicate that ethanol suppression of hypothalamic POMC levels and splenic NK cell functions is associated with a reduced expression of proinflammatory cytokines in neuroendocrine and immune cells.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Citocinas/biosíntesis , Etanol/farmacología , Hipotálamo/metabolismo , Inmunidad Celular/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Sistemas Neurosecretores/metabolismo , Proopiomelanocortina/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Citocinas/antagonistas & inhibidores , Hipotálamo/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/metabolismo , Inyecciones Intraperitoneales , Células Asesinas Naturales/efectos de los fármacos , Lipopolisacáridos/farmacología , Masculino , Sistemas Neurosecretores/citología , Sistemas Neurosecretores/efectos de los fármacos , ARN Mensajero/biosíntesis , Ratas , Ratas Endogámicas F344 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/citología , Bazo/efectos de los fármacos , Bazo/metabolismo
12.
Mol Pharmacol ; 69(3): 706-17, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16326933

RESUMEN

The mechanism by which ethanol induces beta-endorphin (beta-EP) neuronal death during the developmental period was determined using fetal rat hypothalamic cells in primary cultures. The addition of ethanol to hypothalamic cell cultures stimulated apoptotic cell death of beta-EP neurons by increasing caspase-3 activity. Ethanol lowered the levels of adenylyl cyclase (AC)7 mRNA, AC8 mRNA, and/or cAMP in hypothalamic cells, whereas a cAMP analog blocked the apoptotic action of ethanol on beta-EP neurons. The AC inhibitor dideoxyadenosine (DDA) increased cell apoptosis and reduced the number of beta-EP neurons, and it potentiated the apoptotic action of ethanol on these neurons. beta-EP neurons in hypothalamic cultures showed immunoreactivity to transforming growth factor-beta1 (TGF-beta1) protein. Ethanol and DDA increased TGF-beta1 production and/or release from hypothalamic cells. A cAMP analog blocked the activation by ethanol of TGF-beta1 in these cells. TGF-beta1 increased apoptosis of beta-EP neurons, but it did not potentiate the action of ethanol or DDA actions on these neurons. TGF-beta1 neutralizing antibody blocked the apoptotic action of ethanol on beta-EP neurons. Determination of TGF-beta1-controlled cell apoptosis regulatory gene levels in hypothalamic cell cultures and in isolated beta-EP neurons indicated that ethanol, TGF-beta1, and DDA similarly alter the expression of these genes in these cells. These data suggest that ethanol increases beta-EP neuronal death during the developmental period by cellular mechanisms involving, at least partly, the suppression of cAMP production and activation of TGF-beta1-linked apoptotic signaling.


Asunto(s)
Apoptosis , AMP Cíclico/metabolismo , Etanol/toxicidad , Hipotálamo/efectos de los fármacos , Hipotálamo/embriología , Neuronas/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Inhibidores de Adenilato Ciclasa , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Animales , Apoptosis/genética , Caspasa 3 , Caspasas/metabolismo , Células Cultivadas , Didesoxiadenosina/farmacología , Desarrollo Embrionario/efectos de los fármacos , Femenino , Hipotálamo/citología , Neuronas/metabolismo , Embarazo , Ratas , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1 , betaendorfina/metabolismo
13.
Alcohol Clin Exp Res ; 29(4): 648-55, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15834231

RESUMEN

BACKGROUND: beta-Endorphin (beta-EP) neurons are involved in ethanol's action on a variety of brain functions, including positive reinforcement. These neurons are innervated by vasoactive intestinal peptide (VIP)-containing and corticotropin-releasing hormone (CRH)-containing neurons in the hypothalamus. Whether these neuropeptides affect beta-EP neuronal function in the presence or absence of ethanol has not previously been determined. METHODS: The authors determined the effects of VIP and CRH on gene expression and peptide release from beta-EP neurons in primary cultures of mediobasal hypothalamic cells. The effects of receptor antagonists on VIP- and CRH-induced beta-EP release was determined. Furthermore, the authors studied the effects of acute and chronic treatment with ethanol on the response of beta-EP neurons to VIP and CRH. Real-time reverse-transcription polymerase chain reaction was used for messenger RNA (mRNA) detection, and radioimmunoassay was used for hormone measurements. RESULTS: We show that beta-EP neurons responded concentration dependently to VIP and CRH treatments by increasing both beta-EP release and proopiomelanocortin mRNA expression. Simultaneous treatment with a nonspecific receptor antagonist reduced the ability of CRH or VIP to induce beta-EP release from mediobasal hypothalamic cells. Acute treatment with ethanol increased beta-EP neuronal gene expression and the secretory response to CRH and VIP. However, previous exposure to chronic ethanol reduced the CRH and VIP responses of these neurons. CONCLUSIONS: These results indicate that VIP and CRH stimulate beta-EP release from hypothalamic cells in primary cultures and that the stimulatory and adaptive responses of beta-EP neurons to ethanol may involve alteration in the responsiveness of beta-EP-secreting neurons to CRH and VIP.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Hormona Liberadora de Corticotropina/farmacología , Etanol/farmacología , Hipotálamo/metabolismo , Proopiomelanocortina/biosíntesis , ARN Mensajero/biosíntesis , Péptido Intestinal Vasoactivo/farmacología , betaendorfina/metabolismo , Animales , Células Cultivadas , Hormona Liberadora de Corticotropina/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Embarazo , Proopiomelanocortina/metabolismo , ARN/biosíntesis , ARN/aislamiento & purificación , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Péptido Intestinal Vasoactivo/administración & dosificación
14.
Alcohol Clin Exp Res ; 28(8): 1180-6, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15318116

RESUMEN

BACKGROUND: We have previously shown that ethanol administration suppresses natural killer (NK) cell cytolytic activity, partly by decreasing the action of hypothalamic beta-endorphin (beta-EP) on the spleens of male Fischer-344 rats. This study was conducted to examine the effects of ethanol and central administration of beta-EP on perforin, granzyme B, and the cytokine interferon (IFN)-gamma--factors that modulate NK cell cytolytic activity--to understand the mechanism involved in ethanol's suppression of NK cell activity. METHODS: A group of male Fischer-344 rats were fed an ethanol-containing diet (8.7% v/v), and a control group was pair-fed an isocaloric diet. At the end of 2 weeks, both groups were infused with beta-EP 100 ng/hr into the paraventricular nucleus of the hypothalamus for 18 hr, and spleen tissues were immediately removed for analysis of perforin, granzyme B, and IFN-gamma messenger RNA (mRNA) and protein levels. The mRNA levels of perforin, granzyme B, and IFN-gamma were evaluated by quantitative real-time polymerase chain reaction, and the protein levels of perforin and granzyme B were analyzed by Western blot. RESULTS: Paraventricular nucleus administration of beta-EP increased the mRNA and protein expression of granzyme B and mRNA expression of IFN-gamma in pair-fed animals. Ethanol significantly reduced both basal and beta-EP-induced levels of granzyme B and IFN-gamma. CONCLUSIONS: These data suggest that chronic ethanol consumption suppresses beta-EP-induced NK cytolytic activity, granzyme B, and IFN-gamma in male Fischer-344 rats.


Asunto(s)
Citotoxicidad Inmunológica/efectos de los fármacos , Etanol/farmacología , Hipotálamo/efectos de los fármacos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , betaendorfina/farmacología , Animales , Hipotálamo/metabolismo , Células Asesinas Naturales/metabolismo , Masculino , ARN Mensajero/biosíntesis , Ratas , Ratas Endogámicas F344
15.
Alcohol Clin Exp Res ; 27(11): 1813-8, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14634498

RESUMEN

BACKGROUND: Nitric oxide (NO) mediates many pharmacological actions of ethanol. NO's role in regulating ethanol action on hypothalamic beta-endorphin (beta-EP) neurons is not established. METHODS: In this study, we determined the role of NO in ethanol regulation of beta-EP release from primary cultures of rat fetal mediobasal hypothalamic cells. Real-time polymerase chain reaction was used for messenger RNA (mRNA) detection; radioimmunoassay was used for hormone measurements. RESULTS: Acute ethanol treatment for 3 hr increased the release of beta-EP but reduced nitrite levels in the media of hypothalamic cells in primary cultures. In contrast, ethanol exposure for 48 hr reduced the release of beta-EP but increased the release of nitrite from these cells. Alcohol treatments altered the expression of neuronal NO synthase mRNA, but not inducible NO synthase mRNA, in a pattern similar to that of nitrite levels. Alcohol treatments blocked sodium nitroprusside-induced increases in the level of cellular cyclic guanidine monophosphate. The nonspecific NO blocker NG-nitro-l-arginine-methyl-esther, but not the inactive isomer N-nitro-d-arginine-methyl-esther (d-NAME), inhibited ethanol inhibitory actions on beta-EP release. CONCLUSIONS: These results suggest that the cyclic guanidine monophosphate/NO pathway is involved in ethanol alteration of hypothalamic beta-EP release.


Asunto(s)
Etanol/farmacología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Óxido Nítrico/fisiología , betaendorfina/metabolismo , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ratas
16.
J Pharmacol Exp Ther ; 301(1): 119-28, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11907165

RESUMEN

We have previously shown that short-term exposure to ethanol stimulates immunoreactive beta-endorphin (IR-beta-EP) release from hypothalamic neurons and that chronic ethanol exposure decreases the IR-beta-EP release from these neurons. The role of protein kinase C (PKC) in the ethanol-regulated beta-EP release from hypothalamic neurons has not been established. In this study, by using the primary cultures of hypothalamic neurons, we tested the effects of PKC stimulator phorbol ester 4 beta-phorbol 12-myristate-13-acetate (PMA) and PKC inhibitor chelerythrine chloride on ethanol-induced IR-beta-EP release. Additionally, the effects of ethanol with or without PMA on expression and translocation of various PKC isoenzymes from cytosolic to membrane fraction were determined. PMA treatment increased IR-beta-EP release in a time- and dose-dependent manner. Acute ethanol treatment (3 h) increased, while chronic ethanol treatment (24 h) reduced, the magnitude of PMA-induced IR-beta-EP release. The stimulatory effect of acute ethanol on IR-beta-EP release was reduced by chelerythrine chloride. Determination of the effects of ethanol with or without PMA on seven different PKC isoenzymes (PKC-alpha, -beta I, -beta II, -gamma, -delta, -epsilon, and -zeta) revealed that the expression and translocation of only two PKC isoenzymes, PKC-delta and PKC-epsilon, were stimulated by acute treatment with ethanol. Acute ethanol also increased PMA-stimulated expression of these two isoenzymes. Chronic ethanol treatment reduced both basal and PMA-induced increase of PKC-delta and PKC-epsilon expression and translocation. These data provide evidence for the first time that ethanol-regulated IR-beta-EP secretion is controlled by the PKC system, possibly involving PKC-delta and PKC-epsilon isoenzymes.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Hipotálamo/metabolismo , Neuronas/metabolismo , Proteína Quinasa C/fisiología , betaendorfina/metabolismo , Alcaloides , Animales , Benzofenantridinas , Western Blotting , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Células Cultivadas , Citosol/efectos de los fármacos , Citosol/enzimología , Inhibidores Enzimáticos/farmacología , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/fisiología , Neuronas/efectos de los fármacos , Fenantridinas/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA