Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 170(4): 1261-9, 2010 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-20732393

RESUMO

Although most agree that 17ß-estradiol is neuroprotective via a variety of mechanisms, less is known about the role that biological sex plays in receptor-mediated estradiol neuroprotection. To address this issue we isolated primary cortical neurons from rat pups sorted by sex and assessed the ability of estradiol to protect the neurons from death induced by glutamate. Five-minute pretreatment with 10-50 nM 17ß-estradiol protected female but not male neurons from glutamate toxicity 24 h later. Both estrogen receptor alpha (ERα) and estrogen receptor beta (ERß) are expressed in these cultures. Experiments using an ERα selective agonist or antagonist indicate that this receptor is important for neuroprotection in female cortical neurons. The ERß selective agonist conveys a small degree of neuroprotection to both male and female cortical neurons. Interestingly, we found that 17α estradiol and the novel membrane estrogen receptor (mER) agonist STX, but not bovine serum albumin conjugated estradiol or the GPR30 agonist G1 were neuroprotective in both male and female neurons. Taken together these data highlight a role for ERα in sexually dimorphic neuroprotection.


Assuntos
Estradiol/farmacologia , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Ácido Glutâmico/fisiologia , Neurônios/citologia , Fármacos Neuroprotetores/farmacologia , Caracteres Sexuais , Acrilamidas/farmacologia , Animais , Células Cultivadas , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Estrogênios/farmacologia , Feminino , Ácido Glutâmico/toxicidade , Ligantes , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fenóis , Pirazóis/farmacologia , Ratos
2.
Neuroscience ; 153(3): 751-61, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18406537

RESUMO

Estrogen receptors can activate transcription in the nucleus, and activate rapid signal transduction cascades in the cytosol. Multiple reports identify estrogen receptors at the plasma membrane, while others document the dynamic responses of estrogen receptor to ligand binding. However, the function and identity of membrane estrogen receptors remain controversial. We have used confocal microscopy and cell fractionation on the murine hippocampus-derived HT22 cell line and rat primary cortical neurons transfected with estrogen receptor-green fluorescent protein constructs to address the membrane localization of these receptors. We observe translocation of estrogen receptor beta (beta) to the plasma membrane 5 min after exposure to 17beta-estradiol, whereas estrogen receptor alpha (alpha) localization remains unchanged. Membrane localization of estrogen receptor beta is transient, selective for 17beta-estradiol, and is not blocked by ICI182,780. Inhibition of the mitogen-activated protein kinase pathway does not block estrogen-mediated estrogen receptor beta membrane translocation, and in fact prolongs membrane localization. These data suggest that while both estrogen receptor alpha and estrogen receptor beta can be present at the neuronal membrane, their presence is differentially regulated.


Assuntos
Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Neurônios/efeitos dos fármacos , Animais , Western Blotting , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Receptor alfa de Estrogênio/efeitos dos fármacos , Receptor beta de Estrogênio/efeitos dos fármacos , Imuno-Histoquímica , Camundongos , Microscopia Confocal , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neurônios/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos
3.
Minerva Endocrinol ; 32(2): 87-94, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17557034

RESUMO

Estrogen is neuroprotective in a large number of models in vivo and in vitro. Its application in hormone replacement therapy has proven to be more complicated, necessitating better understanding of how estrogen signals in the brain. Estrogen binds to estrogen receptors to regulate gene transcription, and activates a number of rapid signaling cascades from the plasma membrane. These rapid signaling cascades have been shown to play important roles in mediating the neuroprotective effects of estrogen. This review covers evidence that understanding and targeting the membrane effects of estrogen has emerged as an important area in the design of novel neuroprotective drugs.


Assuntos
Encéfalo/efeitos dos fármacos , Membrana Celular/fisiologia , Estrogênios/fisiologia , Fármacos Neuroprotetores/farmacologia , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Transdução de Sinais/fisiologia , Doença de Alzheimer/tratamento farmacológico , Animais , Encéfalo/fisiologia , Estrogênios/farmacologia , Humanos , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia
4.
Endocrine ; 32(3): 317-28, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18247162

RESUMO

The regimen of estrogen replacement can alter the consequences of estrogen therapy and stressors. To determine the long-term effects and interaction of these systems on the brain and periphery, adult female rats were infused with lipopolysaccharide (LPS) into the fourth ventricle of the brain for 4 weeks, and ovariectomized rats were administered either constant or pulsed regimens of estrogen replacement (17beta-estradiol) until sacrifice at 8 weeks. Constant, but not pulsed, estrogen replacement reduced ERalpha and increased HSP90, HSP70, and PR(B) uterine protein levels. Both estrogen regimens increased ERbeta, HSP27, and PR(A) uterine proteins. Both regimens reduced hypothalamic levels of ERalpha, but not ERbeta, HSP, or PR. No changes were observed in the hippocampus. Long-term brain infusion of LPS activated microglia and reduced body weight, but did not alter corticosterone or nitrotyrosine levels. LPS infusion into intact rats suppressed uterine weight, increased ERalpha and decreased HSP90 in the uterus. LPS did not alter uterine weight in ovariectomized rats treated with constant or pulsed estrogen. Together, these data suggest the timing of estrogen replacement and neuroinflammatory stressors can profoundly affect uterine and hypothalamic steroid receptor expression and may be important parameters to consider in the post-menopausal intervention with estrogen.


Assuntos
Encéfalo/efeitos dos fármacos , Terapia de Reposição de Estrogênios , Hipotálamo/metabolismo , Lipopolissacarídeos/farmacologia , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Útero/metabolismo , Animais , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Proteínas de Choque Térmico/metabolismo , Injeções Intraventriculares , Lipopolissacarídeos/administração & dosagem , Ratos , Ratos Endogâmicos F344
5.
Endocrinology ; 148(1): 232-40, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17023524

RESUMO

The effects of estrogen therapy can differ depending on the regimen of estrogen administration. In addition, estrogen can modulate the effects of stressors. To examine the interaction between these systems, we infused adult female rats with lipopolysaccharide (LPS) into the fourth ventricle of the brain for 6 d and compared the effects of constant and pulsed estrogen replacement. Constant, but not pulsed, estrogen treatment reduced estrogen receptor-alpha (ERalpha) protein by 90% in the uterus and increased heat-shock proteins 70 and 90 by 74 and 48%, respectively, whereas progesterone receptor levels increased in all ovariectomized rats receiving estrogen replacement. In contrast to the uterine decline in ERalpha, no changes in ERalpha were observed in the hypothalamus or hippocampus, and ERbeta levels were unchanged in all regions tested. Brain infusion of LPS did not alter these proteins but increased the number of activated microglia in the thalamus and reduced body weight in all rats as well as activated the hypothalamic-pituitary-adrenal axis in ovariectomized rats, as determined by elevations in circulating corticosterone and progesterone. Estrogen treatments did not alter these markers, and no differences were observed in cortical choline acetyltransferase activity or nitrotyrosine for any of the treatment groups. The current study found an unexpected increase in uterine weight in lipopolysaccharide-infused rats treated with constant, but not pulsed, estrogen. This report suggests that constant and pulsed regimens of estrogen administration produce different effects and that stress may be an important factor in the postmenopausal intervention with estrogen.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Lipopolissacarídeos/farmacologia , Útero/efeitos dos fármacos , Útero/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Química Encefálica/efeitos dos fármacos , Interações Medicamentosas , Terapia de Reposição de Estrogênios/métodos , Estrogênios/sangue , Feminino , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Injeções Intraventriculares , Tamanho do Órgão/efeitos dos fármacos , Ovariectomia , Progesterona/sangue , Pulsoterapia , Ratos , Ratos Endogâmicos F344 , Receptores de Progesterona/metabolismo , Estresse Fisiológico/imunologia , Útero/citologia
6.
Neuroscience ; 138(3): 851-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16343790

RESUMO

The aging process is known to coincide with a decline in circulating sex hormone levels in both men and women. Due to an increase in the average lifespan, a growing number of post-menopausal women are now receiving hormone therapy for extended periods of time. Recent findings of the Women's Health Initiative, however, have called into question the benefits of long-term hormone therapy for treating symptoms of menopause. The results of this study are still being evaluated, but it is clear that a better understanding of the molecular effects of estradiol is needed in order to develop new estrogenic compounds that activate specific mechanisms but lack adverse side effects. Traditionally, the effects of estradiol treatment have been ascribed to changes in gene expression, namely transcription at estrogen response elements. This review focuses on emerging information that estradiol can also activate a repertoire of membrane-initiated signaling pathways and that these rapid signaling events lead to functional changes at the cellular level. The various types of cells in the brain can respond differently to estradiol treatment based on the signaling properties of the cell, as well as which receptor, estrogen receptor alpha and/or estrogen receptor beta, is expressed. Taken together, these findings suggest that the estradiol-induced activation of membrane-initiated signaling pathways occurs in a cell-type specific manner and can differentially influence how the cells respond to various insults.


Assuntos
Encéfalo/fisiologia , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Estrogênios/fisiologia , Neuroglia/fisiologia , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Animais , Regulação da Expressão Gênica , Humanos , Cinética , Transcrição Gênica
7.
Neuroscience ; 133(1): 343-52, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15893655

RESUMO

Physiological doses of 17-beta Estradiol (E2) rapidly induce mitogen-activated protein kinase (MAPK) phosphorylation in a variety of cell culture and tissue explant preparations. Rapid MAPK phosphorylation has been implicated as a critical step in estrogen's effects on neuronal activity, gene transcription and neuroprotection. The present series of in vivo experiments were designed to determine whether acute administration of estrogen rapidly increased extracellular signal-regulated protein kinase (ERK) 2 phosphorylation. Brains were harvested 20 min after a single i.p. injection of 15 microg/kg of 17-beta or 17-alpha estradiol. Twelve brain structures were micro-dissected, homogenized and processed for Western blotting. E2-treated rats exhibited a statistically significant increase in ERK2 phosphorylation in the diagonal band of Broca, rostral nucleus accumbens, paraventricular nucleus, arcuate nucleus and anteromedial visual cortex. Administration of the same dose of 17-alpha estradiol did not enhance ERK phosphorylation in any of the brain regions examined. The in vivo data presented here extend previously published in vitro data indicating that E2 rapidly activates MAPK in primary neuronal cultures, explants and cell lines. These data also indicate that MAPK activation is a potential mediator of estrogens effects in some but not all estrogen receptor containing regions of the brain.


Assuntos
Encéfalo/enzimologia , Estradiol/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Algoritmos , Animais , Western Blotting , Encéfalo/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Estradiol/administração & dosagem , Estrogênios/sangue , Feminino , Ovariectomia , Fosforilação , Área Pré-Óptica/fisiologia , Ratos
8.
J Pharmacol Exp Ther ; 299(1): 67-75, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11561064

RESUMO

Genistein is a potent plant-derived isoflavone displaying estrogenic activity at low (nanomolar) concentrations and antiproliferative and antiangiogenic properties at higher concentrations (above 10-50 microM). The antiproliferative potential of genistein has made it an interesting candidate for cancer chemotherapy at high concentrations; however, the potential for genistein toxicity in neurons at such concentrations has not been previously addressed. We show that genistein is toxic to rat primary cortical neurons at a concentration of 50 microM, whereas daidzein, a structural analog, shows no toxicity at similar concentrations. The dying cells display an apoptotic morphology that is characterized by fragmented nuclei, appearance of apoptotic bodies, DNA laddering, and caspase-dependent poly(ADP-ribose) polymerase cleavage. This cell death is partially dependent on caspase activity, independent of estrogen receptors, and does not result in a significant loss of Bcl-2 or Bcl-X(L) protein. Genistein exposure induces delayed and prolonged activation of p42/44 mitogen-activated protein kinase (MAPK) and p38 MAPK but not c-Jun NH2-terminal kinase. The specific p42/44 MAPK kinase inhibitor PD98059 (50 microM) partially blocks genistein-induced apoptosis, whereas the p38 MAPK inhibitor SB202190 (10 microM) has no effect. Genistein elevates intracellular calcium and both 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester (1 microM) and dantrolene (10 microM) inhibit genistein-induced apoptosis, suggesting a link between genistein-induced intracellular calcium release and apoptosis. The combination of dantrolene and PD98059 block genistein-induced apoptosis in an additive manner compared with either compound alone. These findings provide evidence for a proapoptotic function of p42/44 MAPK and raise caution about potential side effects in the nervous system with genistein use as a high-dose therapeutic agent.


Assuntos
Anticarcinógenos/farmacologia , Apoptose/fisiologia , Cálcio/fisiologia , Genisteína/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Neurônios/fisiologia , Animais , Western Blotting , Caspases/metabolismo , Córtex Cerebral/citologia , Fragmentação do DNA/efeitos dos fármacos , Genes bcl-2/genética , Técnicas In Vitro , Indicadores e Reagentes , L-Lactato Desidrogenase/metabolismo , Microscopia de Contraste de Fase , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/metabolismo
9.
Proc Natl Acad Sci U S A ; 98(18): 10451-6, 2001 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-11517332

RESUMO

Motor stereotypies are abnormally repetitive behaviors that can develop with excessive dopaminergic stimulation and are features of some neurologic disorders. To investigate the mechanisms required for the induction of stereotypy, we examined the responses of dopamine-deficient (DD) mice to increasing doses of the dopamine precursor L-DOPA. DD mice lack the ability to synthesize dopamine (DA) specifically in dopaminergic neurons yet exhibit robust hyperlocomotion relative to wild-type (WT) mice when treated with L-DOPA, which restores striatal DA tissue content to approximately 10% of WT levels. To further elevate brain DA content in DD mice, we administered the peripheral L-amino acid decarboxylase inhibitor carbidopa along with L-DOPA (C/l-DOPA). When striatal DA levels reached >50% of WT levels, a transition from hyperlocomotion to intense, focused stereotypy was observed that was correlated with an induction of c-fos mRNA in the ventrolateral and central striatum as well as the somatosensory cortex. WT mice were unaffected by C/L-DOPA treatments. A D1, but not a D2, receptor antagonist attenuated both the C/L-DOPA-induced stereotypy and the c-fos induction. Consistent with these results, stereotypy could be induced in DD mice by a D1, but not by a D2, receptor agonist, with neither agonist inducing stereotypy in WT mice. Intrastriatal injection of a D1 receptor antagonist ameliorated the stereotypy and c-fos induction by C/L-DOPA. These results indicate that activation of D1 receptors on a specific population of striatal neurons is required for the induction of stereotypy in DD mice.


Assuntos
Corpo Estriado/fisiologia , Dopamina/deficiência , Receptores de Dopamina D1/fisiologia , Comportamento Estereotipado/fisiologia , Animais , Benzazepinas/farmacologia , Corpo Estriado/efeitos dos fármacos , Dopamina/fisiologia , Antagonistas dos Receptores de Dopamina D2 , Feminino , Genes fos , Haloperidol/farmacologia , Levodopa/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D1/efeitos dos fármacos , Comportamento Estereotipado/efeitos dos fármacos
10.
Endocrinology ; 142(6): 2336-42, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11356680

RESUMO

The rapid, nongenomic effects of estrogen are increasingly recognized as playing an important role in several aspects of estrogen action. Rapid activation of the mitogen-activated protein kinase (MAPK) signaling pathway by estrogen is among the more recently identified of these effects. To explore the role of estrogen receptors (ERs) in mediating these effects, we have transfected ER-negative Rat-2 fibroblasts with complementary DNA clones encoding either human ERalpha or rat ERbeta and examined their ability to couple to activation of MAPK in response to 17beta-estradiol (17beta-E(2)) and other ligands. For both receptors, addition of E(2) resulted in a rapid phosphorylation of MAPK. Activation of MAPK in ERalpha-transfected cells was partially and completely blocked by the antiestrogens tamoxifen and ICI 182,780, respectively. In ERbeta-transfected cells, MAPK activation was less sensitive to inhibition by tamoxifen and ICI 182,780. We have also observed that, in this model system, a membrane-impermeable estrogen (BSA-E(2)) and 17alpha-E(2) were both able to activate MAPK in a manner similar to E(2) alone. Here also, ICI 182,780 blocked the ability of BSA-E(2) to activate MAPK through ERalpha, but failed to block ERbeta-mediated effects. BSA-E(2) treatment, however, failed to activate nuclear estrogen-response-element-mediated gene transcription. These data show that these nuclear ERs are necessary for estrogen's effects at the membrane. This model system will be useful in identifying molecular interactions involved in the rapid effects mediated by the ERs.


Assuntos
Estradiol/análogos & derivados , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores de Estrogênio/fisiologia , Animais , Western Blotting , Linhagem Celular , Sinergismo Farmacológico , Ativação Enzimática , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Fibroblastos/química , Fibroblastos/metabolismo , Fulvestranto , Humanos , Fosforilação , Ratos , Receptores de Estrogênio/análise , Receptores de Estrogênio/genética , Tamoxifeno/farmacologia , Transfecção
11.
J Neurosci ; 21(7): 2546-52, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11264328

RESUMO

The oxytocin receptor (OTR) is differentially expressed in the CNS. Because there are multiple mechanisms by which the OTR can be transcriptionally induced, we hypothesized that differences in OTR expression may be explained by activation of distinct signal transduction pathways and may be critical for the control of anxiety and sex behaviors. To determine the regulation and functional significance of this expression, we infused female rats with modifiers of protein kinases before assaying for behavior and oxytocin receptor binding. In the ventromedial nucleus of the hypothalamus (VMH), estrogen-dependent induction of oxytocin receptors required protein kinase C activation, and oxytocin infused here promoted female sex behavior but had no effect on anxiety. In contrast, dopamine controlled tonic oxytocin receptor expression in the central nucleus of the amygdala (cAmyg) through activation of protein kinase A, and oxytocin infused here was anxiolytic but had no effect on female sex behavior. Therefore, we have identified brain region-specific regulation of the OTR in the VMH and cAmyg. Distinct signal transduction pathways regulating receptor expression and binding in each brain region may mediate in part the ability of oxytocin to exert these differential behavioral effects.


Assuntos
Ansiedade/metabolismo , Encéfalo/metabolismo , Receptores de Ocitocina/metabolismo , Comportamento Sexual Animal/efeitos dos fármacos , Comportamento Sexual Animal/fisiologia , Animais , Ansiedade/fisiopatologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Ocitocina/administração & dosagem , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Ocitocina/efeitos dos fármacos , Receptores de Ocitocina/fisiologia , Transdução de Sinais/fisiologia
12.
Endocrinology ; 141(11): 4056-64, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11089536

RESUMO

Neuronal expression of vasopressin messenger RNA (mRNA) and peptide has been shown to be estrogen dependent. A 5.5-kb genomic DNA fragment, 5' of the AVP coding region, was used in luciferase reporter assays to measure transcriptional activation by either estrogen receptor alpha or beta in response to various treatments. ER alpha and ER beta displayed differential regulation of the AVP promoter. SK-N-SH cells transfected with ER alpha exhibited increased luciferase activity in response to estrogen, and the selective estrogen receptor modulators (SERMs), Tamoxifen, and ICI 182,780. Cells transfected with ER beta exhibited a high constitutive activity, which is unchanged by exposure to SERMs but can be inhibited by estrogen. Deletion of 1.5 kb from the 5' end or mutation of a single estrogen response element (ERE)-like sequence resulted in loss of estrogen-dependent induction by ER alpha and increased the ability of estrogen to inhibit the high constitutive activity of ER beta. The distal ERE-containing 1.5-kb fragment, when coupled to luciferase, is able to support both ER alpha and ER beta mediated activation of transcription by estrogen. These results suggest that a single ERE in the distal 1.5-kb portion of the 5.5-kb fragment contains the primary positive estrogen responsive sequences for ER alpha and ER beta. The data also suggest that sequences proximal to this element serve to inhibit transcription mediated by ER beta.


Assuntos
Arginina Vasopressina/genética , Estradiol/análogos & derivados , Regulação da Expressão Gênica , Receptores de Estrogênio/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Fulvestranto , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Luciferases/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ratos , Receptores de Estrogênio/genética , Elementos de Resposta , Tamoxifeno/farmacologia , Transcrição Gênica , Transfecção
13.
Brain Res Mol Brain Res ; 78(1-2): 138-45, 2000 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-10891593

RESUMO

Mutations in the presenilin genes PS1 and PS2 cause familial Alzheimer's disease (AD). In a previous study, we reported that PS2 mRNA levels are decreased in the hippocampus, frontal cortex and basal forebrain of subjects with late-onset sporadic AD. In this study, we examined whether this downregulation occurs as the disease progresses from mild to severe stages or whether downregulation of PS2 expression is an early event in AD. We used in situ hybridization histochemistry to quantify the level of expression of PS2 message in the hippocampus of normal subjects and subjects with mild, moderate or severe AD. Several regions of the hippocampus which are sequentially susceptible to AD neuropathology as the disease progresses in severity were analyzed. We demonstrate that specific downregulation of PS2 expression is as severe in subjects with mild AD as it is in subjects in late stages of the disease. In addition, we show that hippocampal regions that are relatively free of AD neuropathology during early stages of the disease exhibit severely compromised PS2 mRNA levels even in mild AD cases. In contrast, PS2 is expressed at normal levels in the cerebellum, a region which succumbs to significantly fewer AD-related insults even at very advanced stages of the disease. These results suggest that the specific downregulation of PS2 gene expression is an early event in sporadic late-onset AD.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Proteínas de Membrana/genética , Idade de Início , Idoso , Idoso de 80 Anos ou mais , Química Encefálica/genética , Cerebelo/fisiopatologia , Progressão da Doença , Feminino , Expressão Gênica/fisiologia , Hipocampo/fisiopatologia , Humanos , Hibridização In Situ , Masculino , Pessoa de Meia-Idade , Presenilina-2 , RNA Mensageiro/análise , Índice de Gravidade de Doença
14.
J Pharmacol Exp Ther ; 291(2): 531-7, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10525068

RESUMO

Acute blockade of dopamine D(2) receptors by the typical antipsychotic drug haloperidol leads to alterations in neuronal gene expression and behavior. In the dorsolateral striatum, the levels of mRNA for the immediate-early gene c-fos and the neuropeptide gene neurotensin/neuromedin N (NT/N) are significantly increased by haloperidol. An acute behavioral response to haloperidol is catalepsy, considered to be a rodent correlate of some of the immediate extrapyramidal motor side effects seen in humans. Several lines of evidence suggest a link between neurotensin induction in the dorsolateral striatum and catalepsy. We hypothesize that both striatal gene induction and catalepsy elicited by haloperidol arise from the combined effect of excitatory adenosinergic and glutamatergic inputs acting at adenosine A(2A) and N-methyl-D-aspartate (NMDA) receptors, respectively. In agreement with our previous reports, adenosine antagonists reduced haloperidol-induced c-fos and neurotensin gene expression as well as catalepsy. In agreement with other reports, the noncompetitive NMDA receptor antagonist MK-801 also reduced gene expression and catalepsy in response to haloperidol. The competitive NMDA receptor antagonist LY235959 decreased haloperidol-induced catalepsy. We show here that blocking both A(2A) and NMDA receptors simultaneously in conjunction with haloperidol resulted in a combined effect on gene expression and behavior that was greater than that for block of either receptor alone. Both c-fos and NT/N mRNA levels were reduced, and catalepsy was completely abolished. These results indicate that the haloperidol-induced increases in c-fos and NT gene expression in the dorsolateral striatum and catalepsy are driven largely by adenosine and glutamatergic inputs acting at A(2A) and NMDA receptors.


Assuntos
Catalepsia/genética , Antagonistas de Dopamina/farmacologia , Genes fos/efeitos dos fármacos , Haloperidol/farmacologia , Receptores de N-Metil-D-Aspartato/fisiologia , Receptores Purinérgicos P1/fisiologia , Animais , Autorradiografia , Catalepsia/prevenção & controle , Maleato de Dizocilpina/farmacologia , Interações Medicamentosas , Hibridização In Situ , Masculino , Fármacos Neuroprotetores/farmacologia , Neurotensina/genética , Ratos , Ratos Sprague-Dawley , Receptores da Bombesina/efeitos dos fármacos , Receptores de Neurotensina/efeitos dos fármacos
15.
Neuroscience ; 89(3): 927-38, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10199625

RESUMO

Typical antipsychotic agents are potent antagonists of Gi-coupled dopamine D2 receptors, but their mechanisms of action following this initial blockade remain poorly understood. We hypothesized that in striatal neurons, interruption of this inhibitory dopamine D2 input would unmask endogenous striatal Gs-coupled receptors. An increase in cAMP levels generated by these unopposed receptors would then lead to the well-described behavioral and molecular effects of antipsychotic administration such as catalepsy and striatal c-fos and neurotensin gene transcription. We examined three striatal Gs-coupled receptor systems (serotonin 5-HT4, serotonin 5-HT6 and adenosine A2a) to assess their potential involvement in the mechanism of action of the typical antipsychotic haloperidol. Antagonists of each of these three receptor systems together with a 1 mg/kg dose of haloperidol were co-administered to Sprague-Dawley rats, and both the degree of catalepsy produced in the animals and the induction of striatal c-fos and neurotensin messenger RNAs were measured. Both the specific adenosine A2a antagonist 8-(3-chlorostyryl)-caffeine and the general adenosine antagonist theophylline reduced haloperidol-dependent induction of striatal neurotensin and c-fos messenger RNA. Administration of these agents also greatly reduced the degree of catalepsy induced by haloperidol. Antagonists of the 5-HT6 receptor failed to block the induction of striatal messenger RNAs, but the 5-HT6 antagonist clozapine (an important atypical antipsychotic agent in its own right) was a potent inhibitor of catalepsy. 5-HT4 agents were unable to alter haloperidol's effects on striatal messenger RNA levels or catalepsy. We conclude that the striatal Gs-coupled adenosine A2a receptor is an important mediator of the molecular and behavioral sequelae following haloperidol administration.


Assuntos
Adenosina/fisiologia , Antipsicóticos/farmacologia , Comportamento Animal/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Haloperidol/farmacologia , Proteínas do Tecido Nervoso/efeitos dos fármacos , Receptores Purinérgicos P1/fisiologia , Receptores de Serotonina/fisiologia , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Serotonina/fisiologia , Animais , Cafeína/análogos & derivados , Cafeína/farmacologia , Catalepsia/induzido quimicamente , Clozapina/farmacologia , Corpo Estriado/citologia , AMP Cíclico/fisiologia , Antagonistas dos Receptores de Dopamina D2 , Subunidades alfa Gs de Proteínas de Ligação ao GTP/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Genes fos/efeitos dos fármacos , Proteínas do Tecido Nervoso/fisiologia , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor A2A de Adenosina , Receptores de Dopamina D2/fisiologia , Receptores Purinérgicos P1/efeitos dos fármacos , Receptores de Serotonina/efeitos dos fármacos , Receptores 5-HT4 de Serotonina , Antagonistas da Serotonina/farmacologia , Teofilina/farmacologia
16.
J Neurosci ; 19(7): 2455-63, 1999 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10087060

RESUMO

Pharmacological and biochemical approaches were used to elucidate the involvement of growth factor signaling pathways mediating estrogen neuroprotection in primary cortical neurons after glutamate excitotoxicity. We addressed the activation of mitogen-activated protein kinase (MAPK) signaling pathways, which are activated by growth factors such as nerve growth factor (NGF). Inhibition of MAPK signaling with the MAPK kinase inhibitor PD98059 blocks both NGF and estrogen neuroprotection in these neurons. These results correlate with a rapid and sustained increase in MAPK activity within 30 min of estrogen exposure. The involvement of signaling molecules upstream from MAPK was also examined to determine whether activation of MAPK by estrogen is mediated by tyrosine kinase activity. Estrogen produces a rapid, transient activation of src-family tyrosine kinases and tyrosine phosphorylation of p21(ras)-guanine nucleotide activating protein. Effects of estrogen on neuroprotection, as well as rapid activation of tyrosine kinase and MAPK activity, are blocked by the anti-estrogen ICI 182,780. This provides evidence that activation of the MAPK pathway by estrogen participates in mediating neuroprotection via an estrogen receptor. These results describe a novel mechanism by which cytoplasmic actions of the estrogen receptor may activate the MAPK pathway, thus broadening the understanding of effects of estrogen in neurons.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Estrogênios/farmacologia , Ácido Glutâmico/toxicidade , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais/fisiologia , 2-Amino-5-fosfonovalerato/farmacologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Maleato de Dizocilpina/farmacologia , Ativação Enzimática , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteínas Tirosina Quinases/metabolismo , Ratos
17.
Neuroreport ; 9(11): 2565-8, 1998 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-9721933

RESUMO

Neuroprotective effects of estrogen have been demonstrated against a variety of cytotoxic insults. We present data here addressing a possible mechanism of estrogen neuroprotection in the human teratocarcinoma cell line NT2 terminally differentiated to a neuronal phenotype. Cell death induced by H2O2 or glutamate results in a dose-dependent cell death of NT2 neurons, while 24 h of estrogen pretreatment significantly enhances neuronal viability. Bcl-2 expression has been shown to reduce oxidative stress and prevent cell death. In NT2 neurons, Bcl-2 levels are dramatically elevated upon differentiation and are further enhanced with estrogen treatment. These results suggest that neuroprotective effects of estrogen may be related to increases in Bcl-2 expression.


Assuntos
Estrogênios/fisiologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Genes bcl-2/genética , Neurônios/fisiologia , Western Blotting , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Diferenciação Celular/fisiologia , Aminoácidos Excitatórios/toxicidade , Ácido Glutâmico/toxicidade , Humanos , Peróxido de Hidrogênio/toxicidade , Oxidantes/toxicidade , Estresse Oxidativo , Células Tumorais Cultivadas
18.
J Neurosci ; 18(17): 6672-80, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9712639

RESUMO

Steroid hormones exert dramatic effects on neuronal expression of genes that encode neuropeptides. Expression of the neurotensin/neuromedin (NT/N) gene in preoptic area neurons is dramatically enhanced by estrogen in vivo, even though its promoter lacks palindromic estrogen response elements. We report here that estrogen promotes transcription of this gene by interactions with the cAMP cascade in a neuronal cell line, SK-N-SH, and in a mouse model. In neuroblastoma cells, estrogen increases cAMP and the phosphorylation of the cAMP response element-binding protein in a time frame that precedes induction of NT/N gene transcription. Interference with the cAMP/protein kinase A signal transduction cascade blocks the ability of estrogen to elicit increases in transcription of this gene. Furthermore, in studies performed in vivo using mice deficient in protein kinase A, estrogen fails to induce increases in NT/N mRNA but retains its ability to promote estrogen response element-dependent progesterone receptor gene transcription. These data represent the first report of a nonclassical effect of estrogen on the expression of an endogenous estrogen-regulated neuropeptide gene through cAMP-mediated mechanisms both in a neuroblastoma cell line and in hypothalamic neurons. More importantly, this "cross-talk" may represent a more generalized mechanism by which steroid hormones act through other signal transduction cascades to regulate the expression of other genes in the brain.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , AMP Cíclico/fisiologia , Estrogênios/farmacologia , Neurotensina/genética , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Animais , Células Cultivadas , Antagonistas de Estrogênios/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo
19.
J Neurosci ; 18(10): 3639-49, 1998 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-9570795

RESUMO

Motor behavior is modulated by dopamine-responsive neurons in the striatum, where dopaminergic signaling uses G-protein-coupled pathways, including those that result in the activation of cAMP-dependent protein kinase (PKA). The RIIbeta isoform of PKA is highly enriched in the striatum, and targeted disruption of the RIIbeta gene in mice leads to a dramatic reduction in total PKA activity in this region. Although the mutant mice show typical locomotor responses after acute administration of dopaminergic drugs, they display abnormalities in two experience-dependent locomotor behaviors: training on the rotarod task and locomotor sensitization to amphetamine. In addition, amphetamine induction of fos is absent, and the basal expression of dynorphin mRNA is reduced in the striatum. These results demonstrate that motor learning and the regulation of neuronal gene expression require RIIbeta PKA, whereas the acute locomotor effects of dopaminergic drugs are relatively unaffected by this PKA deficiency.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/genética , Atividade Motora/fisiologia , Anfetamina/farmacologia , Animais , Comportamento Animal/fisiologia , Corpo Estriado/citologia , Corpo Estriado/efeitos dos fármacos , Subunidade RIIbeta da Proteína Quinase Dependente de AMP Cíclico , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dopamina/fisiologia , Dopaminérgicos/farmacologia , Relação Dose-Resposta a Droga , Dinorfinas/genética , Fertilidade/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Hibridização In Situ , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Longevidade/genética , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Neurônios/enzimologia , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/metabolismo
20.
Brain Res ; 789(2): 343-6, 1998 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-9573399

RESUMO

The effects of one week of estrogen replacement on choline acetyltransferase (ChAT) and trkA mRNA expression are examined in young and aged rodents to determine whether estrogen continues to affect cholinergic neurons in aging brain. Significant increases in ChAT and trkA are observed in the nucleus basalis of Meynert (nBM) of both age groups. ChAT expression is also increased in the HDB without changes in trkA expression. Results indicate modulation of ChAT expression by estrogen is retained in the aged rodent brain and suggests the possibility that changes in ChAT expression may be dissociated from concurrent alterations in trkA.


Assuntos
Envelhecimento/metabolismo , Proteínas de Transporte/genética , Colina O-Acetiltransferase/genética , Estradiol/farmacologia , Proteínas de Membrana/genética , Prosencéfalo/metabolismo , RNA Mensageiro/metabolismo , Receptor trkA , Animais , Feminino , Hibridização In Situ , Ovariectomia , Ratos , Ratos Endogâmicos F344
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...