Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Acta Biomater ; 10(3): 1443-50, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24287162

RESUMO

High-purity (SupT) and reagent-grade (ST), stoichiometric and silicate-containing α-tricalcium phosphate (α-TCP: ST0/SupT0 and Si-TCP x=0.10: ST10/SupT10) were prepared by solid-state reaction based on the substitution mechanism Ca3(PO4)(2-x)(SiO4)x. Samples were determined to be phase pure by X-ray diffraction (XRD), and Rietveld analysis performed on the XRD data confirmed inclusion of Si in the α-TCP structure as determined by increases in unit cell parameters; particularly marked increases in the b-axis and ß-angle were observed. X-ray fluorescence (XRF) confirmed the presence of expected levels of Si in Si-TCP compositions as well as significant levels of impurities (Mg, Al and Fe) present in all ST samples; SupT samples showed both expected levels of Si and a high degree of purity. Phosphorus ((31)P) magic-angle-spinning solid-state nuclear magnetic resonance (MAS NMR) measurements revealed that the high-purity reagents used in the synthesis of SupT0 can resolve the 12 expected peaks in the (31)P spectrum of α-TCP compared to the low-purity ST0 that showed significant spectral line broadening; line broadening was also observed with the inclusion of Si which is indicative of induced structural disorder. Silicon ((29)Si) MAS NMR was also performed on both Si-TCP samples which revealed Q(0) species of Si with additional Si Q(1)/Q(2) species that may indicate a potential charge-balancing mechanism involving the inclusion of disilicate groups; additional Q(4) Si species were also observed, but only for ST10. Heating and cooling rates were briefly investigated by (31)P MAS NMR which showed no significant line broadening other than that associated with the emergence of ß-TCP which was only realised with the reagent-grade sample ST0. This study provides an insight into the structural effects of Si-substitution in α-TCP and could provide a basis for understanding how substitution affects the physicochemical properties of the material.


Assuntos
Fosfatos de Cálcio/química , Espectroscopia de Ressonância Magnética , Silicatos/química , Espectrometria por Raios X , Difração de Raios X , Silício/química
2.
Eur Respir J ; 32(2): 334-43, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18385167

RESUMO

Small-scale clinical trials show that treatment of cystic fibrosis (CF) patients with ibuprofen, a nonsteroidal anti-inflammatory drug, improves the symptoms of CF and slows down the decline of lung function. Paradoxically, ibuprofen inhibits ligand-stimulated CF transmembrance conductance regulator (CFTR) activity. The aim of the present study was to investigate the effects of ibuprofen on CFTR function under different conditions. Patch-clamp recordings were performed in two lines of human airway epithelial cells: IB3-8-3-7 cells, which express wild-type CFTR; and IB3-1 cells, which express the variant CFTR with deletion of phenylalanine 580 (DeltaF580CFTR). Addition of ibuprofen to the extracellular solution caused a rapid inhibition of CFTR activity in IB3-8-3-7 cells in the presence of a high intracellular concentration of cAMP, whereas ibuprofen enhanced the CFTR conductance at low levels of cAMP. Introducing ibuprofen into the interior of cells occluded the enhancing effect of ibuprofen. Notably, the variant CFTR-mediated conductance was detected in IB3-1 cells treated with myoinositol and was enhanced by ibuprofen at endogenous levels of cAMP. In summary, nonsteroidal anti-inflammatory drugs increase the function of both wild-type cystic fibrosis transmembrane conductance regulator and the phenylalanine 580 deletion in cultured human airway epithelial cells at endogenous levels of cAMP.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/metabolismo , Epitélio/microbiologia , Pulmão/microbiologia , Mutação , Regulação para Cima , Linhagem Celular , AMP Cíclico/metabolismo , Epitélio/metabolismo , Humanos , Ibuprofeno/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Modelos Biológicos , Técnicas de Patch-Clamp , Receptores de GABA/metabolismo
3.
J Neurosci ; 25(49): 11374-84, 2005 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-16339032

RESUMO

At CA1 synapses, activation of NMDA receptors (NMDARs) is required for the induction of both long-term potentiation and depression. The basal level of activity of these receptors is controlled by converging cell signals from G-protein-coupled receptors and receptor tyrosine kinases. Pituitary adenylate cyclase activating peptide (PACAP) is implicated in the regulation of synaptic plasticity because it enhances NMDAR responses by stimulating Galphas-coupled receptors and protein kinase A (Yaka et al., 2003). However, the major hippocampal PACAP1 receptor (PAC1R) also signals via Galphaq subunits and protein kinase C (PKC). In CA1 neurons, we showed that PACAP38 (1 nM) enhanced synaptic NMDA, and evoked NMDAR, currents in isolated CA1 neurons via activation of the PAC1R, Galphaq, and PKC. The signaling was blocked by intracellular applications of the Src inhibitory peptide Src(40-58). Immunoblots confirmed that PACAP38 biochemically activates Src. A Galphaq pathway is responsible for this Src-dependent PACAP enhancement because it was attenuated in mice lacking expression of phospholipase C beta1, it was blocked by preventing elevations in intracellular Ca2+, and it was eliminated by inhibiting either PKC or cell adhesion kinase beta [CAKbeta or Pyk2 (proline rich tyrosine kinase 2)]. Peptides that mimic the binding sites for either Fyn or Src on receptor for activated C kinase-1 (RACK1) also enhanced NMDAR in CA1 neurons, but their effects were blocked by Src(40-58), implying that Src is the ultimate regulator of NMDARs. RACK1 serves as a hub for PKC, Fyn, and Src and facilitates the regulation of basal NMDAR activity in CA1 hippocampal neurons.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Hipocampo/fisiologia , Neurônios/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Proteína Quinase C/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Quinases da Família src/metabolismo , Sequência de Aminoácidos , Animais , Ativação Enzimática/fisiologia , Hipocampo/enzimologia , Hipocampo/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Neurônios/enzimologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Ratos , Ratos Wistar
4.
Reprod Fertil Dev ; 17(7): 743-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16364229

RESUMO

The hypothalamic-pituitary-adrenal (HPA) axis is susceptible to programming during fetal life. Such programming occurs, at least partially, at the level of the hippocampus. The hippocampus plays a central role in regulation of the HPA axis and release of endogenous glucocorticoids, via mediation of glucocorticoid negative feedback. Fetal exposure to synthetic glucocorticoids can permanently alter glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) levels within the hippocampus, and serotonin is thought to be involved in this process. In the present study, we hypothesised that dexamethasone, cortisol and serotonin exposure would modify GR mRNA expression within fetal guinea-pig hippocampal cultures. Cultures were derived from 40-day-old guinea-pig fetuses, and were exposed to 0, 1, 10 and 100 nM dexamethasone, cortisol or serotonin for 4 days. Expression of GR and MR mRNA was examined by in situ hybridisation followed by high-resolution silver emulsion autoradiography. Four-day exposure to dexamethasone (P < 0.05; 100 nM) or cortisol (P = 0.08; 100 nM) downregulated the expression of GR mRNA within neurons. There was no change in the expression of MR mRNA levels following cortisol treatment. Exposure to serotonin (100 nM) significantly increased GR mRNA levels in hippocampal neurons. We conclude that synthetic and endogenous glucocorticoids, as well as serotonin, can influence GR expression during hippocampal development and in this way may act to permanently programme HPA function.


Assuntos
Retroalimentação Fisiológica/efeitos dos fármacos , Feto/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Receptores de Glucocorticoides/metabolismo , Animais , Autorradiografia , Dexametasona/farmacologia , Cobaias , Hipocampo/citologia , Hidrocortisona/farmacologia , Hibridização In Situ , Técnicas In Vitro , RNA Mensageiro/genética , Receptores de Glucocorticoides/genética , Serotonina/farmacologia
5.
J Neurophysiol ; 86(5): 2520-6, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11698539

RESUMO

Concentrations of extracellular calcium ([Ca(2+)](e)) in the CNS decrease substantially during seizure activity. We have demonstrated previously that decreases in [Ca(2+)](e) activate a novel calcium-sensing nonselective cation (csNSC) channel in hippocampal neurons. Activation of csNSC channels is responsible for a sustained membrane depolarization and increased neuronal excitability. Our study has suggested that the csNSC channel is likely involved in generating and maintaining seizure activities. In the present study, the effects of anti-epileptic agent lamotrigine (LTG) on csNSC channels were studied in cultured mouse hippocampal neurons using patch-clamp techniques. At a holding potential of -60 mV, a slow inward current through csNSC channels was activated by a step reduction of [Ca(2+)](e) from 1.5 to 0.2 mM. LTG decreased the amplitude of csNSC currents dose dependently with an IC(50) of 171 +/- 25.8 (SE) microM. The effect of LTG was independent of membrane potential. In the presence of 300 microM LTG, the amplitude of csNSC current was decreased by 31 +/- 3% at -60 mV and 29 +/- 2.9% at +40 mV (P > 0.05). LTG depressed csNSC current without affecting the potency of Ca(2+) block of the current (IC(50) for Ca(2+) block of csNSC currents in the absence of LTG: 145 +/- 18 microM; in the presence of 300 microM LTG: 136 +/- 10 microM. n = 5, P > 0.05). In current-clamp recordings, activation of csNSC channel by reducing the [Ca(2+)](e) caused a sustained membrane depolarization and an increase in the frequency of spontaneous firing of action potentials. LTG (300 microM) significantly inhibited csNSC channel-mediated membrane depolarization and the excitation of neurons. Fura-2 ratiometric Ca(2+) imaging experiment showed that LTG also inhibited the increase in intracellular Ca(2+) concentration induced by csNSC channel activation. The effect of LTG on csNSC channels may partially contribute to its broad spectrum of anti-epileptic actions.


Assuntos
Anticonvulsivantes/farmacologia , Cálcio/metabolismo , Cátions/metabolismo , Hipocampo/metabolismo , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/fisiologia , Neurônios/metabolismo , Triazinas/farmacologia , Animais , Cálcio/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Condutividade Elétrica , Eletrofisiologia , Hipocampo/citologia , Membranas Intracelulares/metabolismo , Canais Iônicos/antagonistas & inibidores , Lamotrigina , Camundongos , Concentração Osmolar
6.
J Clin Endocrinol Metab ; 86(11): 5585-92, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11701739

RESUMO

In human pregnancy, cortisol and PGs are involved in the onset of labor and play an important role in the mechanisms leading to parturition. Recent studies have shown that at term, cortisol increases PG synthesis and decreases PG metabolism in chorion trophoblast (CT) cells. In CT, 11 beta-hydroxysteroid oxidase type 1 (11 beta-HSD1) converts biologically inactive cortisone to cortisol to regulate cortisol availability. In the present study, we have investigated whether 11 beta-HSD1 activity could be influenced by PGs. We have shown that in CT, PGF2alpha rapidly increased 11 beta-HSD1 reductase activity in a dose-dependent manner via the PGF2alpha receptor, localized in the fetal membranes. PGF2alpha stimulated 11 beta-HSD1 activity through increased intracellular calcium mobilization, activation of PKC, and the phosphorylation of the 11 beta-HSD enzyme. We propose that within CT there is a novel feed forward loop by which PGF2alpha acts to promote cortisol production from cortisone through increases in 11beta-HSD1, and this in turn leads to further net PG output for the onset of labor and birth.


Assuntos
Dinoprosta/farmacologia , Hidrocortisona/biossíntese , Hidroxiesteroide Desidrogenases/biossíntese , Trabalho de Parto/fisiologia , 11-beta-Hidroxiesteroide Desidrogenases , Adulto , Cálcio/metabolismo , Células Cultivadas , Retroalimentação/fisiologia , Feminino , Feto/metabolismo , Corantes Fluorescentes , Fura-2 , Humanos , Imuno-Histoquímica , Luteolíticos/farmacologia , Membranas/metabolismo , Testes de Precipitina , Gravidez , Prostaglandinas F Sintéticas/farmacologia , Receptores de Prostaglandina/metabolismo , Estimulação Química , Trofoblastos/metabolismo
7.
J Neurosci ; 21(21): 8464-72, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11606635

RESUMO

The postsynaptic density (PSD) at excitatory dendritic synapses comprises a protein complex of glutamate receptors, scaffolding elements, and signaling enzymes. For example, NMDA receptors (NMDARs) are linked to several proteins in the PSD, such as PSD-95, and are also tethered via binding proteins such as alpha-actinin directly to filamentous actin of the cytoskeleton. Depolymerization of the cytoskeleton modulates the activity of NMDARs, and, in turn, strong activation of NMDARs can trigger depolymerization of actin. Myosin, the motor protein of muscular contraction and nonmuscle motility, is also associated with NMDARs and the PSD. We show here that constitutively active myosin light chain kinase (MLCK) enhances NMDAR-mediated whole-cell and synaptic currents in acutely isolated CA1 pyramidal and cultured hippocampal neurons, whereas inhibitors of MLCK depress these currents. This MLCK-dependent regulation was observed in cell-attached patches but was lost after excision to inside-out patches. Furthermore, the enhancement induced by constitutively active MLCK and the depression of MLCK inhibitors were eliminated after depolymerization of the cytoskeleton. NMDARs and MLCK did not colocalize in clusters on the dendrites of cultured hippocampal neurons, further indicating that the effects of MLCK are mediated indirectly via actomyosin. Our results suggest that MLCK enhances actomyosin contractility to either increase the membrane tension on NMDARs or to alter physical relationships between the actin cytoskeleton and the linker proteins of NMDARs.


Assuntos
Actinas/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Cálcio/metabolismo , Carbocianinas , Separação Celular , Células Cultivadas , Dendritos/metabolismo , Inibidores Enzimáticos/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Corantes Fluorescentes , Hipocampo , Camundongos , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Quinase de Cadeia Leve de Miosina/farmacologia , N-Metilaspartato/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos
8.
Curr Drug Targets ; 2(3): 299-312, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11554554

RESUMO

Synaptic plasticity, or long-term potentiation (LTP), of excitatory synapses in the hippocampus contributes to learning and the establishment of spatial memories. In the CA1 region, induction of LTP enhances the function of postsynaptic alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptors (AMPARs) because of the Ca2+-calmodulin kinase II (CaMKII)-dependent phosphorylation of this subtype of glutamate receptor. Entry of Ca2+, via N-methyl-D-aspartate receptors (NMDARs), during strong synaptic stimulation provides the stimulus to trigger phosphorylation of AMPARs. However, this induction also requires activation of a protein kinase C (PKC)-dependent tyrosine kinase signal cascade and a concomitant upregulation of NMDARs. This review focuses upon NMDARs as potential targets of PKC and/or of the PKC-dependent tyrosine kinase cascade. PKC, acting via the CAKbeta/Src tyrosine kinase cascade, enhances NMDAR activation and may increase the number of receptors expressed in synapses. In contrast, direct phosphorylation of NMDARs by PKC increases the sensitivity of NMDA channel inactivation to intracellular Ca2+. In CAI neurons, PKC provides a point of convergence of control of NMDARs and synaptic plasticity for a wide variety of G-protein coupled and growth factor receptors.


Assuntos
Proteína Quinase C/fisiologia , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/fisiologia , Transdução de Sinais/efeitos dos fármacos , Animais , Aminoácidos Excitatórios/fisiologia , Humanos , Plasticidade Neuronal , Receptores Muscarínicos/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Ativação Transcricional/fisiologia
9.
J Neurophysiol ; 86(1): 173-82, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11431499

RESUMO

The actions of the trivalent cation Gd(3+) on whole cell AMPA receptor-mediated currents were studied in isolated hippocampal neurons, in nucleated or outside-out patches taken from cultured hippocampal neurons, and on miniature excitatory postsynaptic currents (mEPSCs) recorded in cultured hippocampal neurons. Glutamate, AMPA, or kainate was employed to activate AMPA receptors. Applications of relatively low concentrations of Gd(3+) (0.1-10 microM) substantially enhanced steady-state whole cell glutamate and kainate-evoked currents without altering peak currents, suggesting that desensitization was reduced. However, higher concentrations (>30 microM) depressed steady-state currents, indicating an underlying inhibition of channel activity. Lower concentrations of Gd(3+) also increased the potency of peak glutamate-evoked currents without altering that of steady-state currents. An ultrafast perfusion system and nucleated patches were then used to better resolve peak glutamate-evoked currents. Low concentrations of Gd(3+) reduced peak currents, enhanced steady-state currents, and slowed the onset of desensitization, providing further evidence that this cation reduces desensitization. In the presence of cyclothiazide, a compound that blocks desensitization, a low concentration Gd(3+) inhibited both peak and steady-state currents, indicating that Gd(3+) both reduces desensitization and inhibits these currents. Gd(3+) reduced the probability of channel opening at the peak of the currents but did not alter the single channel conductance calculated using nonstationary variance analysis. Recovery from desensitization was enhanced, and glutamate-evoked current activation and deactivation were slowed by Gd(3+). The Gd(3+)-induced reduction in desensitization did not require the presence of the GluR2 subunit as this effect was seen in hippocampal neurons from GluR2 null-mutant mice. Gd(3+) reduced the time course of decay of mEPSCs perhaps as a consequence of its slowing of AMPA receptor deactivation although an increase in the frequency of mEPSCs also suggested enhanced presynaptic release of transmitter. These results demonstrate that Gd(3+) potently reduces AMPA receptor desensitization and mimics a number of the properties of the positive modulators of AMPA receptor desensitization such as cyclothiazide.


Assuntos
Gadolínio/farmacologia , Hipocampo/citologia , Ativação do Canal Iônico/efeitos dos fármacos , Neurônios/fisiologia , Receptores de AMPA/metabolismo , Animais , Sinergismo Farmacológico , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Ácido Glutâmico/farmacologia , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Mutantes , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Receptores de AMPA/genética
10.
Can J Physiol Pharmacol ; 79(5): 422-9, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11405246

RESUMO

Positive modulators of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) channels reduce desensitization and alter their gating kinetics. We have discovered a novel compound nitric oxide-mimetic that similarly modulates the AMPA receptor by reducing desensitization. This, designated GT-005, belongs to the organic nitrate family that includes the nitrovasodilator nitroglycerine. In acutely isolated hippocampal neurons, GT-005 enhanced kainate (100 microM)-evoked currents with an EC50 of 1.7+/-0.2 mM and a 176+/-10% maximal increase in the steady-state current response. Similar results were found in cultured hippocampal neurons (EC50 of 1.3+/-0.2 mM and a maximal 83+/-14% increase in the steady-state current response). GT-005 reduced the desensitization of glutamate-evoked currents and slowed the onset of desensitization. This compound also increased the rate of recovery from the desensitized state. With respect to alteration of the excitatory synaptic transmission, GT-005 delayed the decay and increased the frequency of spontaneous miniature excitatory postsynaptic currents (mepsc) recorded in cultured hippocampal neurons.


Assuntos
Neurônios/efeitos dos fármacos , Nitratos/farmacologia , Receptores de AMPA/metabolismo , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/farmacologia , Hipocampo/citologia , Ácido Caínico/farmacologia , Potenciais da Membrana , Camundongos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Ratos
11.
J Neurophysiol ; 85(5): 2030-8, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11353019

RESUMO

Whole-cell or outside-out patch recordings were used to investigate the effects of protons and positive modulators of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors on the desensitization of glutamate-evoked AMPA receptor currents in isolated hippocampal CA1 neurons. Protons inhibited glutamate-evoked currents (IC(50) of 6.2 pH units) but also enhanced the apparent rate and extent of AMPA receptor desensitization. The proton-induced enhancement of desensitization could not be attributed to a reduction in the rate of recovery from desensitization or to a change in the kinetics of deactivation. Non-stationary variance analysis indicated that protons reduced maximum open probability without changing the conductance of AMPA channels. The positive modulators of AMPA receptor desensitization, cyclothiazide and GT-21-005 (an organic nitrate), reduced the proton sensitivity of AMPA receptor desensitization, which suggests that they interact with protons to diminish desensitization. In contrast, the effects of wheat germ agglutinin and aniracetam on AMPA receptor desensitization were independent of pH. These results demonstrate that a reduction in the proton sensitivity of receptor desensitization contributes to the mechanism of action of some positive modulators of AMPA receptors.


Assuntos
Benzotiadiazinas/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hipocampo/efeitos dos fármacos , Proteínas do Tecido Nervoso/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Nitratos/farmacologia , Prótons , Pirrolidinonas/farmacologia , Receptores de AMPA/efeitos dos fármacos , Aglutininas do Germe de Trigo/farmacologia , Regulação Alostérica/efeitos dos fármacos , Animais , Ácido Glutâmico/farmacologia , Hipocampo/citologia , Concentração de Íons de Hidrogênio , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Receptores de AMPA/fisiologia
12.
Anesthesiology ; 94(3): 478-88, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11374610

RESUMO

BACKGROUND: The alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) subtype of glutamate receptor mediates fast excitatory neurotransmission in the central nervous system. Many general anesthetics inhibit AMPA receptors in vitro; however, it is not certain if this inhibition contributes to the behavioral properties of these drugs. AMPA receptors lacking the GluR2 subunit are resistant to blockade by barbiturates in vitro. Paradoxically, GluR2 null mutant (-/-) mice are more sensitive to barbiturate-induced loss of the righting reflex (LORR) compared with wild-type (+/+) littermates. To determine if interactions between anesthetics and AMPA receptors account for the increased sensitivity of (-/-) mice, the effects of volatile anesthetics that do not directly inhibit AMPA receptors were examined. METHODS: Isoflurane, halothane, desflurane, or sevoflurane were administered to (-/-) and (+/+) littermate controls. Anesthetic requirements for LORR, movement to tail clamp (minimum alveolar concentration [MAC]), and hind-paw withdrawal latency (HPWL) were determined. Electrophysiologic methods examined the inhibition of AMPA receptors by isoflurane and halothane. RESULTS: Anesthetic requirements for LORR and HPWL were decreased, whereas MAC values were unchanged in (-/-) mice. Isoflurane and halothane caused minimal inhibition of AMPA receptors at clinically relevant concentrations. CONCLUSIONS: Direct blockade of AMPA receptors did not account for the increased sensitivity to volatile anesthetics in GluR2 null mutant mice for HPWL or LORR. Thus, the deficiency of GluR2-containing AMPA receptors increases the sensitivity of neuronal circuitry mediating these end points, but not MAC. GluR2-containing receptors do not contribute appreciably to MAC in this mouse model. These results illustrate the difficulties in attributing behavioral responses to drug-receptor interactions in genetically engineered animals.


Assuntos
Analgesia , Anestésicos Inalatórios/farmacologia , Comportamento Animal/efeitos dos fármacos , Receptores de AMPA/antagonistas & inibidores , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Eletrofisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Camundongos , Camundongos Mutantes
13.
Brain Res ; 896(1-2): 130-6, 2001 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-11277981

RESUMO

Studies utilizing rats and guinea pigs have demonstrated that the hypothalamo-pituitary-adrenal (HPA) axis can be programmed by glucocorticoids during fetal life. Such programming is believed to occur, at least partially, at the level of hippocampal glucocorticoid receptors (GR) and mineralocorticoid receptors (MR). Studies have also demonstrated that serotonin up regulates GR levels within the developing hippocampus. However, the cell type in which these changes take place has not been determined. We hypothesized that dexamethasone, corticosterone and serotonin exposure modify GR and MR mRNA levels in fetal mouse hippocampal cultures, and that these effects are confined to neurons. Cultures were derived from CD1 mouse fetuses on day 18 of gestation (n=8 dams). Fetal hippocampi were dissected, then mechanically and chemically dispersed. Cultures were exposed to dexamethasone, corticosterone or serotonin (1-100 nM) for 4 days. Levels of GR and MR mRNA were examined by in situ hybridization and high-resolution silver emulsion autoradiography. Four days exposure to dexamethasone or corticosterone (10 or 100 nM) decreased levels of GR mRNA within neurons. There was no significant change in MR mRNA in either experiment. Exposure to serotonin (100 nM) significantly increased expression of GR mRNA in hippocampal neurons. MR mRNA levels were unaffected by serotonin treatment. Dexamethasone, corticosterone or serotonin exposure did not alter expression of GR mRNA within glial cells. We conclude that synthetic and endogenous glucocorticoids, as well as serotonin, can influence neuronal levels of GR mRNA during hippocampal development. However, whether these effects are permanent remains to be determined.


Assuntos
Corticosterona/farmacologia , Neurônios/fisiologia , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/genética , Serotonina/farmacologia , Animais , Células Cultivadas , Dexametasona/farmacologia , Feminino , Feto/citologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glucocorticoides/farmacologia , Hipocampo/citologia , Hibridização In Situ , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos , Neurônios/citologia , Gravidez , RNA Mensageiro/análise
14.
Mol Pharmacol ; 59(4): 814-24, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11259626

RESUMO

gamma-Aminobutyric acid (GABA), the principal inhibitory neurotransmitter, activates a persistent low amplitude tonic current in several brain regions in addition to conventional synaptic currents. Here we demonstrate that GABA(A) receptors mediating the tonic current in hippocampal neurons exhibit functional and pharmacological properties different from those of quantal synaptic currents. Patch-clamp techniques were used to characterize miniature inhibitory postsynaptic currents (mIPSCs) and the tonic GABAergic current recorded in CA1 pyramidal neurons in rat hippocampal slices and in dissociated neurons grown in culture. The competitive GABA(A) receptor antagonists, bicuculline and picrotoxin, blocked both the mIPSCs and the tonic current. In contrast, mIPSCs but not the tonic current were inhibited by gabazine (SR-95531). Coapplication experiments and computer simulations revealed that gabazine bound to the receptors responsible for the tonic current but did not prevent channel activation. However, gabazine competitively inhibited bicuculline blockade. The unitary conductance of the GABA(A) receptors underlying the tonic current (approximately 6 pS) was less than the main conductance of channels activated during quantal synaptic transmission (approximately 15--30 pS). Furthermore, compounds that potentiate GABA(A) receptor function including the benzodiazepine, midazolam, and anesthetic, propofol, prolonged the duration of mIPSCs and increased tonic current amplitude in cultured neurons to different extents. Clinically-relevant concentrations of midazolam and propofol caused a greater increase in tonic current compared with mIPSCs, as measured by total charge transfer. In summary, the receptors underlying the tonic current are functionally and pharmacologically distinct from quantally activated synaptic receptors and these receptors represent a novel target for neurodepressive drugs.


Assuntos
Hipocampo/metabolismo , Neurônios/metabolismo , Receptores de GABA-A/metabolismo , Animais , Bicuculina/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Potenciais Pós-Sinápticos Excitadores/fisiologia , Antagonistas GABAérgicos/farmacologia , Antagonistas de Receptores de GABA-A , Hipocampo/citologia , Hipnóticos e Sedativos/farmacologia , Técnicas In Vitro , Camundongos , Midazolam/farmacologia , Modelos Neurológicos , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Neurônios/citologia , Técnicas de Patch-Clamp , Picrotoxina/farmacologia , Propofol/farmacologia , Piridazinas/farmacologia , Receptores de GABA-A/classificação
15.
Neuron ; 29(2): 485-96, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11239437

RESUMO

Long-term potentiation (LTP) is an activity-dependent enhancement of synaptic efficacy, considered a model of learning and memory. The biochemical cascade producing LTP requires activation of Src, which upregulates the function of NMDA receptors (NMDARs), but how Src becomes activated is unknown. Here, we show that the focal adhesion kinase CAKbeta/Pyk2 upregulated NMDAR function by activating Src in CA1 hippocampal neurons. Induction of LTP was prevented by blocking CAKbeta/Pyk2, and administering CAKbeta/Pyk2 intracellularly mimicked and occluded LTP. Tyrosine phosphorylation of CAKbeta/Pyk2 and its association with Src was increased by stimulation that produced LTP. Finally, CAKbeta/Pyk2-stimulated enhancement of synaptic AMPA responses was prevented by blocking NMDARS, chelating intracellular Ca(2+), or blocking Src. Thus, activating CAKbeta/Pyk2 is required for inducing LTP and may depend upon downstream activation of Src to upregulate NMDA receptors.


Assuntos
Potenciação de Longa Duração/fisiologia , Proteínas Tirosina Quinases/metabolismo , Células Piramidais/fisiologia , Receptores de AMPA/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Quinases da Família src/fisiologia , Animais , Quinase 2 de Adesão Focal , Hipocampo/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia , Quinases da Família src/metabolismo
16.
Neuron ; 29(1): 243-54, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11182095

RESUMO

Long-term potentiation (LTP) of excitatory transmission in the hippocampus likely contributes to learning and memory. The mechanisms underlying LTP at these synapses are not well understood, although phosphorylation and redistribution of AMPA receptors may be responsible for this form of synaptic plasticity. We show here that miniature excitatory postsynaptic currents (mEPSCs) in cultured hippocampal neurons reliably demonstrate LTP when postsynaptic NMDA receptors are briefly stimulated with glycine. LTP of these synapses is accompanied by a rapid insertion of native AMPA receptors and by increased clustering of AMPA receptors at the surface of dendritic membranes. Both LTP and glycine-facilitated AMPA receptor insertion are blocked by intracellular tetanus toxin (TeTx), providing evidence that AMPA receptors are inserted into excitatory synapses via a SNARE-dependent exocytosis during LTP.


Assuntos
Potenciação de Longa Duração/fisiologia , Neurônios/metabolismo , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Animais , Cálcio/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Exocitose/efeitos dos fármacos , Glicina/metabolismo , Glicina/farmacologia , Hipocampo/citologia , Hipocampo/metabolismo , Técnicas In Vitro , Potenciação de Longa Duração/efeitos dos fármacos , Fusão de Membrana/efeitos dos fármacos , Camundongos , Neurônios/citologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores
17.
ScientificWorldJournal ; 1: 213-5, 2001 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-12805676

RESUMO

The "aging" of populations in the developed world is rapidly altering demographics and presents a number of challenges for science and medicine. Foremost among these challenges is the need to enhance the quality of life for this "aging" majority. Paradoxically, improved prevention and treatment of diseases will only increase the number of individuals who will lose quality of life because of cognitive deficits in learning and memory. Such cognitive deficits are particularly vexing in societies where the ability to deal with information technology has become an increasing necessity. Understanding how the human brain encodes and stores information becomes critical in designing required therapeutic strategies.


Assuntos
Potenciação de Longa Duração/fisiologia , Neurônios/fisiologia , Técnicas de Cultura de Células , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Ácido Glutâmico/metabolismo , Glicina/farmacologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
18.
Exp Cell Res ; 257(2): 281-9, 2000 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10837142

RESUMO

S100B is the major low-affinity Ca(2+)-binding protein in astrocytes. In order to study the role of S100B in the maintenance of Ca(2+) homeostasis, we generated S100B null mice by a targeted inactivation of the S100B gene. Absence of S100B expression was demonstrated by Northern and Western blotting for S100B mRNA and protein, respectively, and immunoperoxidase staining of sections of various brain regions. S100B null mice were viable, fertile, and exhibited no overt behavioral abnormalities up to 12 months of age. On the basis of light microscopy and immunohistochemical staining, there were no discernable alterations in the distribution and morphology of astrocytes or neurons in sections of adult brains of these mice. Astrocytes in cerebellar cultures derived from 6-day-old S100B null mice exhibited enhanced Ca(2+) transients in response to treatment with KCl or caffeine. On the other hand, granule neurons, in the same cultures, exhibited normal Ca(2+) transients in response to treatment with KCl, caffeine, or N-methyl-d-aspartate. These results demonstrate a specific decrease in Ca(2+)-handling capacity in astrocytes derived from S100B null mice and suggest that S100B plays a role in the maintenance of Ca(2+) homeostasis in astrocytes.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Cálcio/metabolismo , Fatores de Crescimento Neural/fisiologia , Neuroglia/metabolismo , Proteínas S100 , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Encéfalo/patologia , Cafeína/farmacologia , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/genética , Células Cultivadas , Cerebelo/citologia , Feminino , Fluorometria/métodos , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fatores de Crescimento Neural/biossíntese , Fatores de Crescimento Neural/genética , Neuroglia/efeitos dos fármacos , Cloreto de Potássio/farmacologia , Subunidade beta da Proteína Ligante de Cálcio S100
19.
J Neurosci ; 20(12): 4452-61, 2000 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10844014

RESUMO

The NMDA subtype of the glutamate-gated channel exhibits a high permeability to Ca(2+). The influx of Ca(2+) through NMDA channels is limited by a rapid and Ca(2+)/calmodulin (CaM)-dependent inactivation that results from a competitive displacement of cytoskeleton-binding proteins from the NR1 subunit of the receptor by Ca(2+)/CaM (Zhang et al., 1998; Krupp et al., 1999). The C terminal of this subunit can be phosphorylated by protein kinase C (PKC) (Tingley et al., 1993). The present study sought to investigate whether PKC regulates Ca(2+)-dependent inactivation of the NMDA channel in hippocampal neurons. Activation of endogenous PKC by 4beta-phorbol 12-myristate 13-acetate enhanced peak (I(p)) and depressed steady-state (I(ss)) NMDA-evoked currents, resulting in a reduction in the ratio of these currents (I(ss)/I(p)). We demonstrated previously that PKC activity enhances I(P) via a sequential activation of the focal adhesion kinase cell adhesion kinase beta/proline-rich tyrosine kinase 2 (CAKbeta/Pyk2) and the nonreceptor tyrosine kinase Src (Huang et al., 1999; Lu et al., 1999). Here, we report that the PKC-induced depression of I(ss) is unrelated to the PKC/CAKbeta/Src-signaling pathway but depends on the concentration of extracellular Ca(2+). Intracellular applications of CaM reduced I(ss)/I(p) and occluded the Ca(2+)-dependent effect of phorbol esters on I(ss.) Moreover, increasing the concentration of intracellular Ca(2+) buffer or intracellular application of the inhibitory CaM-binding peptide (KY9) greatly reduced the phorbol ester-induced depression of I(ss). Taken together, these results suggest that PKC enhances Ca(2+)/CaM-dependent inactivation of the NMDA channel, most likely because of a phosphorylation-dependent regulation of interactions between receptor subunits, CaM, and other postsynaptic density proteins.


Assuntos
Cálcio/fisiologia , Potenciais Evocados/fisiologia , Hipocampo/fisiologia , N-Metilaspartato/farmacologia , Proteína Quinase C/metabolismo , Células Piramidais/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Animais , Cálcio/farmacologia , Calmodulina/fisiologia , Ácido Egtázico/farmacologia , Ativação Enzimática , Potenciais Evocados/efeitos dos fármacos , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Glicina/farmacologia , Técnicas In Vitro , Cinética , Proteínas Tirosina Quinases/metabolismo , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Acetato de Tetradecanoilforbol/farmacologia
20.
Nat Neurosci ; 3(6): 559-65, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10816311

RESUMO

In central neurons, the second messenger cGMP is believed to induce long-term changes in efficacy at glutamatergic synapses through activation of protein kinase G (PKG). Stimulating nitric oxide synthase, activating soluble guanylyl cyclase or elevating concentrations of intracellular cGMP depressed excitatory synaptic transmission in CA1 hippocampal neurons. Unexpectedly, intracellular cGMP depressed responses of AMPA receptors and inhibited excitatory postsynaptic currents in hippocampal neurons independently of phosphorylation. Our findings demonstrate that cGMP's modulation of excitatory transmission may involve a coupling of AMPA channel activity to levels of cGMP.


Assuntos
GMP Cíclico/metabolismo , Inibição Neural/fisiologia , Proteínas Quinases/metabolismo , Receptores de AMPA/metabolismo , Animais , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico , Dibutiril GMP Cíclico/farmacologia , Relação Dose-Resposta a Droga , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Retroalimentação/fisiologia , Ácido Glutâmico/farmacologia , Hipocampo/citologia , Hipocampo/metabolismo , Técnicas In Vitro , N-Metilaspartato/farmacologia , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Técnicas de Patch-Clamp , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Ratos , Ratos Wistar , Receptores de AMPA/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...