Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Neurophysiol ; 120(3): 1264-1273, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29897859

RESUMO

The current electrophysiological study investigated the functional roles of high- and low-voltage-activated Ca2+ channel subtypes on glutamatergic small mossy fiber nerve terminals (SMFTs) that synapse onto rat hippocampal CA3 neurons. Experiments combining both the "synapse bouton" preparation and single-pulse focal stimulation technique were performed using the conventional whole cell patch configuration under voltage-clamp conditions. Nifedipine, at a high concentration, and BAY K 8644 inhibited and facilitated the glutamatergic excitatory postsynaptic currents (eEPSCs) that were evoked by 0.2-Hz stimulation, respectively. However, these drugs had no effects on spontaneous EPSCs (sEPSCs). Following the use of a high stimulation frequency of 3 Hz, however, nifedipine markedly inhibited eEPSCs at the low concentration of 0.3 µM. Moreover, ω-conotoxin GVIA and ω-agatoxin IVA significantly inhibited both sEPSCs and eEPSCs. Furthermore, SNX-482 slightly inhibited eEPSCs. R(-)-efonidipine had no effects on either sEPSCs or eEPSCs. It was concluded that glutamate release from SMFTs depends largely on Ca2+ entry through N- and P/Q-type Ca2+ channels and, to a lesser extent, on R-type Ca2+ channels. The contribution of L-type Ca2+ channels to eEPSCs was small at low-firing SMFTs but more significant at high-firing SMFTs. T-type Ca2+ channels did not appear to be involved in neurotransmission at SMFTs. NEW & NOTEWORTHY Action potential-evoked glutamate release from small mossy fiber nerve terminals (SMFTs) that synapse onto rat hippocampal CA3 neurons is regulated by high-threshold but not low-threshold Ca2+ channel subtypes. The functional contribution mainly depends on N- and P/Q-type Ca2+ channels and, to a lesser extent, on R-type Ca2+ channels. However, in SMFTs stimulated at a high 3-Hz frequency, L-type Ca2+ channels contributed significantly to the currents. The present results are consistent with previous findings from fluorometric studies of large mossy fiber boutons.


Assuntos
Potenciais de Ação , Região CA3 Hipocampal/fisiologia , Canais de Cálcio/fisiologia , Ácido Glutâmico/fisiologia , Fibras Musgosas Hipocampais/fisiologia , Terminações Pré-Sinápticas/fisiologia , Animais , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores , Ratos Wistar
2.
J Neurophysiol ; 114(2): 1109-18, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26019316

RESUMO

The effects of heavy water (deuterium oxide, D2O) on GABAergic and glutamatergic spontaneous and evoked synaptic transmission were investigated in acute brain slice and isolated "synaptic bouton" preparations of rat hippocampal CA3 neurons. The substitution of D2O for H2O reduced the frequency and amplitude of GABAergic spontaneous inhibitory postsynaptic currents (sIPSCs) in a concentration-dependent manner but had no effect on glutamatergic spontaneous excitatory postsynaptic currents (sEPSCs). In contrast, for evoked synaptic responses in isolated neurons, the amplitude of both inhibitory and excitatory postsynaptic currents (eIPSCs and eEPSCs) was decreased in a concentration-dependent manner. This was associated with increases of synaptic failure rate (Rf) and paired-pulse ratio (PPR). The effect was larger for eIPSCs compared with eEPSCs. These results clearly indicate that D2O acts differently on inhibitory and excitatory neurotransmitter release machinery. Furthermore, D2O significantly suppressed GABAA receptor-mediated whole cell current (IGABA) but did not affect glutamate receptor-mediated whole cell current (IGlu). The combined effects of D2O at both the pre- and postsynaptic sites may explain the greater inhibition of eIPSCs compared with eEPSCs. Finally, D2O did not enhance or otherwise affect the actions of the general anesthetics nitrous oxide and propofol on spontaneous or evoked GABAergic and glutamatergic neurotransmissions, or on IGABA and IGlu. Our results suggest that previously reported effects of D2O to mimic and/or modulate anesthesia potency result from mechanisms other than modulation of GABAergic and glutamatergic neurotransmission.


Assuntos
Região CA3 Hipocampal/efeitos dos fármacos , Fármacos do Sistema Nervoso Central/farmacologia , Óxido de Deutério/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Região CA3 Hipocampal/fisiologia , Relação Dose-Resposta a Droga , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/fisiologia , Ácido Glutâmico/metabolismo , Potenciais Pós-Sinápticos Inibidores/fisiologia , Neurônios/fisiologia , Óxido Nitroso/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/fisiologia , Propofol/farmacologia , Ratos Wistar , Receptores de GABA-A/metabolismo , Técnicas de Cultura de Tecidos , Ácido gama-Aminobutírico/metabolismo
3.
J Pharmacol Exp Ther ; 348(2): 246-59, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24259680

RESUMO

Levetiracetam (LEV) is an antiepileptic drug with a unique but as yet not fully resolved mechanism of action. Therefore, by use of a simplified rat-isolated nerve-bouton preparation, we have investigated how LEV modulates glutamatergic transmission from mossy fiber terminals to hippocampal CA3 neurons. Action potential-evoked excitatory postsynaptic currents (eEPSCs) were recorded using a conventional whole-cell patch-clamp recording configuration in voltage-clamp mode. The antiepileptic drug phenytoin decreased glutamatergic eEPSCs in a concentration-dependent fashion by inhibiting voltage-dependent Na⁺ and Ca²âº channel currents. In contrast, LEV had no effect on eEPSCs or voltage-dependent Na⁺ or Ca²âº channel currents. Activation of presynaptic GABA type A (GABA(A)) receptors by muscimol induced presynaptic inhibition of eEPSCs, resulting from depolarization block. Low concentrations of Zn²âº, which had no effect on eEPSCs, voltage-dependent Na⁺ or Ca²âº channel currents, or glutamate receptor-mediated whole cell currents, reduced the muscimol-induced presynaptic inhibition. LEV applied in the continuous presence of 1 µM muscimol and 1 µM Zn²âº reversed this Zn²âº modulation on eEPSCs. The antagonizing effect of LEV on Zn²âº-induced presynaptic GABA(A) receptor inhibition was also observed with the Zn²âº chelators Ca-EDTA and RhodZin-3. Our results clearly show that LEV removes the Zn²âº-induced suppression of GABA(A)-mediated presynaptic inhibition, resulting in a presynaptic decrease in glutamate-mediated excitatory transmission. Our results provide a novel mechanism by which LEV may inhibit neuronal activity.


Assuntos
Anticonvulsivantes/farmacologia , Região CA3 Hipocampal/efeitos dos fármacos , Neurônios GABAérgicos/efeitos dos fármacos , Inibição Neural/efeitos dos fármacos , Piracetam/análogos & derivados , Receptores de GABA-A/metabolismo , Zinco/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Região CA3 Hipocampal/citologia , Região CA3 Hipocampal/metabolismo , Células Cultivadas , Quelantes/farmacologia , Potenciais Evocados/efeitos dos fármacos , Feminino , Agonistas de Receptores de GABA-A/farmacologia , Antagonistas de Receptores de GABA-A/farmacologia , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Levetiracetam , Masculino , Fibras Musgosas Hipocampais/efeitos dos fármacos , Fibras Musgosas Hipocampais/metabolismo , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Piracetam/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Ratos , Receptores de GABA-A/química , Zinco/química
4.
Gen Physiol Biophys ; 33(2): 145-55, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24448372

RESUMO

Human plasma contains wide variety of bioactive proteins that have proved essential in therapeutic discovery. However many human plasma proteins remain orphans with unknown biological functions. Evidences suggest that some plasma components target the respiratory system. In the present study we adapted heparin affinity chromatography to fractionate human plasma for functional bioassay. Fractions from pooled human plasma yielded particular plasma fractions with strong cough suppressing effects. Purification yielded a fraction that was finally identified as an activated blood coagulation factor fXIa using liquid chromatography-tandem mass spectrometry (LC-MS/MS) and matrix-assisted laser desorption ionization/time-of-flight mass spectrometry (MALDI/TOF-MS). The fraction almost completely suppressed coughs induced by either chemical or mechanical stimulation applied to larynx or bifurcation of guinea-pig trachea. Cough suppressing effect of the fraction and commercially available fXIa were one million times stronger than codeine and codeine only partially suppressed the mechanically triggered coughing in animal model. Recent reviews highlighted prominent shortcomings of current available antitussives, including narcotic opioids such as codeine and their unpleasant or intolerable side effects. Therefore, safer and more effective cough suppressants would be welcome, and present findings indicate that fXIa in human plasma as a very promising, new therapeutic candidate for effective antitussive action.


Assuntos
Antitussígenos/sangue , Antitussígenos/farmacologia , Tosse/tratamento farmacológico , Animais , Antitussígenos/isolamento & purificação , Antitussígenos/metabolismo , Bioensaio , Análise Química do Sangue , Capsaicina/farmacologia , Cromatografia de Afinidade , Codeína/farmacologia , Tosse/induzido quimicamente , Descoberta de Drogas , Fator XIa/isolamento & purificação , Fator XIa/metabolismo , Fator XIa/farmacologia , Cobaias , Heparina/metabolismo , Masculino , Fenômenos Mecânicos
5.
J Physiol ; 591(4): 1031-43, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23109108

RESUMO

Type A botulinum toxin blocks not only ACh release from motor nerve terminals but also central synaptic transmission, including glutamate, noradrenaline, dopamine, ATP, GABA and glycine. Neurotoxins (NTXs) are transported by both antero- and retrogradely along either motor or sensory axons for bidirectional delivery between peripheral tissues or the CNS. A newly developed type A2 NTX (A2NTX) injected into one rat foreleg muscle was transported to the contralateral muscle. This finding was consistent with the NTX traveling retrogradely via spinal neurons and then transsynaptically through motor neurons to the contralateral motor neurons within the spinal cord and on to the soleus muscle. In the present study we found that toxin injection into the rat left soleus muscle clearly induced bilateral muscle relaxation in a dose-dependent fashion, although the contralateral muscle relaxation followed the complete inhibition of toxin-injected ipsilateral muscles. The toxin-injected ipsilateral muscle relaxation was faster and stronger in A2NTX-treated rats than A1LL (BOTOX). A1LL was transported almost equally to the contralateral muscle via neural pathways and the bloodstream. In contrast, A2NTX was mainly transported to contralateral muscles via the blood. A1LL was more successfully transported to contralateral spinal neurons than A2NTX. We also demonstrated that A1LL and A2NTX were carried from peripheral to CNS and vice versa by dual antero- and retrograde axonal transport through either motor or sensory neurons.


Assuntos
Toxinas Botulínicas Tipo A/farmacologia , Músculo Esquelético/efeitos dos fármacos , Animais , Antitoxina Botulínica/farmacologia , Colchicina/farmacologia , Estimulação Elétrica , Feminino , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Contração Isométrica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Músculo Esquelético/inervação , Músculo Esquelético/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Ratos , Ratos Wistar , Substância Gelatinosa/citologia , Nervo Tibial/efeitos dos fármacos , Nervo Tibial/fisiologia
6.
Mol Pain ; 8: 58, 2012 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-22906126

RESUMO

BACKGROUND: 5-hydroxytryptamine (5-HT) is one of the major neurotransmitters widely distributed in the CNS. Several 5-HT receptor subtypes have been identified in the spinal dorsal horn which act on both pre- and postsynaptic sites of excitatory and inhibitory neurons. However, the receptor subtypes and sites of actions as well as underlying mechanism are not clarified rigorously. Several electrophysiological studies have been performed to investigate the effects of 5-HT on excitatory transmission in substantia gelatinosa (SG) of the spinal cord. In the present study, to understand the effects of 5-HT on the inhibitory synaptic transmission and to identify receptor subtypes, the blind whole cell recordings were performed from SG neurons of rat spinal cord slices. RESULTS: Bath applied 5-HT (50 µM) increased the frequency but not amplitudes of spontaneous inhibitory postsynaptic currents (sIPSCs) in 58% of neurons, and both amplitude and frequency in 23% of neurons. The frequencies of GABAergic and glycinergic mIPSCs were both enhanced. TTX (0.5 µM) had no effect on the increasing frequency, while the enhancement of amplitude of IPSCs was eliminated. Evoked-IPSCs (eIPSCs) induced by focal stimulation near the recording neurons in the presence of CNQX and APV were enhanced in amplitude by 5-HT. In the presence of Ba(2+) (1 mM), a potassium channel blocker, 5-HT had no effect on both frequency and amplitude. A 5-HT(2A) receptor agonist, TCB-2 mimicked the 5-HT effect, and ketanserin, an antagonist of 5-HT(2A) receptor, inhibited the effect of 5-HT partially and TCB-2 almost completely. A 5-HT(2C) receptor agonist WAY 161503 mimicked the 5-HT effect and this effect was blocked by a 5-HT(2C) receptor antagonist, N-desmethylclozapine. The amplitudes of sIPSCs were unaffected by 5-HT(2A) or 5-HT(2C) agonists. A 5-HT(3) receptor agonist mCPBG enhanced both amplitude and frequency of sIPSCs. This effect was blocked by a 5-HT(3) receptor antagonist ICS-205,930. The perfusion of 5-HT(2B) receptor agonist had no effect on sIPSCs. CONCLUSIONS: Our results demonstrated that 5-HT modulated the inhibitory transmission in SG by the activation of 5-HT(2A) and 5-HT(2C) receptors subtypes located predominantly at inhibitory interneuron terminals, and 5-HT(3) receptors located at inhibitory interneuron terminals and soma-dendrites, consequently enhanced both frequency and amplitude of IPSCs.


Assuntos
Potenciais Pós-Sinápticos Inibidores/fisiologia , Células do Corno Posterior/fisiologia , Receptores de Serotonina/metabolismo , Animais , Bário/farmacologia , Glicina/metabolismo , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Masculino , Neurotransmissores/metabolismo , Células do Corno Posterior/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Serotonina/farmacologia , Antagonistas da Serotonina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Substância Gelatinosa/efeitos dos fármacos , Substância Gelatinosa/fisiologia , Ácido gama-Aminobutírico/metabolismo
7.
J Pharmacol Exp Ther ; 341(3): 809-19, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22434676

RESUMO

Ethanol (EtOH) has a number of behavioral effects, including intoxication, amnesia, and/or sedation, that are thought to relate to the activation of GABA(A) receptors. However, GABA(A) receptors at different cellular locations have different sensitivities to EtOH. The present study used the "synaptic bouton" preparation where we could stimulate nerve endings on mechanically dissociated single rat hippocampal CA1 and CA3 pyramidal neurons and investigate the effects of EtOH on presynaptic and postsynaptic GABA(A) receptors. Low concentrations of EtOH (10 mM) had no effect on postsynaptic GABA(A) and glutamate receptors or voltage-dependent Na(+) and Ca(2+) channels. Higher concentrations (≥100 mM) could significantly inhibit these current responses. EtOH at 10 mM had no direct effect on inhibitory postsynaptic currents (IPSCs) and excitatory postsynaptic currents (EPSCs) evoked by focal stimulation of single boutons [evoked IPSCs (eIPSCs) and evoked EPSCs (eEPSCs)]. However, coapplication of 10 mM EtOH with muscimol decreased the amplitude of eIPSCs and eEPSCs and increased their paired-pulse ratio. The effects on eEPSCs were reversed by bicuculline. Coapplication of muscimol and EtOH significantly increased the frequency of spontaneous IPSCs and EPSCs. The EtOH effects on the postsynaptic responses and eEPSCs were similar in neurons from neonatal and mature rats. These results revealed that low concentrations of EtOH can potentiate the activation of presynaptic GABA(A) receptors to inhibit evoked GABA and glutamate release. These results indicate a high sensitivity of presynaptic GABA(A) receptor to EtOH, which needs to be accounted for when considering the cellular mechanisms of EtOH's physiological responses.


Assuntos
Anti-Infecciosos Locais/farmacologia , Etanol/farmacologia , Neurônios GABAérgicos/metabolismo , Terminações Pré-Sinápticas/efeitos dos fármacos , Receptores de GABA-A/metabolismo , Receptores de Glutamato/metabolismo , Animais , Bicuculina/farmacologia , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Região CA3 Hipocampal/efeitos dos fármacos , Região CA3 Hipocampal/metabolismo , Estimulação Elétrica , Agonistas de Receptores de GABA-A/farmacologia , Antagonistas de Receptores de GABA-A/farmacologia , Muscimol/farmacologia , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/metabolismo , Ratos , Ratos Wistar
8.
J Pharmacol Sci ; 118(1): 75-81, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-32092840

RESUMO

Our recent study showed a possibility that newly developed A2 type botulinum toxin (A2NTX) inhibits both spontaneous and evoked transmitter release from inhibitory (glycinergic or GABAergic) and excitatory (glutamatergic) nerve terminals using rat spinal sacral dorsal commissural nucleus neurons. In the present study, to determine the modulatory effect of A2NTX on glycinergic and glutamatergic release probabilities, we tested the effects of A2NTX on a single inhibitory or excitatory nerve ending adherent to a dissociated neuron that was activated by paired-pulse stimuli by using the focal electrical stimulation technique. The results of the present paired-pulse experiments showed clearly that A2NTX enhanced paired-pulse facilitation of evoked glycinergic inhibitory postsynaptic currents and glutamatergic excitatory postsynaptic currents and increased the failure rate (Rf) of the first postsynaptic currents (P1) and both the responses. These effects of A2NTX on the amplitude and Rf of the P1 and the second postsynaptic currents (P2) and paired-pulse ratio were rescued by application of 4-aminophthalimide. In summary, the present results showed that A2NTX acts purely presynaptically and inhibits the release machinery of transmitters such as glycine and glutamate, and the transmitter release machinery became less sensitive to intracellular free-Ca2+ in A2NTX poisoned nerve terminals.

9.
J Pharmacol Sci ; 118(1): 75-81, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22198005

RESUMO

Our recent study showed a possibility that newly developed A2 type botulinum toxin (A2NTX) inhibits both spontaneous and evoked transmitter release from inhibitory (glycinergic or GABAergic) and excitatory (glutamatergic) nerve terminals using rat spinal sacral dorsal commissural nucleus neurons. In the present study, to determine the modulatory effect of A2NTX on glycinergic and glutamatergic release probabilities, we tested the effects of A2NTX on a single inhibitory or excitatory nerve ending adherent to a dissociated neuron that was activated by paired-pulse stimuli by using the focal electrical stimulation technique. The results of the present paired-pulse experiments showed clearly that A2NTX enhanced paired-pulse facilitation of evoked glycinergic inhibitory postsynaptic currents and glutamatergic excitatory postsynaptic currents and increased the failure rate (Rf) of the first postsynaptic currents (P(1)) and both the responses. These effects of A2NTX on the amplitude and Rf of the P(1) and the second postsynaptic currents (P(2)) and paired-pulse ratio were rescued by application of 4-aminophthalimide. In summary, the present results showed that A2NTX acts purely presynaptically and inhibits the release machinery of transmitters such as glycine and glutamate, and the transmitter release machinery became less sensitive to intracellular free-Ca(2+) in A2NTX poisoned nerve terminals.


Assuntos
Toxinas Botulínicas Tipo A/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Neurotoxinas/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Animais , Região CA3 Hipocampal/citologia , Ácido Glutâmico/fisiologia , Glicina/fisiologia , Técnicas In Vitro , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Ratos , Ratos Wistar , Medula Espinal/citologia , Ácido gama-Aminobutírico/fisiologia
10.
J Pharmacol Sci ; 118(1): 33-42, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-32092836

RESUMO

Recent studies have demonstrated that the botulinum neurotoxins inhibit the release of acetylcholine, glutamate, GABA, and glycine in central nerve system (CNS) neurons. The Na+ current (INa) is of major interest because it acts as the trigger for many cellular functions such as transmission, secretion, contraction, and sensation. Thus, these observations raise the possibility that A type neurotoxin might also alter the INa of neuronal excitable membrane. To test our idea, we examined the effects of A type neurotoxins on INa of central and peripheral neurons. The neurotoxins in femtomolar to picomolar concentrations produced substantial decreases of the neuronal INa, but interestingly the current inhibition was saturated at about maximum 50% level of control INa. The inhibitory pattern in the concentration-response curve for the neurotoxins differed from tetrodotoxin (TTX), local anesthetic, and antiepileptic drugs that completely inhibited INa in a concentration-dependent manner. We concluded that A type neurotoxins inhibited membrane Na+-channel activity in CNS neurons and that INa of both TTX-sensitive and-insensitive peripheral dorsal ganglion cells were also inhibited similarly to a maximum 40% of the control by the neurotoxins. The results suggest evidently that A2NTX could be also used as a powerful drug in treating epilepsy and several types of pain.

11.
J Pharmacol Sci ; 118(1): 33-42, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22156364

RESUMO

Recent studies have demonstrated that the botulinum neurotoxins inhibit the release of acetylcholine, glutamate, GABA, and glycine in central nerve system (CNS) neurons. The Na(+) current (I(Na)) is of major interest because it acts as the trigger for many cellular functions such as transmission, secretion, contraction, and sensation. Thus, these observations raise the possibility that A type neurotoxin might also alter the I(Na) of neuronal excitable membrane. To test our idea, we examined the effects of A type neurotoxins on I(Na) of central and peripheral neurons. The neurotoxins in femtomolar to picomolar concentrations produced substantial decreases of the neuronal I(Na), but interestingly the current inhibition was saturated at about maximum 50% level of control I(Na). The inhibitory pattern in the concentration-response curve for the neurotoxins differed from tetrodotoxin (TTX), local anesthetic, and antiepileptic drugs that completely inhibited I(Na) in a concentration-dependent manner. We concluded that A type neurotoxins inhibited membrane Na(+)-channel activity in CNS neurons and that I(Na) of both TTX-sensitive and -insensitive peripheral dorsal ganglion cells were also inhibited similarly to a maximum 40% of the control by the neurotoxins. The results suggest evidently that A2NTX could be also used as a powerful drug in treating epilepsy and several types of pain.


Assuntos
Toxinas Botulínicas Tipo A/farmacologia , Região CA1 Hipocampal/fisiologia , Gânglios Espinais/fisiologia , Neurônios/efeitos dos fármacos , Neurotoxinas/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Animais , Região CA1 Hipocampal/citologia , Gânglios Espinais/citologia , Neurônios/fisiologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Ratos , Ratos Wistar , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/fisiologia , Tetrodotoxina/farmacologia
12.
Chemosphere ; 184: 337-346, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28605704

RESUMO

4,5-Dichloro-2-octyl-4-isothiazolin-3-one (DCOIT) is an alternative to organotin antifoulants, such as tributyltin and triphenyltin. Since DCOIT is found in harbors, bays, and coastal areas worldwide, this chemical compound may have some impacts on ecosystems. To determine whether DCOIT possesses neurotoxic activity by modifying synaptic transmission, we examined the effects of DCOIT on synaptic transmission in a 'synaptic bouton' preparation of rat brain. DCOIT at concentrations of 0.03-1 µM increased the amplitudes of evoked synaptic currents mediated by GABA and glutamate, while it reduced the amplitudes of these currents at 3-10 µM. However, the currents elicited by exogenous applications of GABA and glutamate were not affected by DCOIT. DCOIT at 1-10 µM increased the frequency of spontaneous synaptic currents mediated by GABA. It also increased the frequency of glutamate-mediated spontaneous currents at0.3-10 µM. The frequencies of miniature synaptic currents mediated by GABA and glutamate, observed in the presence of tetrodotoxin under external Ca2+-free conditions, were increased by 10 µM DCOIT. With the repetitive applications of DCOIT, the frequency of miniature synaptic currents mediated by glutamate was not increased by the second and third applications of DCOIT. Voltage-dependent Ca2+ channels were not affected by DCOIT, but DCOIT slowed the inactivation of voltage-dependent Na+ channels. These results suggest that DCOIT increases Ca2+ release from intracellular Ca2+ stores, resulting in the facilitation of both action potential-dependent and spontaneous neurotransmission, possibly leading to neurotoxicity.


Assuntos
Região CA3 Hipocampal/patologia , Neurônios/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Tiazóis/farmacologia , Animais , Cálcio/metabolismo , Ecossistema , Poluentes Ambientais/farmacologia , Masculino , Neurônios/efeitos dos fármacos , Terminações Pré-Sinápticas/efeitos dos fármacos , Ratos , Poluentes Químicos da Água/farmacologia
13.
Brain Res ; 1631: 147-56, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26616339

RESUMO

The effects of the intravenous anesthetic, propofol, on glycinergic transmission and on glycine receptor-mediated whole-cell currents (IGly) were examined in the substantia gelatinosa (SG) neuronal cell body, mechanically dissociated from the rat spinal cord. This "synaptic bouton" preparation, which retains functional native nerve endings, allowed us to evaluate glycinergic inhibitory postsynaptic currents (IPSCs) and whole-cell currents in a preparation in which experimental solution could rapidly access synaptic terminals. Synaptic IPSCs were measured as spontaneous (s) and evoked (e) IPSCs. The eIPSCs were elicited by applying paired-pulse focal electrical stimulation, while IGly was evoked by a bath application of glycine. A concentration-dependent enhancement of IGly was observed for ≥10µM propofol. Propofol (≥3µM) significantly increased the frequency of sIPSCs and prolonged the decay time without altering the current amplitude. However, propofol (≥3µM) also significantly increased the mean amplitude of eIPSCs and decreased the failure rate (Rf). A decrease in the paired-pulse ratio (PPR) was noted at higher concentrations (≥10µM). The decay time of eIPSCs was prolonged only at the maximum concentration tested (30µM). Propofol thus acts at both presynaptic glycine release machinery and postsynaptic glycine receptors. At clinically relevant concentrations (<1µM) there was no effect on IGly, sIPSCs or eIPSCs suggesting that at anesthetic doses propofol does not affect inhibitory glycinergic synapses in the spinal cord.


Assuntos
Glicinérgicos/farmacologia , Neurônios/efeitos dos fármacos , Propofol/farmacologia , Sinapses/efeitos dos fármacos , Anestésicos Intravenosos/farmacologia , Animais , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Glicina/metabolismo , Potenciais Pós-Sinápticos Inibidores/fisiologia , Masculino , Neurônios/metabolismo , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de Glicina/metabolismo , Medula Espinal/efeitos dos fármacos , Sinapses/metabolismo , Transmissão Sináptica/fisiologia
14.
Toxicon ; 103: 12-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25959619

RESUMO

The present study utilised a 'synaptic bouton' preparation of mechanically isolated rat hippocampal CA3 pyramidal neurons, which permits direct physiological and pharmacological quantitative analyses at the excitatory and inhibitory single synapse level. Evoked excitatory and inhibitory postsynaptic currents (eEPSCs and eIPSCs) were generated by focal paired-pulse electrical stimulation of single boutons. The sensitivity of eEPSC to tetrodotoxin (TTX) was higher than that of the voltage-dependent Na(+) channel whole-cell current (INa) in the postsynaptic CA3 soma membrane. The synaptic transmission was strongly inhibited by 3 nM TTX, at which concentration the INa was hardly suppressed. The IC50 values of eEPSC and INa for TTX were 2.8 and 37.9 nM, respectively, and complete inhibition was 3-10 nM for eEPSC and 1000 nM for INa. On the other hand, both eEPSC and eIPSC were equally and gradually inhibited by decreasing the external Na(+) concentration ([Na]o), which decreases the Na(+)gradient across the cell membrane. The results indicate that TTX at 3-10 nM could block most of voltage-dependent Na(+) channels on presynaptic nerve terminal, resulting in abruptly inhibition of action potential dependent excitatory neurotransmission.


Assuntos
Sistema Nervoso Central/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Tetrodotoxina/toxicidade , Animais , Sistema Nervoso Central/metabolismo , Estimulação Elétrica , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Concentração Inibidora 50 , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Ratos , Ratos Wistar , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo , Ácido gama-Aminobutírico/metabolismo
15.
Brain Res Bull ; 118: 34-45, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26343381

RESUMO

We evaluated the effects of N2O on synaptic transmission using a preparation of mechanically dissociated rat hippocampal CA3 neurons that allowed assays of single bouton responses evoked from native functional nerve endings. We studied the effects of N2O on GABAA, glutamate, AMPA and NMDA receptor-mediated currents (IGABA, IGlu, IAMPA and INMDA) elicited by exogenous application of GABA, glutamate, (S)-AMPA, and NMDA and spontaneous, miniature, and evoked GABAergic inhibitory and glutamatergic excitatory postsynaptic current (sIPSC, mIPSC, eIPSC, sEPSC, mEPSC and eEPSC) in mechanically dissociated CA3 neurons. eIPSC and eEPSC were evoked by focal electrical stimulation of a single bouton. Administration of 70% N2O altered neither IGABA nor the frequency and amplitude of both sIPSCs and mIPSCs. In contrast, N2O decreased the amplitude of eIPSCs, while increasing failure rates (Rf) and paired-pulse ratios (PPR) in a concentration-dependent manner. On the other hand, N2O decreased IGlu, IAMPA and INMDA. Again N2O did not change the frequency and amplitude of either sEPSCs of mEPSCs. N2O also decreased amplitudes of eEPSCs with increased Rf and PPR. The decay phases of all synaptic responses were unchanged. The present results indicated that N2O inhibits the activation of AMPA/KA and NMDA receptors and also that N2O preferentially depress the action potential-dependent GABA and glutamate releases but had little effects on spontaneous and miniature releases.


Assuntos
Região CA3 Hipocampal/efeitos dos fármacos , Óxido Nitroso/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Estimulação Elétrica , Feminino , Neurônios GABAérgicos/efeitos dos fármacos , Ácido Glutâmico/farmacologia , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Masculino , N-Metilaspartato/farmacologia , Ratos , Ratos Wistar , Transmissão Sináptica/efeitos dos fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia , Ácido gama-Aminobutírico/farmacologia
16.
Chemosphere ; 120: 598-607, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25462303

RESUMO

Triphenyltin (TPT) is an organometallic compound that poses a known environmental hazard to some fish and mollusks, as well as mammals. However, its neurotoxic mechanisms in the mammalian brain are still unclear. Thus, we have investigated mechanisms through which TPT modulates glutamatergic synaptic transmission, including spontaneous, miniature, and evoked excitatory postsynaptic currents (sEPSCs, mEPSCs, and eEPSCs respectively), in a rat hippocampal CA3 'synaptic-bouton' preparation. TPT, at environmentally relevant concentrations (30 nM to 1 µM), significantly increased the frequency of sEPSCs and mEPSCs in a concentration-dependent manner, without affecting the currents' amplitudes. The facilitatory effects of TPT on mEPSC frequency were seen even in a Ca(2+)-free external solution containing tetrodotoxin. These effects were further prolonged by adding caffeine, which releases Ca(2+) from intracellular Ca(2+) storage sites. In glutamatergic eEPSCs evoked by paired-pulse stimuli, TPT at concentrations greater than or equal to 100 nM markedly increased the current amplitude by the first pulse and decreased failure rate and pair-pulse ratio. On the other hand, both voltage-dependent Na(+) and Ca(2+) channels were unaffected by submicromolar concentrations of TPT. Overall, these results suggest that TPT, at environmentally relevant concentrations, affects presynaptic transmitter release machinery by directly modulating Ca(2+) storage. Further, findings of this study imply that excitotoxic mechanisms may underlie TPT-induced neuronal damage.


Assuntos
Cálcio/metabolismo , Poluentes Ambientais/toxicidade , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Compostos Orgânicos de Estanho/toxicidade , Terminações Pré-Sinápticas/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Análise de Variância , Animais , Cafeína/farmacologia , Hipocampo/citologia , Japão , Masculino , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Transmissão Sináptica/efeitos dos fármacos , Tetrodotoxina/química
17.
Brain Res Bull ; 117: 39-44, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26247839

RESUMO

The acute effects of high-dose Li(+) treatment on glutamatergic and GABAergic transmissions were studied in the "synaptic bouton" preparation of isolated rat hippocampal pyramidal neurons by using focal electrical stimulation. Both action potential-dependent glutamatergic excitatory and GABAergic inhibitory postsynaptic currents (eEPSC and eIPSC, respectively) were dose-dependently inhibited in the external media containing 30-150 mM Li(+), but the sensitivity for Li(+) was greater tendency for eEPSCs than for eIPSCs. When the effects of Li(+) on glutamate or GABAA receptor-mediated whole-cell responses (IGlu and IGABA) elicited by an exogenous application of glutamate or GABA were examined in the postsynaptic soma membrane of CA3 neurons, Li(+) slightly inhibited both IGlu and IGABA at the 150 mM Li(+) concentration. Present results suggest that acute treatment with high concentrations of Li(+) acts preferentially on presynaptic terminals, and that the Li(+)-induced inhibition may be greater for excitatory than for inhibitory transmission.


Assuntos
Fármacos do Sistema Nervoso Central/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Compostos de Lítio/farmacologia , Células Piramidais/efeitos dos fármacos , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/fisiologia , Região CA3 Hipocampal/efeitos dos fármacos , Região CA3 Hipocampal/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/fisiologia , Ácido Glutâmico/metabolismo , Potenciais Pós-Sinápticos Inibidores/fisiologia , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/fisiologia , Células Piramidais/fisiologia , Ratos Wistar , Ácido gama-Aminobutírico/metabolismo
18.
Brain Res Bull ; 93: 39-46, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23174309

RESUMO

Neurosteroids such as allopregnanolone (Allo) are widely distributed in the brain and may modulate neuronal excitability under physiological or pathological states. Allo modulates GABAA receptor responses, and in this study we investigated the functional effects of Allo on presynaptic GABAA receptors on single glutamatergic nerve terminal projecting on CA3 neurons. In the present study, we measured spontaneous and evoked excitatory postsynaptic currents (sEPSCs and eEPSCs), the latter was elicited with single or paired-pulse focal electrical stimulation, using mechanically isolated 'synaptic bouton' preparation. Allo (10 nM) increased significantly eEPSC amplitude while decreasing the failure rate (Rf) and the paired-pulse response ratio (PPR). Conversely high concentration (100 nM) of Allo decreased eEPSC amplitude and increased Rf and PPR. Allo also increased significantly the frequency and amplitude of sEPSCs at low concentrations (10-30 nM) but at high concentration (100 nM) it had no effect on current amplitude but modestly decreased sEPSC frequency. Application of Allo at nanomolar concentrations facilitated exogenous muscimol-induced outward postsynaptic currents but had no effect on glutamate-induced inward postsynaptic currents. Our results demonstrate that Allo modulates glutamate release via presynaptic GABAA receptors, in addition to its better characterized effects to modulate postsynaptic GABAA responses. Both pre- and postsynaptic GABAA receptor modulation is likely to contribute to the physiological actions of neurosteroids.


Assuntos
Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Pregnanolona/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Bicuculina/farmacologia , Biofísica , Região CA1 Hipocampal/citologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Estimulação Elétrica , Antagonistas de Aminoácidos Excitatórios/farmacologia , Agonistas de Receptores de GABA-A/farmacologia , Antagonistas de Receptores de GABA-A/farmacologia , Ácido Glutâmico/farmacologia , Muscimol/farmacologia , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Terminações Pré-Sinápticas/fisiologia , Ratos , Ratos Wistar , Sinapses/ultraestrutura , Tetraetilamônio/farmacologia , Valina/análogos & derivados , Valina/farmacologia
19.
Brain Res Bull ; 90: 10-8, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23026118

RESUMO

Pentobarbital (PB) modulates GABA(A) receptor-mediated postsynaptic responses through various mechanisms, and can directly activate the channel at higher doses. These channels exist both pre- and postsynaptically, and on the soma outside the synapse. PB also inhibits voltage-dependent Na⁺ and Ca²âº channels to decrease excitatory synaptic transmission. Just how these different sites of action combine to contribute to the overall effects of PB on inhibitory and excitatory synaptic transmission is less clear. To compare these pre- and postsynaptic actions of PB, we used a 'synaptic bouton' preparation of isolated rat hippocampal CA3 pyramidal neurons where we could measure in single neurons the effects of PB on spontaneous and single bouton evoked GABAergic inhibitory and glutamatergic excitatory postsynaptic currents (sIPSCs, sEPSCs, eIPSCs and eEPSCs), respectively. Low (sedative) concentrations (3-10 µM) of PB increased the frequency and amplitude of sIPSCs and sEPSCs, and also presynaptically increased the amplitude of both eIPSCs and eEPSCs. There was no change in current kinetics at this low concentration. At higher concentrations (30-300 µM), PB decreased the frequency, and increased the amplitude of sIPSCs, and presynaptically decreased the amplitude of eIPSCs. The current decay phase of sIPSCs and eIPSCs was increased. An increase in both frequency and amplitude was seen for sEPSCs, while the eIPSCs was also decreased by a bicuculline-sensitive presynaptic effect. The results confirm the multiple sites of action of PB on inhibitory and excitatory transmission and demonstrate that the most sensitive site of action is on transmitter release, via effects on presynaptic GABA(A) receptors. At low concentrations, however, both glutamate and GABA release is similarly enhanced, making the final effects on neuronal excitability difficult to predict and dependent on the particular systems involved and/or on subtle differences in susceptibility amongst individuals. At higher concentrations, release of both transmitters is decreased, while the postsynaptic effects to increase IPSPs and decrease EPSCs would be expected to both results in reduced neuronal excitability.


Assuntos
Região CA3 Hipocampal/citologia , Moduladores GABAérgicos/farmacologia , Neurônios/citologia , Neurotransmissores/metabolismo , Pentobarbital/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Fenômenos Biofísicos/efeitos dos fármacos , Biofísica , Relação Dose-Resposta a Droga , Estimulação Elétrica , Potenciais Evocados/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Ácido Glutâmico/farmacologia , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar , Fatores de Tempo , Ácido gama-Aminobutírico/farmacologia
20.
Eur J Pharmacol ; 718(1-3): 63-73, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24051267

RESUMO

We evaluated the effects of propofol on synaptic transmission using a mechanically dissociated preparation of rat hippocampal CA3 neurons to allow assays of single bouton responses evoked from retained functional native nerve endings. We studied synaptic and extrasynaptic GABAA and glutamate receptor responses in a preparation in which experimental solutions rapidly accessed synaptic terminals. Whole-cell responses were evoked by bath application of GABA and glutamate. Synaptic inhibitory and excitatory postsynaptic currents (IPSC and EPSC) were measured as spontaneous and evoked postsynaptic responses. Evoked currents were elicited by focal electrical stimulation. Propofol (1-100 µM) enhanced extrasynaptic GABAA-receptor mediated responses but the increase at clinically relevant concentrations (1 µM) were minor. In contrast, 1 µM propofol significantly increased both the amplitude and frequency of spontaneous IPSCs (sIPSCs) and increased the amplitudes of evoked IPSCs (eIPSCs) while decreasing failure rates (Rf) and paired-pulse ratios (PPR). Decay times of sIPSCs and eIPSCs were significantly prolonged. Although propofol had no effect on extrasynaptic glutamate responses, only supra-clinical propofol concentrations (≥ 10 µM) increased the spontaneous EPSCs (sEPSCs, amplitudes and frequencies) but suppressed evoked EPSCs (eEPSCs decreased amplitudes with increased Rf and PPR). The decay phases of sEPSCs and eEPSCs were not changed. The propofol-induced changes in sEPSCs and eEPSCs resulted from presynaptic GABAA receptor-mediated depolarization, because these actions were blocked by bicuculline. These results suggest that propofol acts at presynaptic and postsynaptic GABAA receptors within GABAergic synapses, but also increases extrasynaptic GABA responses. Our results expand the locus of propofol actions to GABAergic and glutamatergic synapses.


Assuntos
Ácido Glutâmico/metabolismo , Hipocampo/citologia , Neurônios/citologia , Propofol/farmacologia , Sinapses/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo , Animais , Cálcio/metabolismo , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Hipocampo/fisiologia , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar , Sódio/metabolismo , Sinapses/fisiologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA