Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Neurosci ; 36(34): 8957-66, 2016 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-27559176

RESUMO

UNLABELLED: Most craniosensory afferents have unmyelinated axons expressing TRP Vanilloid 1 (TRPV1) receptors in synaptic terminals at the solitary tract nucleus (NTS). Neurotransmission from these synapses is characterized by substantial asynchronous EPSCs following action potential-synched EPSCs and high spontaneous rates that are thermally sensitive. The present studies blocked voltage-activated calcium channels (CaV) using the nonselective CaV blocker Cd(2+) or the specific N-type blocker ω-conotoxin GVIA to examine the calcium dependence of the synchronous, asynchronous, spontaneous, and thermally gated modes of release. In rat brainstem slices containing caudal NTS, shocks to the solitary tract (ST) triggered synchronous ST-EPSCs and trailing asynchronous EPSCs. Cd(2+) or GVIA efficiently reduced both synchronous and asynchronous EPSCs without altering spontaneous or thermal-evoked transmission. Activation of TRPV1 with either the selective agonist resiniferatoxin (150 pm) or temperature augmented basal sEPSC rates but failed to alter the synchronous or asynchronous modes of release. These data indicate that calcium sourced through TRPV1 has no access to the synchronous or asynchronous release mechanism(s) and conversely that CaV-sourced calcium does not interact with the thermally evoked mode of release. Buffering intracellular calcium with EGTA-AM or BAPTA-AM reduced asynchronous EPSC rates earlier and to a greater extent than synchronous ST-EPSC amplitudes without altering sEPSCs or thermal sensitivity. Buffering therefore distinguishes asynchronous vesicles as possessing a highly sensitive calcium sensor located perhaps more distant from CaV than synchronous vesicles or thermally evoked vesicles from TRPV1. Together, our findings suggest separate mechanisms of release for spontaneous, asynchronous and synchronous vesicles that likely reside in unique, spatially separated vesicle domains. SIGNIFICANCE STATEMENT: Most craniosensory fibers release glutamate using calcium entry from two sources: CaVs and TRPV1. We demonstrate that calcium segregation distinguishes three vesicle release mechanisms. Most surprisingly, asynchronous release is associated with CaV and not TRPV1 calcium entry. This reveals that asynchronous release is an additional and separate phenotypic marker of unmyelinated afferents rather than operated by TRPV1. The functional independence of the two calcium sources expands the regulatory repertoire of transmission and imbues these inputs with additional modulation targets for synaptic release not present at conventional CaV synapses. Peptides and lipid mediators may target one or both of these calcium sources at afferent terminals within the solitary tract nucleus to independently modify release from distinct, functionally segregated vesicle pools.


Assuntos
Vias Aferentes/fisiologia , Cálcio/metabolismo , Neurônios/fisiologia , Sinapses/metabolismo , Acetilcolina/metabolismo , Análise de Variância , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Quelantes/farmacologia , Diterpenos/farmacologia , Relação Dose-Resposta a Droga , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Ácido Glutâmico/metabolismo , Técnicas In Vitro , Masculino , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Núcleo Solitário/citologia , Sinapses/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo
2.
J Neurosci ; 34(24): 8324-32, 2014 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-24920635

RESUMO

Action potentials trigger synaptic terminals to synchronously release vesicles, but some vesicles release spontaneously. G-protein-coupled receptors (GPCRs) can modulate both of these processes. At cranial primary afferent terminals, the GPCR cannabinoid 1 (CB1) is often coexpressed with transient receptor potential vanilloid 1 (TRPV1), a nonselective cation channel present on most afferents. Here we tested whether CB1 activation modulates synchronous, action potential-evoked (eEPSCs) and/or spontaneous (sEPSCs) EPSCs at solitary tract nucleus neurons. In rat horizontal brainstem slices, activation of solitary tract (ST) primary afferents generated ST-eEPSCs that were rapidly and reversibly inhibited from most afferents by activation of CB1 with arachidonyl-2'-chloroethylamide (ACEA) or WIN 55,212-2 [R-(+)-(2,3-dihydro-5-methyl-3-[(4-morpholinyl)methyl]pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl)(1-naphthalenyl) methanone monomethanesulfonate]. The CB1 antagonist/inverse agonist AM251 [N-1-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-1-piperidinyl-1H-pyrazole-3-carboxamide] blocked these responses. Despite profound depression of ST-eEPSCs during CB1 activation, sEPSCs in these same neurons were unaltered. Changes in temperature changed sEPSC frequency only from TRPV1(+) afferents (i.e., thermal sEPSC responses only occurred in TRPV1(+) afferents). CB1 activation failed to alter these thermal sEPSC responses. However, the endogenous arachidonate metabolite N-arachidonyldopamine (NADA) promiscuously activated both CB1 and TRPV1 receptors. NADA inhibited ST-eEPSCs while simultaneously increasing sEPSC frequency, and thermally triggered sEPSC increases in neurons with TRPV1(+) afferents. We found no evidence for CB1/TRPV1 interactions suggesting independent regulation of two separate vesicle pools. Together, these data demonstrate that action potential-evoked synchronous glutamate release is modulated separately from TRPV1-mediated glutamate release despite coexistence in the same central terminations. This two-pool arrangement allows independent and opposite modulation of glutamate release by single lipid metabolites.


Assuntos
Ácido Glutâmico/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Transmissão Sináptica/fisiologia , Canais de Cátion TRPV/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Analgésicos/farmacologia , Animais , Ácidos Araquidônicos/farmacologia , Benzoxazinas/farmacologia , Tronco Encefálico/citologia , Tronco Encefálico/fisiologia , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Dopamina/análogos & derivados , Dopamina/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Temperatura Alta , Técnicas In Vitro , Masculino , Morfolinas/farmacologia , Naftalenos/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Piperidinas/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/antagonistas & inibidores , Transmissão Sináptica/efeitos dos fármacos , Canais de Cátion TRPV/antagonistas & inibidores
3.
J Neurophysiol ; 112(11): 2697-706, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25185814

RESUMO

The cell-impermeant lidocaine derivative QX-314 blocks sodium channels via intracellular mechanisms. In somatosensory nociceptive neurons, open transient receptor potential vanilloid type 1 (TRPV1) receptors provide a transmembrane passageway for QX-314 to produce long-lasting analgesia. Many cranial primary afferents express TRPV1 at synapses on neurons in the nucleus of the solitary tract and caudal trigeminal nucleus (Vc). Here, we investigated whether QX-314 interrupts neurotransmission from primary afferents in rat brain-stem slices. Shocks to the solitary tract (ST) activated highly synchronous evoked excitatory postsynaptic currents (ST-EPSCs). Application of 300 µM QX-314 increased the ST-EPSC latency from TRPV1+ ST afferents, but, surprisingly, it had similar actions at TRPV1- ST afferents. Continued exposure to QX-314 blocked evoked ST-EPSCs at both afferent types. Neither the time to onset of latency changes nor the time to ST-EPSC failure differed between responses for TRPV1+ and TRPV1- inputs. Likewise, the TRPV1 antagonist capsazepine failed to prevent the actions of QX-314. Whereas QX-314 blocked ST-evoked release, the frequency and amplitude of spontaneous EPSCs remained unaltered. In neurons exposed to QX-314, intracellular current injection evoked action potentials suggesting a presynaptic site of action. QX-314 acted similarly at Vc neurons to increase latency and block EPSCs evoked from trigeminal tract afferents. Our results demonstrate that QX-314 blocked nerve conduction in cranial primary afferents without interrupting the glutamate release mechanism or generation of postsynaptic action potentials. The TRPV1 independence suggests that QX-314 either acted extracellularly or more likely entered these axons through an undetermined pathway common to all cranial primary afferents.


Assuntos
Nervos Cranianos/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores , Lidocaína/análogos & derivados , Neurônios Aferentes/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo , Anestésicos Locais/farmacologia , Animais , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Nervos Cranianos/metabolismo , Nervos Cranianos/fisiologia , Lidocaína/farmacologia , Masculino , Neurônios Aferentes/metabolismo , Neurônios Aferentes/fisiologia , Ratos , Ratos Sprague-Dawley , Tempo de Reação , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/genética
4.
Am J Physiol Regul Integr Comp Physiol ; 303(12): R1207-16, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23076872

RESUMO

Cranial primary afferent sensory neurons figure importantly in homeostatic control of visceral organ systems. Of the two broad classes of visceral afferents, the role of unmyelinated or C-type class remains poorly understood. This review contrasts key aspects of peripheral discharge properties of C-fiber afferents and their glutamate transmission mechanisms within the solitary tract nucleus (NTS). During normal prevailing conditions, most information arrives at the NTS through myelinated A-type nerves. However, most of visceral afferent axons (75-90%) in NTS are unmyelinated, C-type axons. Centrally, C-type solitary tract (ST) afferent terminals have presynaptic transient receptor potential vanilloid type 1 (TRPV1) receptors. Capsaicin activation of TRPV1 blocks phasic or synchronous release of glutamate but facilitates release of glutamate from a separate pool of vesicles. This TRPV1-operated pool of vesicles is active at normal temperatures and is responsible for actively driving a 10-fold higher release of glutamate at TRPV1 compared with TRPV1- terminals even in the absence of afferent action potentials. This novel TRPV1 mechanism is responsible for an additional asynchronous release of glutamate that is not present in myelinated terminals. The NTS is rich with presynaptic G protein-coupled receptors, and the implications of TRPV1-operated glutamate offer unique targets for signaling in C-type sensory afferent terminals from neuropeptides, inflammatory mediators, lipid metabolites, cytokines, and cannabinoids. From a homeostatic view, this combination could have broad implications for integration in chronic pathological disturbances in which the numeric dominance of C-type endings and TRPV1 would broadly disturb multisystem control mechanisms.


Assuntos
Fibras Nervosas Amielínicas/fisiologia , Neurônios Aferentes/fisiologia , Núcleo Solitário/fisiologia , Transmissão Sináptica/fisiologia , Canais de Cátion TRPV/fisiologia , Potenciais de Ação/fisiologia , Animais , Glutamatos/metabolismo , Humanos , Camundongos , Camundongos Knockout , Modelos Animais , Canais de Cátion TRPV/deficiência , Canais de Cátion TRPV/genética
5.
J Physiol ; 589(Pt 15): 3801-21, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21646412

RESUMO

We report a novel excitatory effect of cannabinoid agonists on action potential-independent GABAergic transmission in the rat dentate gyrus. Specifically, we find that both WIN55,212-2 and anandamide increase the frequency of miniature IPSCs (mIPSCs)recorded from hilar mossy cells without altering event amplitude, area, rise time, or decay. The effect of WIN55,212-2 on mIPSCs is insensitive to AM251 and preserved in CB1 −/− animals,indicating that it does not depend on activation of CB1 receptors. It is also insensitive to AM630 and unaffected by capsazepine suggesting that neither CB2 nor TRPV1 receptors are involved. Further, it is blocked by pre-incubation in suramin and by a selective protein kinase A inhibitor (H-89), and is mimicked (and occluded) by bath application of forskolin. Similar CB1 receptor-independent facilitation of exocytosis is not apparent when recording evoked IPSCs in the presence of AM251, suggesting that the exocytotic mechanism that produces WIN55,212-2 sensitive mIPSCs is distinct from that which produces CB1 sensitive and action potential-dependent release. Despite clear independence from action potentials, WIN55,212-2 mediated facilitation of mIPSCs requires calcium, and yet is insensitive to chelation of calcium in the postsynaptic cell. Finally, we demonstrate that both bath application of 2-arachidonoylglycerol(2-AG) and depolarization-induced release of endogenous cannabinoids have minimal effect on mIPSC frequency. Cumulatively, our results indicate that cannabinoid ligands can selectively facilitate action potential-independent exocytosis of GABA in the rat dentate gyrus, and further emphasize that this new cannabinoid sensitive signalling system is distinct from previously described CB1 receptor-dependent systems in numerous respects.


Assuntos
Canabinoides/metabolismo , Fibras Musgosas Hipocampais/fisiologia , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/metabolismo , Ácido gama-Aminobutírico/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Ácidos Araquidônicos/farmacologia , Benzoxazinas/farmacologia , Cálcio/metabolismo , Canabinoides/agonistas , Endocanabinoides , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/fisiologia , Glicerídeos/farmacologia , Masculino , Morfolinas/farmacologia , Fibras Musgosas Hipocampais/efeitos dos fármacos , Naftalenos/farmacologia , Piperidinas/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/antagonistas & inibidores , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
6.
Neuropharmacology ; 55(7): 1140-6, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18675282

RESUMO

Recent advances in immunohistochemical techniques have, contrary to earlier reports, positively identified CB1 receptors on glutamatergic terminals in the hippocampus. Further work has implicated these receptors in modulation of susceptibility to kainic acid induced seizures. Based on these results, the current study was designed to test the hypothesis that both exogenous and endogenous cannabinoids can selectively modulate glutamatergic afferents to CA3 pyramidal cells, and that such modulation is mediated by cannabinoid type 1 (CB1) receptors. Towards that end we employed either conventional or two-photon guided minimal stimulation techniques to isolate mossy fiber and/or associational/commissural (A/C) inputs to CA3 pyramidal cells. We report that bath application of WIN55,212-2 selectively inhibits minimally evoked A/C inputs to CA3 pyramidal cells, without significantly altering simultaneously recorded mossy fiber inputs. Further, we find that WIN55,212-2 mediated inhibition of A/C inputs is completely blocked by the CB1 selective antagonist AM-251 and absent in CB1(-/-) animals, suggesting a dependence on CB1 receptors. Finally, we demonstrate that depolarization of CA3 pyramidal cells leads to calcium dependent release of endogenous cannabinoids that transiently inhibit A/C mediated responses, and that this effect is also sensitive to both AM-251 and the muscarinic acetylcholine receptor antagonist atropine. To our knowledge this represents the first demonstration of depolarization induced suppression of excitation in area CA3 of the hippocampus. Collectively, these results provide new information relevant to developing a thorough understanding of how ECs modulate excitatory transmission in an area that is both essential for the acquisition of new memories and intimately involved in epileptogenesis.


Assuntos
Neurônios Eferentes/fisiologia , Células Piramidais/fisiologia , Receptor CB1 de Canabinoide/fisiologia , Transmissão Sináptica/fisiologia , Animais , Cálcio/metabolismo , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibras Musgosas Hipocampais/fisiologia , Neurônios Eferentes/efeitos dos fármacos , Técnicas de Patch-Clamp , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/efeitos dos fármacos , Receptor CB1 de Canabinoide/genética , Receptores Muscarínicos/efeitos dos fármacos
7.
Neuropsychopharmacology ; 43(6): 1395-1404, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29317777

RESUMO

Previous work indicated that activation of D1-like dopamine receptors (D1DRs) in the nucleus accumbens shell promoted cocaine seeking through a process involving the activation of PKA and GluA1-containing AMPA receptors (AMPARs). A-kinase anchoring proteins (AKAPs) localize PKA to AMPARs leading to enhanced phosphorylation of GluA1. AKAP150, the most well-characterized isoform, plays an important role in several forms of neuronal plasticity. However, its involvement in drug addiction has been minimally explored. Here we examine the role of AKAP150 in cocaine reinstatement, an animal model of relapse. We show that blockade of PKA binding to AKAPs in the nucleus accumbens shell of Sprague-Dawley rats attenuates reinstatement induced by either cocaine or a D1DR agonist. Moreover, this effect is specific to AKAP150, as viral overexpression of a PKA-binding deficient mutant of AKAP150 also impairs cocaine reinstatement. This viral-mediated attenuation of cocaine reinstatement was accompanied by decreased phosphorylation of GluA1-containing AMPARs and attenuated AMPAR eEPSCs. Collectively, these results suggest that AKAP150 facilitates the reinstatement of cocaine-seeking behavior by amplifying D1DR/PKA-dependent AMPA transmission in the nucleus accumbens.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Núcleo Accumbens/metabolismo , Receptores de AMPA/metabolismo , Transmissão Sináptica/fisiologia , Proteínas de Ancoragem à Quinase A/genética , Animais , Cocaína/administração & dosagem , Sacarose Alimentar , Modelos Animais de Doenças , Inibidores da Captação de Dopamina/administração & dosagem , Comportamento de Procura de Droga/efeitos dos fármacos , Comportamento de Procura de Droga/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Vetores Genéticos/administração & dosagem , Núcleo Accumbens/efeitos dos fármacos , Ratos Sprague-Dawley , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/metabolismo , Recidiva , Autoadministração , Transmissão Sináptica/efeitos dos fármacos
8.
PLoS One ; 12(3): e0174915, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28358887

RESUMO

The recycling of vesicle membrane fused during exocytosis is essential to maintaining neurotransmission. The GTPase dynamin is involved in pinching off membrane to complete endocytosis and can be inhibited by dynasore resulting in activity-dependent depletion of release-competent synaptic vesicles. In rat brainstem slices, we examined the effects of dynasore on three different modes of glutamate release-spontaneous, evoked, and asynchronous release-at solitary tract (ST) inputs to neurons in the nucleus of the solitary tract (NTS). Intermittent bursts of stimuli to the ST interspersed with pauses in stimulation allowed examination of these three modes in each neuron continuously. Application of 100 µM dynasore rapidly increased the spontaneous EPSC (sEPSC) frequency which was followed by inhibition of both ST-evoked EPSCs (ST-EPSC) as well as asynchronous EPSCs. The onset of ST-EPSC failures was not accompanied by amplitude reduction-a pattern more consistent with conduction block than reduced probability of vesicle release. Neither result suggested that dynasore interrupted endocytosis. The dynasore response profile resembled intense presynaptic TRPV1 activation. The TRPV1 antagonist capsazepine failed to prevent dynasore increases in sEPSC frequency but did prevent the block of the ST-EPSC. In contrast, the TRPV1 antagonist JNJ 17203212 prevented both actions of dynasore in neurons with TRPV1-expressing ST inputs. In a neuron lacking TRPV1-expressing ST inputs, however, dynasore promptly increased sEPSC rate followed by block of ST-evoked EPSCs. Together our results suggest that dynasore actions on ST-NTS transmission are TRPV1-independent and changes in glutamatergic transmission are not consistent with changes in vesicle recycling and endocytosis.


Assuntos
Hidrazonas/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Fibras Aferentes Viscerais/efeitos dos fármacos , Fibras Aferentes Viscerais/metabolismo , Aminopiridinas/farmacologia , Animais , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Masculino , Técnicas de Patch-Clamp , Piperazinas/farmacologia , Ratos , Ratos Sprague-Dawley , Núcleo Solitário/efeitos dos fármacos , Núcleo Solitário/metabolismo , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo
9.
Neuropharmacology ; 101: 401-11, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26471418

RESUMO

Vanilloids, high temperature, and low pH activate the transient receptor potential vanilloid type 1 (TRPV1) receptor. In spinal dorsal root ganglia, co-activation of one of these gating sites on TRPV1 sensitized receptor gating by other modes. Here in rat brainstem slices, we examined glutamate synaptic transmission in nucleus of the solitary tract (NTS) neurons where most cranial primary afferents express TRPV1, but TRPV1 sensitization is unknown. Electrical shocks to the solitary tract (ST) evoked EPSCs (ST-EPSCs). Activation of TRPV1 with capsaicin (100 nM) increased spontaneous EPSCs (sEPSCs) but inhibited ST-EPSCs. High concentrations of the ultra-potent vanilloid resiniferatoxin (RTX, 1 nM) similarly increased sEPSC rates but blocked ST-EPSCs. Lowering the RTX concentration to 150 pM modestly increased the frequency of the sEPSCs without causing failures in the evoked ST-EPSCs. The sEPSC rate increased with raising bath temperature to 36 °C. Such thermal responses were larger in 150 pM RTX, while the ST-EPSCs remained unaffected. Vanilloid sensitization of thermal responses persisted in TTX but was blocked by the TRPV1 antagonist capsazepine. Our results demonstrate that multimodal activation of TRPV1 facilitates sEPSC responses in more than the arithmetic sum of the two activators, i.e. co-activation sensitizes TRPV1 control of spontaneous glutamate release. Since action potential evoked glutamate release is unaltered, the work provides evidence for cooperativity in gating TRPV1 plus a remarkable separation of calcium mechanisms governing the independent vesicle pools responsible for spontaneous and evoked release at primary afferents in the NTS.


Assuntos
Diterpenos/farmacologia , Ácido Glutâmico/metabolismo , Núcleo Solitário/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo , Animais , Capsaicina/farmacologia , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Técnicas In Vitro , Masculino , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Fármacos do Sistema Sensorial/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Núcleo Solitário/citologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Tetrodotoxina/farmacologia
10.
PLoS One ; 10(5): e0127764, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25992717

RESUMO

Temperature is fundamentally important to all biological functions including synaptic glutamate release. Vagal afferents from the solitary tract (ST) synapse on second order neurons in the nucleus of the solitary tract, and glutamate release at this first central synapse controls autonomic reflex function. Expression of the temperature-sensitive Transient Receptor Potential Vanilloid Type 1 receptor separates ST afferents into C-fibers (TRPV1+) and A-fibers (TRPV1-). Action potential-evoked glutamate release is similar between C- and A-fiber afferents, but TRPV1 expression facilitates a second form of synaptic glutamate release in C-fibers by promoting substantially more spontaneous glutamate release. The influence of temperature on different forms of glutamate release is not well understood. Here we tested how temperature impacts the generation of evoked and spontaneous release of glutamate and its relation to TRPV1 expression. In horizontal brainstem slices of rats, activation of ST primary afferents generated synchronous evoked glutamate release (ST-eEPSCs) at constant latency whose amplitude reflects the probability of evoked glutamate release. The frequency of spontaneous EPSCs in these same neurons measured the probability of spontaneous glutamate release. We measured both forms of glutamate from each neuron during ramp changes in bath temperature of 4-5 °C. Spontaneous glutamate release from TRPV1+ closely tracked with these thermal changes indicating changes in the probability of spontaneous glutamate release. In the same neurons, temperature changed axon conduction registered as latency shifts but ST-eEPSC amplitudes were constant and independent of TRPV1 expression. These data indicate that TRPV1-operated glutamate release is independent of action potential-evoked glutamate release in the same neurons. Together, these support the hypothesis that evoked and spontaneous glutamate release originate from two pools of vesicles that are independently modulated and are distinct processes.


Assuntos
Potenciais Evocados/fisiologia , Crânio/inervação , Temperatura , Fibras Aferentes Viscerais/fisiologia , Potenciais de Ação/fisiologia , Animais , Potenciais Pós-Sinápticos Excitadores/fisiologia , Ácido Glutâmico , Masculino , Neurônios/fisiologia , Fenótipo , Ratos Sprague-Dawley , Tempo de Reação/fisiologia , Núcleo Solitário/fisiologia , Canais de Cátion TRPV/metabolismo
11.
Exp Neurol ; 244: 43-50, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22178327

RESUMO

Phytocannabinoids isolated from the cannabis plant have broad potential in medicine that has been well recognized for many centuries. It is presumed that these lipid soluble signaling molecules exert their effects in both the central and peripheral nervous system in large part through direct interaction with metabotropic cannabinoid receptors. These same receptors are also targeted by a variety of endogenous cannabinoids including 2-arachidonoyl glycerol and anandamide. Significant effort over the last decade has produced an enormous advance in our understanding of both the cellular and the synaptic physiology of endogenous lipid signaling systems. This increase in knowledge has left us better prepared to carefully evaluate the potential for both natural and synthetic cannabinoids in the treatment of a variety of neurological disorders. In the case of epilepsy, long standing interest in therapeutic approaches that target endogenous cannabinoid signaling systems are, for the most part, not well justified by available clinical data from human epileptics. Nevertheless, basic science experiments have clearly indicated a key role for endogenous cannabinoid signaling systems in moment to moment regulation of neuronal excitability. Further it has become clear that these systems can both alter and be altered by epileptiform activity in a wide range of in vitro and in vivo models of epilepsy. Collectively these observations suggest clear potential for effective therapeutic modulation of endogenous cannabinoid signaling systems in the treatment of human epilepsy, and in fact, further highlight key obstacles that would need to be addressed to reach that goal.


Assuntos
Canabinoides/uso terapêutico , Cannabis/química , Epilepsia/tratamento farmacológico , Epilepsia/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Epilepsia/patologia , Humanos
12.
Front Neurosci ; 6: 191, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23335875

RESUMO

The brainstem nucleus of the solitary tract (NTS) holds the first central neurons in major homeostatic reflex pathways. These homeostatic reflexes regulate and coordinate multiple organ systems from gastrointestinal to cardiopulmonary functions. The core of many of these pathways arise from cranial visceral afferent neurons that enter the brain as the solitary tract (ST) with more than two-thirds arising from the gastrointestinal system. About one quarter of ST afferents have myelinated axons but the majority are classed as unmyelinated C-fibers. All ST afferents release the fast neurotransmitter glutamate with remarkably similar, high-probability release characteristics. Second order NTS neurons receive surprisingly limited primary afferent information with one or two individual inputs converging on single second order NTS neurons. A- and C-fiber afferents never mix at NTS second order neurons. Many transmitters modify the basic glutamatergic excitatory postsynaptic current often by reducing glutamate release or interrupting terminal depolarization. Thus, a distinguishing feature of ST transmission is presynaptic expression of G-protein coupled receptors for peptides common to peripheral or forebrain (e.g., hypothalamus) neuron sources. Presynaptic receptors for angiotensin (AT1), vasopressin (V1a), oxytocin, opioid (MOR), ghrelin (GHSR1), and cholecystokinin differentially control glutamate release on particular subsets of neurons with most other ST afferents unaffected. Lastly, lipid-like signals are transduced by two key ST presynaptic receptors, the transient receptor potential vanilloid type 1 and the cannabinoid receptor that oppositely control glutamate release. Increasing evidence suggests that peripheral nervous signaling mechanisms are repurposed at central terminals to control excitation and are major sites of signal integration of peripheral and central inputs particularly from the hypothalamus.

13.
Brain Res ; 1318: 42-51, 2010 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-20079344

RESUMO

In the present study we used electrophysiological techniques in an in vitro preparation of the rat dentate gyrus to examine the effect of muscarinic acetylcholine receptor activation on the intrinsic excitability of hilar neurons. We found that bath application of muscarine caused a direct depolarization in approximately 80% of mossy cells tested, and also produced a clear afterdepolarization (ADP) in nearly 100% of trials. The ADP observed in hilar mossy cells is produced by the opening of a Na(+) permeant and yet largely TTX insensitive ion channel. It requires an increase in postsynaptic calcium for activation, and is blocked by flufenamic acid, an antagonist of a previously identified calcium activated non-selective cation channel (I(CAN)). Further, we demonstrate that induction of an ADP in current clamp causes release of cannabinoids, and subsequent depression of GABAergic transmission that is comparable to that produced in the same cells by a more conventional 5s depolarization in voltage clamp. By contrast, other types of hilar neurons were less strongly depolarized by bath application of muscarinic agonists, and uniformly lacked a similar muscarinic ADP. Overall, the data presented here extend our understanding of the specific mechanisms through which muscarinic agonists are likely to modulate neuronal excitability in the hilar network, and further reveal a mechanism that could plausibly promote endocannabinoid mediated signaling in vivo.


Assuntos
Potenciais da Membrana/fisiologia , Fibras Musgosas Hipocampais/fisiologia , Receptores Muscarínicos/metabolismo , Animais , Cálcio/metabolismo , Moduladores de Receptores de Canabinoides/metabolismo , Fármacos do Sistema Nervoso Central/farmacologia , Giro Denteado/efeitos dos fármacos , Giro Denteado/fisiologia , Ácido Flufenâmico/farmacologia , Técnicas In Vitro , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Fibras Musgosas Hipocampais/efeitos dos fármacos , Muscarina/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Canais de Sódio/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Ácido gama-Aminobutírico/metabolismo
14.
J Neurophysiol ; 96(5): 2501-12, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16807350

RESUMO

Hilar mossy cells represent a unique population of local circuit neurons in the hippocampus and dentate gyrus. Here we use electrophysiological techniques in acute preparations of hippocampal slices to demonstrate that depolarization of a single hilar mossy cell can produce robust inhibition of local GABAergic afferents. This depolarization-induced suppression of inhibition (DSI) can be observed as a transient reduction in frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) or as a transient reduction in amplitude of evoked IPSCs (eIPSCs). We find that DSI of eIPSCs as observed in hilar mossy cells is enhanced by activation of muscarinic acetylcholine receptors, blocked by chelation of postsynaptic calcium, and critically dependent on retrograde activation of presynaptic cannabinoid type 1 (CB1) receptors. We further report that activation of CB1 receptors on GABAergic afferents to hilar mossy cells (by either endogenous or exogenous agonists) preferentially inhibits calcium-dependent exocytosis and that endocannabinoid-dependent retrograde signaling in this system is subject to tight spatial constraints.


Assuntos
Moduladores de Receptores de Canabinoides/fisiologia , Giro Denteado/fisiologia , Endocanabinoides , Fibras Musgosas Hipocampais/fisiologia , Animais , Benzoxazinas , Cálcio/fisiologia , Interpretação Estatística de Dados , Giro Denteado/citologia , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Masculino , Potenciais da Membrana/fisiologia , Morfolinas/farmacologia , Naftalenos/farmacologia , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/efeitos dos fármacos , Receptor CB1 de Canabinoide/metabolismo , Receptores Pré-Sinápticos/efeitos dos fármacos , Receptores Pré-Sinápticos/metabolismo , Transmissão Sináptica/fisiologia , Ácido gama-Aminobutírico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA