Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cereb Cortex ; 29(9): 3778-3795, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30295710

RESUMO

Epilepsy is a multifactorial disorder associated with neuronal hyperexcitability that affects more than 1% of the human population. It has long been known that adenosine can reduce seizure generation in animal models of epilepsies. However, in addition to various side effects, the instability of adenosine has precluded its use as an anticonvulsant treatment. Here we report that a stable analogue of diadenosine-tetraphosphate: AppCH2ppA effectively suppresses spontaneous epileptiform activity in vitro and in vivo in a Tuberous Sclerosis Complex (TSC) mouse model (Tsc1+/-), and in postsurgery cortical samples from TSC human patients. These effects are mediated by enhanced adenosine signaling in the cortex post local neuronal adenosine release. The released adenosine induces A1 receptor-dependent activation of potassium channels thereby reducing neuronal excitability, temporal summation, and hypersynchronicity. AppCH2ppA does not cause any disturbances of the main vital autonomous functions of Tsc1+/- mice in vivo. Therefore, we propose this compound to be a potent new candidate for adenosine-related treatment strategies to suppress intractable epilepsies.


Assuntos
Adenosina/fisiologia , Anticonvulsivantes/administração & dosagem , Fosfatos de Dinucleosídeos/administração & dosagem , Neocórtex/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Convulsões/fisiopatologia , Animais , Feminino , Humanos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Neocórtex/fisiopatologia , Neurônios/fisiologia , Canais de Potássio/fisiologia , Receptor A1 de Adenosina/fisiologia , Convulsões/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Proteína 1 do Complexo Esclerose Tuberosa/genética
2.
Mol Pain ; 122016.
Artigo em Inglês | MEDLINE | ID: mdl-27030723

RESUMO

BACKGROUND: A growing body of evidence suggests that ATP-gated P2X3 receptors (P2X3Rs) are implicated in chronic pain. We address the possibility that stable, synthetic analogs of diadenosine tetraphosphate (Ap4A) might induce antinociceptive effects by inhibiting P2X3Rs in peripheral sensory neurons. RESULTS: The effects of two stable, synthetic Ap4A analogs (AppNHppA and AppCH2ppA) are studied firstly in vitro on HEK293 cells expressing recombinant rat P2XRs (P2X2Rs, P2X3Rs, P2X4Rs, and P2X7Rs) and then using native rat brain cells (cultured trigeminal, nodose, or dorsal root ganglion neurons). Thereafter, the action of these stable, synthetic Ap4A analogs on inflammatory pain and thermal hyperalgesia is studied through the measurement of antinociceptive effects in formalin and Hargreaves plantar tests in rats in vivo. In vitro inhibition of rat P2X3Rs (not P2X2Rs, P2X4Rs nor P2X7Rs) is shown to take place mediated by high-affinity desensitization (at low concentrations; IC50 values 100-250 nM) giving way to only weak partial agonism at much higher concentrations (EC50 values ≥ 10 µM). Similar inhibitory activity is observed with human recombinant P2X3Rs. The inhibitory effects of AppNHppA on nodose, dorsal root, and trigeminal neuron whole cell currents suggest that stable, synthetic Ap4A analogs inhibit homomeric P2X3Rs in preference to heteromeric P2X2/3Rs. Both Ap4A analogs mediate clear inhibition of pain responses in both in vivo inflammation models. CONCLUSIONS: Stable, synthetic Ap4A analogs (AppNHppA and AppCH2ppA) being weak partial agonist provoke potent high-affinity desensitization-mediated inhibition of homomeric P2X3Rs at low concentrations. Therefore, both analogs demonstrate clear potential as potent analgesic agents for use in the management of chronic pain associated with heightened P2X3R activation.


Assuntos
Fosfatos de Dinucleosídeos/uso terapêutico , Inflamação/complicações , Inflamação/tratamento farmacológico , Dor/complicações , Dor/tratamento farmacológico , Antagonistas do Receptor Purinérgico P2X/uso terapêutico , Receptores Purinérgicos P2X3/metabolismo , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Fosfatos de Dinucleosídeos/farmacologia , Células HEK293 , Humanos , Hiperalgesia/complicações , Hiperalgesia/tratamento farmacológico , Injeções Subcutâneas , Masculino , Multimerização Proteica/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Antagonistas do Receptor Purinérgico P2X/farmacologia , Ratos Wistar , Proteínas Recombinantes/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo
3.
Cereb Cortex ; 25(9): 2440-55, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24646614

RESUMO

Cannabinoids are known to regulate inhibitory synaptic transmission via activation of presynaptic G protein-coupled cannabinoid CB1 receptors (CB1Rs). Additionally, recent studies suggest that cannabinoids can also directly interact with recombinant GABAA receptors (GABAARs), potentiating currents activated by micromolar concentrations of γ-aminobutyric acid (GABA). However, the impact of this direct interaction on GABAergic inhibition in central nervous system is unknown. Here we report that currents mediated by recombinant GABAARs activated by high (synaptic) concentrations of GABA as well as GABAergic inhibitory postsynaptic currents (IPSCs) at neocortical fast spiking (FS) interneuron to pyramidal neuron synapses are suppressed by exogenous and endogenous cannabinoids in a CB1R-independent manner. This IPSC suppression may account for disruption of inhibitory control of pyramidal neurons by FS interneurons. At FS interneuron to pyramidal neuron synapses, endocannabinoids induce synaptic low-pass filtering of GABAAR-mediated currents evoked by high-frequency stimulation. The CB1R-independent suppression of inhibition is synapse specific. It does not occur in CB1R containing hippocampal cholecystokinin-positive interneuron to pyramidal neuron synapses. Furthermore, in contrast to synaptic receptors, the activity of extrasynaptic GABAARs in neocortical pyramidal neurons is enhanced by cannabinoids in a CB1R-independent manner. Thus, cannabinoids directly interact differentially with synaptic and extrasynaptic GABAARs, providing a potent novel context-dependent mechanism for regulation of inhibition.


Assuntos
Canabinoides/metabolismo , Potenciais Pós-Sinápticos Inibidores/fisiologia , Inibição Neural/fisiologia , Receptores de GABA/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Animais , Animais Recém-Nascidos , Canabinoides/farmacologia , GABAérgicos/farmacologia , Hipocampo/citologia , Humanos , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibição Neural/efeitos dos fármacos , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptor CB1 de Canabinoide/genética , Receptor CB1 de Canabinoide/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética , Transfecção
4.
Brain ; 136(Pt 8): 2457-73, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23831613

RESUMO

Altered development of the human cerebral cortex can cause severe malformations with often intractable focal epileptic seizures and may participate in common pathologies, notably epilepsy. This raises important conceptual and therapeutic issues. Two missense mutations in the sushi repeat-containing protein SRPX2 had been previously identified in epileptic disorders with or without structural developmental alteration of the speech cortex. In the present study, we aimed to decipher the precise developmental role of SRPX2, to have a better knowledge on the consequences of its mutations, and to start addressing therapeutic issues through the design of an appropriate animal model. Using an in utero Srpx2 silencing approach, we show that SRPX2 influences neuronal migration in the developing rat cerebral cortex. Wild-type, but not the mutant human SRPX2 proteins, rescued the neuronal migration phenotype caused by Srpx2 silencing in utero, and increased alpha-tubulin acetylation. Following in utero Srpx2 silencing, spontaneous epileptiform activity was recorded post-natally. The neuronal migration defects and the post-natal epileptic consequences were prevented early in embryos by maternal administration of tubulin deacetylase inhibitor tubacin. Hence epileptiform manifestations of developmental origin could be prevented in utero, using a transient and drug-based therapeutic protocol.


Assuntos
Anilidas/farmacologia , Movimento Celular/genética , Córtex Cerebral/metabolismo , Epilepsia/genética , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Proteínas de Membrana/genética , Neurônios/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Epilepsia/metabolismo , Inativação Gênica , Humanos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Ratos , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
5.
J Neurosci ; 32(50): 18047-53, 2012 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-23238720

RESUMO

Cellular electrophysiological signatures of Parkinson's disease described in the pharmacological 6-hydroxydopamine (6-OHDA) animal models of Parkinson's disease include spontaneous repetitive giant GABAergic currents in a subpopulation of striatal medium spiny neurons (MSNs), and spontaneous rhythmic bursts of spikes generated by subthalamic nucleus (STN) neurons. We investigated whether similar signatures are present in Pink1(-/-) mice, a genetic rodent model of the PARK6 variant of Parkinson's disease. Although 9- to 24-month-old Pink1(-/-) mice show reduced striatal dopamine content and release, and impaired spontaneous locomotion, the relevance of this model to Parkinson's disease has been questioned because mesencephalic dopaminergic neurons do not degenerate during the mouse lifespan. We show that 75% of the MSNs of 5- to 7-month-old Pink1(-/-) mice exhibit giant GABAergic currents, occurring either singly or in bursts (at 40 Hz), rather than the low-frequency (2 Hz), low-amplitude, tonic GABAergic drive common to wild-type MSNs of the same age. STN neurons from 5- to 7-month-old Pink1(-/-) mice spontaneously generated bursts of spikes instead of the control tonic drive. Chronic kainic acid lesion of the STN or chronic levodopa treatment reliably suppressed the giant GABAergic currents of MSNs after 1 month and replaced them with the control tonic activity. The similarity between the in vitro resting states of Pink1 MSNs and those of fully dopamine (DA)-depleted MSNs of 6-OHDA-treated mice, together with the beneficial effect of levodopa treatment, strongly suggest that dysfunction of mesencephalic dopaminergic neurons in Pink1(-/-) mice is more severe than expected. The beneficial effect of the STN lesion also suggests that pathological STN activity strongly influences striatal networks in Pink1(-/-) mice.


Assuntos
Levodopa/farmacologia , Neurônios/efeitos dos fármacos , Transtornos Parkinsonianos/fisiopatologia , Proteínas Quinases/deficiência , Núcleo Subtalâmico/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo , Animais , Antiparkinsonianos/farmacologia , Corpo Estriado/patologia , Corpo Estriado/fisiopatologia , Condutividade Elétrica , Agonistas de Aminoácidos Excitatórios/toxicidade , Feminino , Imuno-Histoquímica , Ácido Caínico/toxicidade , Masculino , Camundongos , Camundongos Knockout , Neurônios/fisiologia , Transtornos Parkinsonianos/patologia , Técnicas de Patch-Clamp , Proteínas Quinases/genética , Núcleo Subtalâmico/lesões , Núcleo Subtalâmico/patologia , Núcleo Subtalâmico/fisiopatologia
6.
J Pharmacol Exp Ther ; 331(2): 618-26, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19684252

RESUMO

N-Methyl-d-aspartate (NMDA) receptor antagonists that are highly selective for specific NMDA receptor 2 (NR2) subunits have several potential therapeutic applications; however, to date, only NR2B-selective antagonists have been described. Whereas most glutamate binding site antagonists display a common pattern of NR2 selectivity, NR2A > NR2B > NR2C > NR2D (high to low affinity), (2S*,3R*)-1-(phenanthrene-2-carbonyl)piperazine-2,3-dicarboxylic acid (PPDA) has a low selectivity for NR2C- and NR2D-containing NMDA receptors. A series of PPDA derivatives were synthesized and then tested at recombinant NMDA receptors expressed in Xenopus laevis oocytes. In addition, the optical isomers of PPDA were resolved; the (-) isomer displayed a 50- to 80-fold greater potency than the (+) isomer. Replacement of the phenanthrene moiety of PPDA with naphthalene or anthracene did not improve selectivity. However, phenylazobenzoyl (UBP125) or phenylethynylbenzoyl (UBP128) substitution significantly improved selectivity for NR2B-, NR2C-, and NR2D-containing receptors over NR2A-containing NMDA receptors. Phenanthrene attachment at the 3 position [(2R*,3S*)-1-(phenanthrene-3-carbonyl)piperazine-2,3-dicarboxylic acid (UBP141); (2R*,3S*)-1-(9-bromophenanthrene-3-carbonyl)piperazine-2,3-dicarboxylic acid (UBP145); (2R*,3S*)-1-(9-chlorophenanthrene-3-carbonyl)piperazine-2,3-dicarboxylic acid (UBP160); and (2R*,3S*)-1-(9-iodophenanthrene-3-carbonyl)piperazine-2,3-dicarboxylic acid (UBP161)] displayed improved NR2D selectivity. UBP141 and its 9-brominated homolog (UBP145) both display a 7- to 10- fold selectivity for NR2D-containing receptors over NR2B- or NR2A-containing receptors. Schild analysis indicates that these two compounds are competitive glutamate binding site antagonists. Consistent with a physiological role for NR2D-containing receptors in the hippocampus, UBP141 (5 muM) displayed greater selectivity than PPDA for inhibiting the slow-decaying component of the NMDA receptor-mediated CA3-CA1 synaptic response in rat hippocampal slices. UBP125, UBP128, UBP141, and UBP145 may be useful tools for determining the function of NMDA receptor subtypes.


Assuntos
Ácidos Dicarboxílicos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hipocampo/metabolismo , Piperazinas/farmacologia , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Animais , DNA Complementar/biossíntese , DNA Complementar/genética , Desenho de Fármacos , Eletrofisiologia , Hipocampo/efeitos dos fármacos , Técnicas In Vitro , Ácido Caínico/metabolismo , Oócitos , Ratos , Ratos Wistar , Receptores de AMPA/efeitos dos fármacos , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/química , Relação Estrutura-Atividade , Especificidade por Substrato , Xenopus laevis
7.
Front Cell Neurosci ; 9: 195, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26041996

RESUMO

GABAergic inhibition, which is instrumental in the generation of hippocampal gamma oscillations, undergoes significant changes during development. However, the development of hippocampal gamma oscillations remains largely unknown. Here, we explored the developmental features of kainate-induced oscillations (KA-Os) in CA3 region of rat hippocampal slices. Up to postnatal day P5, the bath application of kainate failed to evoke any detectable oscillations. KA-Os emerged by the end of the first postnatal week; these were initially weak, slow (20-25 Hz, beta range) and were poorly synchronized with CA3 units and synaptic currents. Local field potential (LFP) power, synchronization of units and frequency of KA-Os increased during the second postnatal week to attain gamma (30-40 Hz) frequency by P15-21. Both beta and gamma KA-Os are characterized by alternating sinks and sources in the pyramidal cell layer, likely generated by summation of the action potential-associated currents and GABAergic synaptic currents, respectively. Blockade of GABA(A) receptors with gabazine completely suppressed KA-Os at all ages indicating that GABAergic mechanisms are instrumental in their generation. Bumetanide, a NKCC1 chloride co-transporter antagonist which renders GABAergic responses inhibitory in the immature hippocampal neurons, failed to induce KA-Os at P2-4 indicating that the absence of KA-Os in neonates is not due to depolarizing actions of GABA. The linear developmental profile, electrographic features and pharmacological properties indicate that CA3 hippocampal beta and gamma KA-Os are fundamentally similar in their generative mechanisms and their delayed onset and developmental changes likely reflect the development of perisomatic GABAergic inhibition.

8.
Science ; 346(6206): 176, 2014 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-25301611

RESUMO

Bambini-Junior et al. questioned whether our treatment in two rodent models of autism has a long-lasting effect into adulthood. In response, we show that bumetanide treatment around delivery attenuates autistic behavioral features in adult offspring. Therefore, the polarity of γ-aminobutyric acid (GABA) actions during delivery exerts long-lasting priming actions after birth.


Assuntos
Transtorno Autístico/induzido quimicamente , Transtorno Autístico/genética , Citoproteção , Ocitocina/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Feminino , Gravidez
9.
Science ; 343(6171): 675-9, 2014 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-24503856

RESUMO

We report that the oxytocin-mediated neuroprotective γ-aminobutyric acid (GABA) excitatory-inhibitory shift during delivery is abolished in the valproate and fragile X rodent models of autism. During delivery and subsequently, hippocampal neurons in these models have elevated intracellular chloride levels, increased excitatory GABA, enhanced glutamatergic activity, and elevated gamma oscillations. Maternal pretreatment with bumetanide restored in offspring control electrophysiological and behavioral phenotypes. Conversely, blocking oxytocin signaling in naïve mothers produced offspring having electrophysiological and behavioral autistic-like features. Our results suggest a chronic deficient chloride regulation in these rodent models of autism and stress the importance of oxytocin-mediated GABAergic inhibition during the delivery process. Our data validate the amelioration observed with bumetanide and oxytocin and point to common pathways in a drug-induced and a genetic rodent model of autism.


Assuntos
Transtorno Autístico/induzido quimicamente , Transtorno Autístico/genética , Citoproteção , Ocitocina/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Transtorno Autístico/metabolismo , Comportamento Animal , Bumetanida/administração & dosagem , Cloretos/metabolismo , Modelos Animais de Doenças , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Troca Materno-Fetal , Camundongos , Parto , Gravidez , Ratos , Ácido Valproico/farmacologia
10.
Nat Genet ; 45(9): 1061-6, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23933820

RESUMO

Epileptic encephalopathies are severe brain disorders with the epileptic component contributing to the worsening of cognitive and behavioral manifestations. Acquired epileptic aphasia (Landau-Kleffner syndrome, LKS) and continuous spike and waves during slow-wave sleep syndrome (CSWSS) represent rare and closely related childhood focal epileptic encephalopathies of unknown etiology. They show electroclinical overlap with rolandic epilepsy (the most frequent childhood focal epilepsy) and can be viewed as different clinical expressions of a single pathological entity situated at the crossroads of epileptic, speech, language, cognitive and behavioral disorders. Here we demonstrate that about 20% of cases of LKS, CSWSS and electroclinically atypical rolandic epilepsy often associated with speech impairment can have a genetic origin sustained by de novo or inherited mutations in the GRIN2A gene (encoding the N-methyl-D-aspartate (NMDA) glutamate receptor α2 subunit, GluN2A). The identification of GRIN2A as a major gene for these epileptic encephalopathies provides crucial insights into the underlying pathophysiology.


Assuntos
Epilepsias Parciais/genética , Síndrome de Landau-Kleffner/genética , Mutação , Receptores de N-Metil-D-Aspartato/genética , Substituição de Aminoácidos , Linhagem Celular , Eletroencefalografia , Feminino , Expressão Gênica , Genótipo , Humanos , Masculino , Linhagem , Fenótipo
11.
Cell Calcium ; 46(3): 154-62, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19682741

RESUMO

The neuromodulatory effects of cannabinoids in the central nervous system have mainly been associated with G-protein coupled cannabinoid receptor (CB1R) mediated inhibition of voltage-gated calcium channels (VGCCs). Numerous studies show, however, that cannabinoids can also modulate VGCCs independent of CB1R activation. Nevertheless, despite the fact that endocannabinoids have a nearly equal efficacy for direct and CB1R-mediated effects on VGCC, the role of the direct cannabinoid-VGCC interaction has been largely underestimated. In this review, we summarize recent studies on the modulation of different types of VGCCs by cannabinoids, highlight the evidence for and implications of the CB1R-independent modulation, and put forward the concept, that direct interaction of cannabinoids and VGCCs is as important in regulation of VGCCs function as the CB1R-mediated effects.


Assuntos
Canais de Cálcio/metabolismo , Moduladores de Receptores de Canabinoides/farmacologia , Sistema Nervoso Central/fisiologia , Animais , Ácidos Araquidônicos/farmacologia , Sinalização do Cálcio , Endocanabinoides , Alcamidas Poli-Insaturadas/farmacologia , Isoformas de Proteínas/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Transdução de Sinais
12.
J Neurophysiol ; 96(3): 1267-77, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16738209

RESUMO

Endocannabinoids released by postsynaptic cells inhibit neurotransmitter release in many central synapses by activating presynaptic cannabinoid CB1 receptors. In particular, in the cerebellum, endocannabinoids inhibit synaptic transmission at granule cell to Purkinje cell synapses by modulating presynaptic calcium influx via N-, P/Q-, and R-type calcium channels. Using whole cell patch-clamp techniques, we show that in addition to this presynaptic action, both synthetic and endogenous cannabinoids inhibit P-type calcium currents in isolated rat Purkinje neurons independent of CB1 receptor activation. The IC50 for the anandamide (AEA)-induced inhibition of P-current peak amplitude was 1.04 +/- 0.04 microM. In addition, we demonstrate that all the tested cannabinoids in a physiologically relevant range of concentrations strongly accelerate inactivation of P currents. The effects of AEA cannot be attributed to the metabolism of AEA because a nonhydrolyzing analogue of AEA, methanandamide inhibited P-type currents with a similar efficacy. All effects of cannabinoids on P-type Ca2+ currents were insensitive to antagonists of CB1 cannabinoid or vanilloid TRPV1 receptors. In cerebellar slices, WIN 55,212-2 significantly affected spontaneous firing of Purkinje neurons in the presence of CB1 receptor antagonist, in a manner similar to that of a specific P-type channel antagonist, indicating a possible functional implication of the direct effects of cannabinoids on P current. Taken together these findings demonstrate a functionally important direct action of cannabinoids on P-type calcium currents.


Assuntos
Canais de Cálcio Tipo P/fisiologia , Canabinoides/farmacologia , Células de Purkinje/fisiologia , Animais , Ácidos Araquidônicos/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo P/efeitos dos fármacos , Canais de Cálcio Tipo R/efeitos dos fármacos , Canais de Cálcio Tipo R/fisiologia , Cerebelo/fisiologia , Endocanabinoides , Técnicas In Vitro , Cinética , Técnicas de Patch-Clamp , Alcamidas Poli-Insaturadas , Células de Purkinje/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Canais de Cátion TRPV/efeitos dos fármacos , Canais de Cátion TRPV/fisiologia
13.
J Pharmacol Exp Ther ; 318(2): 579-88, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16709679

RESUMO

Previously, we have described the modulatory effect of diadenosine polyphosphates Ap4A and Ap5A on synaptic transmission in the rat hippocampal slices mediated by presynaptic receptors (Klishin et al., 1994). In contrast, we now describe how nonhydrolyzable Ap4A analog diadenosine-5',5'''-P1,P4-[beta,beta'-methylene]tetraphosphate (AppCH2ppA) at low micromolar concentrations exerts strong nondesensitizing inhibition of orthodromically evoked field potentials (OFPs) without affecting the amplitude of excitatory postsynaptic currents and antidromically evoked field potentials, as recorded in hippocampal CA1 zone. The effects of AppCH2ppA on OFPs are eliminated by a P2 receptor antagonist pyridoxal-phosphate-6-azophenyl-2',4'-disulfonic acid (PPADS) but not mimicked by purinoceptor agonists alpha,beta-methylene-ATP and adenosine 5'-O-(3-thio)-triphosphate, indicating that a P2-like receptor is involved but not one belonging to the conventional P2X/P2Y receptor classes. Diadenosine polyphosphate receptor (P4) antagonist Ip4I (diinosine tetraphosphate) was unable to modulate AppCH2ppA effects. Thus, the PPADS-sensitive P2-like receptor for AppCH2ppA seems to control selectively dendritic excitation of the CA1 neurons. The specific nitric oxide (NO)-scavenger 2-phenyl-4,4,5,5-tetramethyl-imidazoline-1-oxyl-3-oxide is shown to significantly attenuate AppCH2ppA-mediated inhibitory effects, indicating that NO is involved in the cascade of events initiated by AppCH2ppA. Further downstream mediation by adenosine A1 receptors is also demonstrated. Hence, AppCH2ppA-mediated effects involve PPADS-sensitive P2-like receptor activation leading to the production of NO that stimulates intracellular synthesis of adenosine, causing in turn postsynaptic A1 receptor activation and subsequent postsynaptic CA1 dendritic inhibition. Such spatially selective postsynaptic dendritic inhibition may influence dendritic electrogenesis in pyramidal neurons and consequently mediate control of neuronal network activity.


Assuntos
Fosfatos de Dinucleosídeos/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Óxido Nítrico/fisiologia , Adenosina/farmacologia , Animais , Óxidos N-Cíclicos/farmacologia , Dendritos/efeitos dos fármacos , Estimulação Elétrica , Eletrofisiologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hidrólise , Imidazóis/farmacologia , Nitroglicerina/farmacologia , Técnicas de Patch-Clamp , Agonistas Purinérgicos , Antagonistas Purinérgicos , Células Piramidais/efeitos dos fármacos , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacologia , Ratos , Ratos Wistar , Transmissão Sináptica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA