Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Eur J Pharmacol ; 969: 176396, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38325793

RESUMEN

Olanzapine is an antipsychotic drug applied in psychiatry to treat psychoses, especially schizophrenia and schizoaffective disorders with similar or better improvement than haloperidol and risperidone in the treatment of depressive and negative symptoms. The effect of olanzapine on neural synchrony remains to be explored. We investigated the effects of olanzapine on gamma oscillations in the CA3 region of the hippocampus and frontal association cortex. Olanzapine reduced carbachol (CCh)-induced gamma oscillation power in CA3 slice and gamma oscillation power in the frontal association cortex in vivo. The power of theta oscillations was increased in the presence of olanzapine. The phase amplitude coupling of theta and gamma wave was strengthened by the administration of olanzapine in the frontal association cortex in vivo. Taken together, these results show that olanzapine modulates local field potential and the neuronal activity.


Asunto(s)
Antipsicóticos , Olanzapina/farmacología , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Hipocampo , Haloperidol/farmacología , Corteza Prefrontal
2.
Neuroscience ; 502: 107-116, 2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-36038038

RESUMEN

5-HT2A receptors (5-HT2ARs) are widely expressed in the central nervous system, including in the ventrolateral orbital cortex (VLO). The VLO is an important cortical component for pain processing. Brain 5-HT2ARs are implicated in both pro- and anti- nociceptive functions. However, the roles of 5-HT2ARs in the VLO in trigeminal neuralgia and neuronal synaptic function remain to be understood. We used chronic constriction injury of infraorbital nerve (IoN-CCI) model and shRNA mediated gene knockdown in mice to investigate the role of 5-HT2ARs in the VLO in trigeminal neuralgia. We found that knockdown of 5-HT2ARs in the VLO aggravated spontaneous pain and mechanical allodynia in mice after IoN-CCI. At the synaptic level, decreasing 5-HT2AR expression by shRNA or inhibition of 5-HT2AR activity by its antagonist ketanserin decreased the frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) of the neurons in the VLO, whereas 5-HT2AR partial agonist 2,5-Dimethoxy-4-iodoamphetamine (DOI) enhanced sEPSCs of the neurons in the VLO. In summary, 5-HT2ARs in the VLO modulate the trigeminal pain by regulating neuronal glutamatergic activity.


Asunto(s)
Neuralgia del Trigémino , Ratas , Animales , Ratones , Neuralgia del Trigémino/metabolismo , Receptor de Serotonina 5-HT2A/metabolismo , Ratas Sprague-Dawley , Ketanserina/metabolismo , Serotonina/metabolismo , ARN Interferente Pequeño/metabolismo , Dolor/metabolismo , Corteza Prefrontal/metabolismo
3.
Front Pharmacol ; 13: 1084108, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36588734

RESUMEN

The reduced antidepressant and antihyperalgesic effects of selective serotonin reuptake inhibitors (SSRIs) such as fluoxetine during maintenance treatment has been reported, but little is known about the molecular mechanism of this phenomenon. In three comorbid pain and depression animal models (genetic predisposition, chronic social stress, arthritis), we showed that the fluoxetine's antidepressant and antihyperalgesic effects were reduced during the maintenance treatment. Fluoxetine exposure induced upregulation of the 5-hydroxytryptamine 1A (5-HT1A) auto-receptor and indoleamine 2,3 dioxygenase 1 (IDO1, a rate-limiting enzyme of tryptophan metabolism) in the brainstem dorsal raphe nucleus (DRN), which shifted the tryptophan metabolism away from the 5-HT biosynthesis. Mechanistically, IDO1 upregulation was downstream to fluoxetine-induced 5-HT1A receptor expression because 1) antagonism of the 5-HT1A receptor with WAY100635 or 5-HT1A receptor knockout blocked the IDO1 upregulation, and 2) inhibition of IDO1 activity did not block the 5-HT1A receptor upregulation following fluoxetine exposure. Importantly, inhibition of either the 5-HT1A receptor or IDO1 activity sustained the fluoxetine's antidepressant and antihyperalgesic effects, indicating that 5-HT1A-mediated IDO1 upregulation in the brainstem DRN contributed to the reduced antidepressant and antihyperalgesic effects of fluoxetine. These results suggest a new strategy to improving the therapeutic efficacy of SSRI during maintenance treatment.

4.
Int J Mol Sci ; 23(1)2021 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-35008465

RESUMEN

Benzodiazepines (BZDs) produce versatile pharmacological actions through positive modulation of GABAA receptors (GABAARs). A previous study has demonstrated that high concentrations of diazepam potentiate GABA currents on the α1ß2γ2 and α1ß2 GABAARs in a flumazenil-insensitive manner. In this study, the high-concentration effects of BZDs and their sensitivity to flumazenil were determined on synaptic (α1ß2γ2, α2ß2γ2, α5ß2γ2) and extra-synaptic (α4ß2δ) GABAARs using the voltage-clamp electrophysiology technique. The in vivo evaluation of flumazenil-insensitive BZD effects was conducted in mice via the loss of righting reflex (LORR) test. Diazepam induced biphasic potentiation on the α1ß2γ2, α2ß2γ2 and α5ß2γ2 GABAARs, but did not affect the α4ß2δ receptor. In contrast to the nanomolar component of potentiation, the second potentiation elicited by micromolar diazepam was insensitive to flumazenil. Midazolam, clonazepam, and lorazepam at 200 µM exhibited similar flumazenil-insensitive effects on the α1ß2γ2, α2ß2γ2 and α5ß2γ2 receptors, whereas the potentiation induced by 200 µM zolpidem or triazolam was abolished by flumazenil. Both the GABAAR antagonist pentylenetetrazol and Fa173, a proposed transmembrane site antagonist, abolished the potentiation induced by 200 µM diazepam. Consistent with the in vitro results, flumazenil antagonized the zolpidem-induced LORR, but not that induced by diazepam or midazolam. Pentylenetetrazol and Fa173 antagonized the diazepam-induced LORR. These findings support the existence of non-classical BZD binding sites on certain GABAAR subtypes and indicate that the flumazenil-insensitive effects depend on the chemical structures of BZD ligands.


Asunto(s)
Benzodiazepinas/farmacología , Flumazenil/farmacología , Receptores de GABA-A/metabolismo , Animales , Animales no Consanguíneos , Clonazepam/farmacología , Diazepam/farmacología , Femenino , Antagonistas del GABA/farmacología , Masculino , Ratones , Midazolam/farmacología , Xenopus laevis/metabolismo , Ácido gamma-Aminobutírico/metabolismo
5.
Front Cell Neurosci ; 14: 205, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32733209

RESUMEN

Although acid-sensing ion channels (ASICs) are widely expressed in the central nervous system, their distribution and roles in axonal growth cones remain unclear. In this study, we examined ASIC localization and function in the axonal growth cones of cultured immature hippocampal neurons. Our immunocytochemical data showed that native and overexpressed ASIC1a and ASIC2a are both localized in growth cones of cultured young hippocampal neurons. Calcium imaging and electrophysiological assay results were utilized to validate their function. The calcium imaging test results indicated that the ASICs (primarily ASIC1a) present in growth cones mediate calcium influx despite the addition of voltage-gated Ca2+ channels antagonists and the depletion of intracellular calcium stores. The electrophysiological tests results suggested that a rapid decrease in extracellular pH at the growth cones of voltage-clamped neurons elicits inward currents that were blocked by bath application of the ASIC antagonist amiloride, showing that the ASICs expressed at growth cones are functional. The subsequent immuno-colocalization test results demonstrated that ASIC1a and ASIC2a are both colocalized with Neurofilament-H and Bassoon in mature hippocampal neurons. This finding demonstrated that after reaching maturity, ASIC1a and ASIC2a are both distributed in axons and the presynaptic membrane. Our data reveal the distribution of functional ASICs in growth cones of immature hippocampal neurons and the presence of ASICs in the axons and presynaptic membrane of mature hippocampal neurons, indicating a possible role for ASICs in axonal guidance, synapse formation and neurotransmitter release.

6.
Biochem Biophys Res Commun ; 527(1): 98-103, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32446398

RESUMEN

Effective therapies for Methamphetamine (METH) induced stereotyped behavior are still being explored. It is unclear whether Neuropeptide S (NPS) is involved in the mechanism of METH-induced stereotyped behavior. In the contemporary behavioral study, pretreatment with NPS reduces stereotyped circling significantly, but didn't have any impact on the total incidence of stereotypy and stereotyped sniffing and biting induced by METH (10 mg/kg). When METH (10 mg/kg) was administered to rats, the level of NPS in the cerebrospinal fluid was not affected, but pretreatment with NPS reversed METH-induced glutamate release in the hippocampus and striatum. The findings suggest that NPS receptor system is likely to involve in the METH-overdose-induced behaviors.


Asunto(s)
Estimulantes del Sistema Nervioso Central/farmacología , Metanfetamina/farmacología , Neuropéptidos/farmacología , Conducta Estereotipada/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Ácido Glutámico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley
7.
Brain Res ; 1717: 204-213, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-30940440

RESUMEN

Chronic neuropathic pain has demonstrated that coexisting psychiatric disorders are associated with disability and poorer treatment outcomes. Hyperpolarization-activated cyclic nucleotide-gated (HCN, Ih) channels play a major role in pain via hyperexcitability and facilitation of ectopic firing in neurons. Neuronal hyperexcitability contributes to pain maintenance and anxiety/depression. GABA-mediated inhibitory postsynaptic neurotransmission in the brain is impaired in the pathophysiology of chronic neuropathic pain with comorbidity mood disorders. Currently, interaction of HCN channels and GABAergic synaptic transmission inhibition in neuropathic pain and the associated comorbidity anxiety/depression mechanism remains relatively unknown. To address this, the HCN channel inhibitor, ZD7288, was administrated to Wistar Kyoto (WKY) rats after spared nerve injury (SNI). Our findings show that intracerebroventricular injection of ZD7288 concurrently attenuates co-existing nociceptive and depression-like behaviors, and increases glutamicacid decarboxylase (GAD67/65) expression and GABA levels in the hippocampus and thalamus with High-performance liquid chromatography technique. It suggests that inhibition of HCN channels is likely to decrease the hyperexcitability of neurons in rat SNI and improve the level of GABA. Further, HCN channel may offer a new strategy to alleviate both neuropathic pain and comorbidity for depression.


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Neuralgia/metabolismo , Pirimidinas/farmacología , Animales , Encéfalo/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Depresión/tratamiento farmacológico , Depresión/metabolismo , Depresión/fisiopatología , Trastorno Depresivo/metabolismo , Trastorno Depresivo/fisiopatología , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Glutamato Descarboxilasa/metabolismo , Hipocampo/metabolismo , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Masculino , Neuralgia/fisiopatología , Neuronas/metabolismo , Ratas , Ratas Endogámicas WKY , Tálamo/metabolismo
8.
Pain ; 159(8): 1518-1528, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29613911

RESUMEN

Clinical evidence indicates that cognitive impairment is a common comorbid condition of chronic pain. However, the cellular basis for chronic pain-mediated cognitive impairment remains unclear. We report here that rats exhibited memory deficits after spared nerve injury (SNI). We found that levels of stable microtubule (MT) were increased in the hippocampus of the rats with memory deficits. This increase in stable MT is marked by α-tubulin hyperacetylation. Paclitaxel, a pharmacological MT stabilizer, increased the level of stable MT in the hippocampus and induced learning and memory deficits in normal rats. Furthermore, paclitaxel reduced long-term potentiation in hippocampal slices and increased stable MT (evidenced by α-tubulin hyperacetylation) levels in hippocampal neuronal cells. Intracerebroventricular infusion of nocodazole, an MT destabilizer, ameliorated memory deficits in rats with SNI-induced nociceptive behavior. Expression of HDAC6, an α-tubulin deacetylase, was reduced in the hippocampus in rats with cognitive impairment. These findings indicate that peripheral nerve injury (eg, SNI) affects the MT dynamic equilibrium, which is critical to neuronal structure and synaptic plasticity.


Asunto(s)
Disfunción Cognitiva/etiología , Hipocampo/metabolismo , Microtúbulos/metabolismo , Neuralgia/complicaciones , Animales , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/psicología , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Neuralgia/metabolismo , Neuralgia/psicología , Nocodazol/farmacología , Paclitaxel/farmacología , Ratas , Ratas Sprague-Dawley , Moduladores de Tubulina/farmacología
9.
CNS Neurosci Ther ; 23(12): 930-939, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28990373

RESUMEN

INTRODUCTION: Sigma-1 receptors (Sig-1Rs) are unique endoplasmic reticulum proteins that have been implicated in both neurodegenerative and ischemic diseases, such as Alzheimer's disease and stroke. Accumulating evidence has suggested that Sig-1R plays a role in neuroprotection and axon outgrowth. The underlying mechanisms of Sig-1R-mediated neuroprotection have been well elucidated. However, the mechanisms underlying the effects of Sig-1R on axon outgrowth are not fully understood. METHODS: To clarify this issue, we utilized immunofluorescence to compare the axon lengths of cultured naïve hippocampal neurons before and after the application of the Sig-1R agonist, SA4503. Then, electrophysiology and immunofluorescence were used to examine voltage-gated calcium ion channel (VGCCs) currents in the cell membranes and growth cones. RESULTS: We found that Sig-1R activation dramatically enhanced the axonal length of the naïve hippocampal neurons. Application of the Sig-1R antagonist NE100 and gene knockdown techniques both demonstrated the effects of Sig-1R. The growth-promoting effect of SA4503 was accompanied by the inhibition of voltage-gated Ca2+ influx and was recapitulated by incubating the neurons with the L-type, N-type, and P/Q-type VGCC blockers, nimodipine, MVIIA and ω-agatoxin IVA, respectively. This effect was unrelated to glial cells. The application of SA4503 transformed the growth cone morphologies from complicated to simple, which favored axon outgrowth. CONCLUSION: Sig-1R activation can enhance axon outgrowth and may have a substantial influence on neurogenesis and neurodegenerative diseases.


Asunto(s)
Anisoles/farmacología , Axones/efectos de los fármacos , Canales de Calcio/metabolismo , Hipocampo/citología , Neuronas , Propilaminas/farmacología , Receptores sigma/agonistas , Análisis de Varianza , Animales , Animales Recién Nacidos , Bloqueadores de los Canales de Calcio/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Potenciales de la Membrana/efectos de los fármacos , Morfolinas/farmacología , Neuritas/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Nootrópicos/farmacología , Técnicas de Placa-Clamp , Piperazinas/farmacología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Tubulina (Proteína)/metabolismo , Receptor Sigma-1
10.
J Mol Neurosci ; 62(1): 17-27, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28341892

RESUMEN

The water channel aquaporin 4 (AQP4) is abundantly expressed in astrocytes and provides a mechanism by which water permeability of the plasma membrane can be regulated. Evidence suggests that AQP4 is associated with glutamate transporter-1 (GLT-1) for glutamate clearance and contributes to morphine dependence. Previous studies show that AQP4 deficiency changed the mu opioid receptor expression and opioid receptors' characteristics as well. In this study, we focused on whether AQP4 could form macromolecular complex with GLT-1 and mu opioid receptor (MOR) and participates in morphine dependence. By using immunofluorescence staining, fluorescence resonance energy transfer, and co-immunoprecipitation, we demonstrated that AQP4 forms protein complexes with GLT-1 and MOR in both brain tissue and primary cultured astrocytes. We then showed that the C-terminus of AQP4 containing the amino acid residues 252 to 323 is the site of interaction with GLT-1. Protein kinase C, activated by morphine, played an important role in regulating the expression of these proteins. These findings may help to reveal the mechanism that AQP4, GLT-1, and MOR form protein complex and participate in morphine dependence, and deeply understand the reason that AQP4 deficiency maintains extracellular glutamate homeostasis and attenuates morphine dependence, moreover emphasizes the function of astrocyte in morphine dependence.


Asunto(s)
Acuaporina 4/metabolismo , Astrocitos/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Dependencia de Morfina/metabolismo , Receptores Opioides mu/metabolismo , Animales , Acuaporina 4/química , Sitios de Unión , Encéfalo/metabolismo , Células Cultivadas , Masculino , Unión Proteica , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Psychopharmacology (Berl) ; 233(15-16): 3017-29, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27329413

RESUMEN

RATIONALE: Methamphetamine addiction is believed to primarily result from increased dopamine release and the inhibition of dopamine uptake. Some evidence suggests that hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play important roles in the functional modulation of dopaminergic neurons and the pathophysiology of related diseases. However, little is known about the effects of HCN channels on methamphetamine addiction. OBJECTIVES: The present study investigated the role of brain HCN channels in methamphetamine addiction. RESULTS: Acute intracerebroventricular (i.c.v.) injection or bilateral intra-accumbens microinjections of non-selective HCN channel blocker ZD7288 (0.3125 and 0.625 µg) significantly reduced both methamphetamine (0.0125 or 0.05 mg/kg/infusion)-induced self-administration under fixed ratio 2 reinforcement and the breakpoint of methamphetamine (0.05 mg/kg/infusion) under progressive ratio reinforcement in rats. Moreover, compared with i.c.v. injection, bilateral intra-accumbens microinjections of ZD7288 exerted stronger inhibitory effects, suggesting that blockade of HCN channels in the nucleus accumbens reduced the reinforcing effects of and motivation for methamphetamine. We also found that ZD7288 (0.625 and 1.25 µg, i.c.v.) significantly decreased methamphetamine (1 mg/kg, intraperitoneal (i.p.))-induced hyperactivity with no effect on the spontaneous activity in rats. Finally, in vivo microdialysis experiments showed that the HCN channel blockade using ZD7288 (0.625 and 1.25 µg, i.c.v.) decreased methamphetamine (1 mg/kg, i.p.)-induced elevation of extracellular dopamine levels in the nucleus accumbens. CONCLUSIONS: These results indicate that HCN channels in the nucleus accumbens are involved in the reinforcing properties of methamphetamine and highlight the importance of HCN channels in the regulation of dopamine neurotransmission underlying methamphetamine addiction.


Asunto(s)
Conducta Animal/efectos de los fármacos , Fármacos Cardiovasculares/farmacología , Estimulantes del Sistema Nervioso Central/administración & dosificación , Dopamina/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Metanfetamina/administración & dosificación , Núcleo Accumbens/efectos de los fármacos , Pirimidinas/farmacología , Animales , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Masculino , Microdiálisis , Núcleo Accumbens/metabolismo , Ratas , Ratas Sprague-Dawley , Autoadministración
12.
Front Cell Neurosci ; 10: 71, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27047336

RESUMEN

The striatum is the most prominent nucleus in the basal ganglia and plays an important role in motor movement regulation. The cholinergic interneurons (ChIs) in striatum are involved in the motion regulation by releasing acetylcholine (ACh) and modulating the output of striatal projection neurons. Here, we report that muscarinic ACh receptor (M receptor) agonists, ACh and Oxotremorine (OXO-M), decreased the firing frequency of ChIs by blocking the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. Scopolamine (SCO), a nonselective antagonist of M receptors, abolished the inhibition. OXO-M exerted its function by activating the Gi/o cAMP signaling cascade. The single-cell reverse transcription polymerase chain reaction (scRT-PCR) revealed that all the five subtypes of M receptors and four subtypes of HCN channels were expressed on ChIs. Among them, M2 receptors and HCN2 channels were the most dominant ones and expressed in every single studied cholinergic interneuron (ChI).Our results suggest that ACh regulates not only the output of striatal projection neurons, but also the firing activity of ChIs themselves by activating presynaptic M receptors in the dorsal striatum. The activation of M2 receptors and blockage of HCN2 channels may play an important role in ACh inhibition on the excitability of ChIs. This finding adds a new G-protein coupled receptor mediated regulation on ChIs and provides a cellular mechanism for control of cholinergic activity and ACh release in the dorsal striatum.

13.
Neuropharmacology ; 105: 420-433, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26855147

RESUMEN

Neuropeptide S (NPS), an endogenous anxiolytic, has been shown to protect against chronic pain through interacting with its cognate NPS receptor (NPSR) in the brain. However, the cellular mechanism of this NPS action remains unclear. We report that NPS inhibits hyperpolarization-activated cyclic nucleotide-gated (HCN) channel current (Ih) in the rat's amygdala through activation of NPSR. This NPS effect is mediated through ERK1/2 phosphorylation in a subset of pyramidal-like neurons located in the medial amygdala. The characters of the recorded Ih suggest a major role for HCN1 activity in this process. Inhibition of Ih by NPS stimulates the glutamatergic drive onto fast spiking intra-amygdalolidal GABAergic interneurons, which in turn facilitates GABA release onto pyramidal-like neurons. Moreover, the HCN1 expression is increased in the amygdala of rats with peripheral nerve injury and intra-amygdaloidal administration of the HCN channel inhibitor ZD7288 attenuates nociceptive behavior in these rats. These results suggest that NPS-mediated modulation of intra-amygdaloidal HCN channel activities may be an important central inhibitory mechanism for regulation of chronic pain.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Dolor Crónico/tratamiento farmacológico , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/efectos de los fármacos , Neuropéptidos/farmacología , Canales de Potasio/efectos de los fármacos , Animales , Ácido Glutámico/metabolismo , Hiperalgesia/tratamiento farmacológico , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Técnicas de Placa-Clamp , Células Piramidales/efectos de los fármacos , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Taquicininas , Ácido gamma-Aminobutírico/metabolismo , Ácido gamma-Aminobutírico/fisiología
14.
J Physiol Biochem ; 71(4): 601-10, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26271386

RESUMEN

TREK-2 (TWIK-related K(+) channel-2), a member of two-pore domain potassium (K2P) channel family, tunes cellular excitability via conducting leak or background currents. In TREK-2, the isoforms generated by alternative translation initiation (ATI) mechanism exhibit large divergence in unitary conductance, but similar in selectivity to K(+). Up to now, the structural basis for this similarity in ion selectivity is unknown. Here, we report that externally applied Ba(2+) inhibits the currents of TREK-2 in a concentration- and time-dependent manner. The blocking effect is blunted by elevated extracellular K(+) or mutation of S4 K(+) binding site, which suggests that the inhibitory mechanism of Ba(2+) is due to its competitive docking properties within the selectivity filter (SF). Next, we demonstrate that all the ATI isoforms exhibit analogous behaviors upon the application of Ba(2+) and alteration of extracellular pH (pHo), which acts on the outer position of the SF. These results strongly support the notion that all the ATI isoforms of TREK-2 possess resembled SF conformation in S4 site and the position defined by pHo, which implicates that neither the role of N-terminus (Nt) nor the unitary conductance is associated with SF conformation. Our findings might help to understand the detail gating mechanism of TREK-2 and K2P channels.


Asunto(s)
Canales de Potasio de Dominio Poro en Tándem/fisiología , Secuencia de Aminoácidos , Animales , Compuestos de Bario/farmacología , Sitios de Unión , Células Cultivadas , Cloruros/farmacología , Femenino , Humanos , Concentración 50 Inhibidora , Potenciales de la Membrana , Datos de Secuencia Molecular , Iniciación de la Cadena Peptídica Traduccional , Potasio/farmacología , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/química , Unión Proteica , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína , Xenopus laevis
15.
Biochem Biophys Res Commun ; 463(3): 200-4, 2015 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25998380

RESUMEN

The corticotropin releasing factor (CRF) plays a central role in regulating the activities of hypothalamic-pituitary-adrenal (HPA) axis in the presence of a variety of stressful stimuli via binding to its type 1 receptors (CRFR1). Despite that many peptidic or non-peptidic antagonists of CRFR1 have been developed to serve as therapeutic tools to CRF-related pathologies, none of them have been utilized clinically. Targeting the extracellular domain 1 (EC1) of CRFR1, the CRF-binding site, represents a new strategy to inhibit the function of the receptor. However, no such agents have been identified up to now. Herein, by using an 87-amino acid fragment corresponding to the EC1 region as the bait, we screened the binding polypeptides from a phage display (Ph.D.-12) peptide library. After 3-round biopanning, positive clones were selected and the polypeptides carried by them were identified. 5 polypeptides were found to bind with the target specifically. Among them, the P7 exhibited the highest affinity. By evaluating the cAMP accumulation in the CRFR1 or CRFR2-expressing HEK293 cells, we demonstrated that P7 blocking the function of CRFR1, but not CRFR2. In addition, we also found that P7 and CRF act on CRFR1 competitively. Taken together, we reveal that P7, a novel polypeptide identified from phage display library, inhibits the function of CRFR1 effectively and specifically by binding at its EC1 domain. The new polypeptide might provide a promising agent for diagnostic or therapeutic utilities in CRF-related disorders.


Asunto(s)
Péptidos/química , Péptidos/farmacología , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Secuencia de Aminoácidos , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Biblioteca de Péptidos , Estructura Terciaria de Proteína , Receptores de Hormona Liberadora de Corticotropina/química
16.
Brain Res ; 1605: 12-21, 2015 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-25681549

RESUMEN

Small molecular inhibitors of Cav2.2 have been reported for the treatment of neuropathic pain; however, low selectivity and side effects limit their further development. In our study, a series of new compounds were designed and synthesized by optimizing the 4-amino-piperidine template. The results show that ZC88 inhibits transiently expressed Cav2.2 in state-dependent manner in oocytes with an IC50 of 0.45 ± 0.09 µM. The steady-state inactivation relationship curve is shifted to more negative potentials for the calcium channels, suggesting that ZC88 blocks inactivated state of the channel. ZC88 does not present any remarkable effects on voltage-gated P/Q-type calcium channel currents, l-type calcium channel currents, potassium channel and sodium channel currents. Taken together, these in vitro data suggest that ZC88 is a voltage-dependent, subtype-selective Cav2.2 channel inhibitor and can achieve an improved therapeutic window over the relatively state-independent Cav2.2-selective inhibitor, which may have potential to be developed into a novel analgesic agent.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Piperidinas/farmacología , Proteínas de Xenopus/antagonistas & inhibidores , Analgésicos no Narcóticos/farmacología , Animales , Canales de Calcio Tipo N , Células Cultivadas , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Potenciales de la Membrana/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Conejos , Ratas , Ratas Wistar , Canales de Sodio Activados por Voltaje/metabolismo , Xenopus laevis
17.
Brain Res ; 1582: 1-11, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25091639

RESUMEN

Methamphetamine (METH) is a psychostimulant drug. Abuse of METH produces long-term behavioral changes including behavioral, sensitization, tolerance, and dependence. It induces neurotoxic effects in several areas of the brain via enhancing dopamine (DA) level abnormally, which may cause a secondary release of glutamate (GLU). However, repeated administration of METH still increases release of GLU even when dopamine content in tissue is significantly depleted. It implies that some other mechanisms are likely to involve in METH-induced GLU release. The goal of this study was to observe METH affected glutamatergic synaptic transmission in rat primary cultured hippocampal neurons and to explore the mechanism of METH modulated GLU release. Using whole-cell patch-clamp recordings, we found that METH (0.1-50.0µM) increased the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) and miniature excitatory postsynaptic currents (mEPSCs). However, METH decreased the frequency of sEPSCs and mEPSCs at high concentration of 100µM. The postsynaptic NMDA receptor currents and P/Q-type calcium channel were not affected by the use of METH (10,100µM). METH did not present visible effect on N-type Ca(2+) channel current at the concentration lower than 50.0µM, but it was inhibited by use of METH at a 100µM. The effect of METH on glutamatergic synaptic transmission was not revered by pretreated with DA receptor antagonist SCH23390. These results suggest that METH directly modulated presynaptic GLU release at a different concentration, while dopaminergic system was not involved in METH modulated release of GLU in rat primary cultured hippocampal neurons.


Asunto(s)
Estimulantes del Sistema Nervioso Central/farmacología , Ácido Glutámico/metabolismo , Hipocampo/efectos de los fármacos , Metanfetamina/farmacología , Neuronas/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Benzazepinas/farmacología , Canales de Calcio Tipo N/metabolismo , Células Cultivadas , Dopamina/metabolismo , Antagonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Hipocampo/fisiología , Inmunohistoquímica , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/fisiología , Ratas Wistar , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/fisiología , Tirosina 3-Monooxigenasa/metabolismo
18.
Neuroreport ; 25(12): 887-93, 2014 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-25006846

RESUMEN

In an attempt to investigate the underlying mechanisms of cancer-induced bone pain, we investigated the presence of acid-sensing ion channel 3 (ASIC3) in dorsal root ganglia (DRG) neurons in an animal model of bone cancer pain. Forty-five female Sprague-Dawley rats were randomized into three groups: sham-operation group (sham), cancer-bearing animals killed after 7 days (C7), and cancer-bearing animals killed after 14 days (C14). After establishment of the bone cancer pain model, pain-related behavioral tests were performed to determine the paw withdrawal threshold of mechanical allodynia and thermal hyperalgesia, respectively. Reverse transcription-PCR, western blot, and immunofluorescence were used to determine mRNA and protein expression of ASIC3 in ipsilateral and contralateral lumbar 4-5 DRG neurons. Compared with the sham group, paw withdrawal threshold of mechanical allodynia and thermal hyperalgesia in the C14 group showed a significant decrease (P<0.01) from postoperation day 7 to the termination of the experiment. Compared with the sham group, the ipsilateral but not contralateral mRNA of ASIC3 was upregulated in the C14 group. Meanwhile, the ipsilateral protein expression of ASIC3 was increased in the C7 and C14 group compared with the sham group. Double-labeled immunofluorescence showed that ASIC3 and isolectin-B4 (IB4)-colocalized small DRG neurons in the C14 group were more than that in the sham group. Furthermore, we also found that there were more ASIC3 and neurofilament 200 (NF200)-colocalized DRG neurons in the C14 group than in the sham group. The upregulation of mRNA and protein levels of ASIC3 suggested its potential involvement in the development and maintenance of cancer-induced bone pain.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Neoplasias Óseas/metabolismo , Carcinoma/metabolismo , Ganglios Espinales/metabolismo , Dolor/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Lateralidad Funcional , Glicoproteínas/metabolismo , Calor , Hiperalgesia/metabolismo , Lectinas/metabolismo , Vértebras Lumbares , Trasplante de Neoplasias , Neuronas/metabolismo , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas Sprague-Dawley , Factores de Tiempo , Tacto , Versicanos
19.
Pain ; 155(8): 1504-1515, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24793908

RESUMEN

Anxiety disorder is a comorbid condition of chronic pain. Analgesics and anxiolytics, subject to addiction and abuse, are currently used to manage pain and anxiety symptoms. However, the cellular mechanism underlying chronic pain and anxiety interaction remains to be elucidated. We report that persistent nociception following peripheral nerve injury induced anxiety-like behavior in rodents. Brain expression and release of neuropeptide S (NPS), a proposed endogenous anxiolytic peptide, was diminished in rodents with coexisting nociceptive and anxiety-like behaviors. Intracerebroventricular administration of exogenous NPS concurrently improved both nociceptive and anxiety-like behaviors. At the cellular level, NPS enhanced intra-amygdaloidal inhibitory transmission by increasing presynaptic gamma-aminobutyric acid (GABA) release from interneurons. These findings indicate that the interaction between nociceptive and anxiety-like behaviors in rodents may be regulated by the altered NPS-mediated intra-amygdaloidal GABAergic inhibition. The data suggest that enhancing the brain NPS function may be a new strategy to manage comorbid pain and anxiety.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ansiedad/metabolismo , Conducta Animal/fisiología , Neuropéptidos/metabolismo , Nocicepción/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiopatología , Animales , Ansiedad/fisiopatología , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Masculino , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Neuropéptidos/farmacología , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/metabolismo
20.
Brain Res ; 1535: 137-47, 2013 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-23998984

RESUMEN

Previous studies have shown that the morphology and number of cells in the spinal cord dorsal horn could change following peripheral nerve injury and that the Hippo signaling pathway plays an important role in cell growth, proliferation, apoptosis, and dendritic remolding. In the present study, we examined whether the expression of YAP and TAZ, two critical components regulated by Hippo signaling, in the spinal cord dorsal horn would be altered by chronic constriction sciatic nerve injury (CCI). We found that (1) YAP was mainly expressed on CGRP- and IB4-immunoreactive primary afferent nerve terminals without noticeable expression on glial cells, whereas TAZ was mainly expressed on spinal cord second order neurons as well as microglia; (2) upregulation of YAP and TAZ expression followed two distinct temporal patterns after CCI, such that the highest expression of YAP and TAZ was on day 14 and day 1 after CCI, respectively; (3) there were also unique topographic patterns of YAP and TAZ distribution in the spinal cord dorsal horn consistent with their distinctive association with primary afferents and second order neurons; (4) changes in the YAP expression were selectively induced by CCI but not CFA-induced hindpaw inflammation; and (5) the number of nuclear profiles of TAZ expression was significantly increased after CCI, indicating translocation of TAZ from the cytoplasma to nucleus. These findings indicate that peripheral nerve injury induced time-dependent and region-specific changes in the spinal YAP and TAZ expression. A role for Hippo signaling in synaptic and structural plasticity is discussed in relation to the cellular mechanism of neuropathic pain.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Transducción de Señal/fisiología , Médula Espinal/metabolismo , Factores de Transcripción/metabolismo , Aciltransferasas , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Hiperalgesia/metabolismo , Lectinas/metabolismo , Masculino , Neuralgia/metabolismo , Neuronas/metabolismo , Dimensión del Dolor , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Regulación hacia Arriba , Proteínas Señalizadoras YAP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...