Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 728, 2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-35132099

RESUMEN

Postsynaptic NMDARs at spinal synapses are required for postsynaptic long-term potentiation and chronic pain. However, how presynaptic NMDARs (PreNMDARs) in spinal nociceptor terminals control presynaptic plasticity and pain hypersensitivity has remained unclear. Here we report that PreNMDARs in spinal nociceptor terminals modulate synaptic transmission in a nociceptive tone-dependent manner. PreNMDARs depresses presynaptic transmission in basal state, while paradoxically causing presynaptic potentiation upon injury. This state-dependent modulation is dependent on Ca2+ influx via PreNMDARs. Small conductance Ca2+-activated K+ (SK) channels are responsible for PreNMDARs-mediated synaptic depression. Rather, tissue inflammation induces PreNMDARs-PKG-I-dependent BDNF secretion from spinal nociceptor terminals, leading to SK channels downregulation, which in turn converts presynaptic depression to potentiation. Our findings shed light on the state-dependent characteristics of PreNMDARs in spinal nociceptor terminals on modulating nociceptive transmission and revealed a mechanism underlying state-dependent transition. Moreover, we identify PreNMDARs in spinal nociceptor terminals as key constituents of activity-dependent pain sensitization.


Asunto(s)
Dolor Crónico/fisiopatología , Nociceptores/metabolismo , Terminales Presinápticos/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Calcio/metabolismo , Dolor Crónico/genética , Dolor Crónico/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Ganglios Espinales/citología , Ganglios Espinales/fisiología , Inflamación , Potenciación a Largo Plazo , Depresión Sináptica a Largo Plazo , Ratones , Ratones Transgénicos , Sustancia Gris Periacueductal/citología , Sustancia Gris Periacueductal/fisiología , Canales de Potasio Calcio-Activados/genética , Canales de Potasio Calcio-Activados/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Transmisión Sináptica
2.
Nat Neurosci ; 22(3): 503, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30643300

RESUMEN

In the version of this article initially published, what was originally described as 'conditioned place preference' in a two-chamber mouse experiment could be better described as 'conditioned place avoidance'.

3.
Mol Pain ; 13: 1744806917707127, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28587505

RESUMEN

Cervical radiculopathic pain is a very common symptom that may occur with cervical spondylosis. Mechanical allodynia is often associated with cervical radiculopathic pain and is inadequately treated with current therapies. However, the precise mechanisms underlying cervical radiculopathic pain-associated mechanical allodynia have remained elusive. Compelling evidence from animal models suggests a role of large-diameter dorsal root ganglion neurons and plasticity of spinal circuitry attached with Aß fibers in mediating neuropathic pain. Whether cervical radiculopathic pain condition induces plastic changes of large-diameter dorsal root ganglion neurons and what mechanisms underlie these changes are yet to be known. With combination of patch-clamp recording, immunohistochemical staining, as well as behavioral surveys, we demonstrated that upon chronic compression of C7/8 dorsal root ganglions, large-diameter cervical dorsal root ganglion neurons exhibited frequent spontaneous firing together with hyperexcitability. Quantitative analysis of hyperpolarization-activated cation current ( Ih) revealed that Ih was greatly upregulated in large dorsal root ganglion neurons from cervical radiculopathic pain rats. This increased Ih was supported by the enhanced expression of hyperpolarization-activated, cyclic nucleotide-modulated channels subunit 3 in large dorsal root ganglion neurons. Blockade of Ih with selective antagonist, ZD7288 was able to eliminate the mechanical allodynia associated with cervical radiculopathic pain. This study sheds new light on the functional plasticity of a specific subset of large-diameter dorsal root ganglion neurons and reveals a novel mechanism that could underlie the mechanical allodynia associated with cervical radiculopathy.


Asunto(s)
Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Neuralgia/etiología , Neuralgia/metabolismo , Neuronas/citología , Neuronas/metabolismo , Radiculopatía/etiología , Radiculopatía/metabolismo , Animales , Dolor Crónico/etiología , Dolor Crónico/metabolismo , Dolor Crónico/patología , Masculino , Potenciales de la Membrana/fisiología , Neuralgia/patología , Neuronas Aferentes/citología , Neuronas Aferentes/metabolismo , Radiculopatía/patología , Ratas , Ratas Sprague-Dawley
4.
Nat Neurosci ; 20(7): 917-926, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28530662

RESUMEN

Programmed cell death ligand-1 (PD-L1) is typically produced by cancer cells and suppresses immunity through the receptor PD-1 expressed on T cells. However, the role of PD-L1 and PD-1 in regulating pain and neuronal function is unclear. Here we report that both melanoma and normal neural tissues including dorsal root ganglion (DRG) produce PD-L1 that can potently inhibit acute and chronic pain. Intraplantar injection of PD-L1 evoked analgesia in naive mice via PD-1, whereas PD-L1 neutralization or PD-1 blockade induced mechanical allodynia. Mice lacking Pd1 (Pdcd1) exhibited thermal and mechanical hypersensitivity. PD-1 activation in DRG nociceptive neurons by PD-L1 induced phosphorylation of the tyrosine phosphatase SHP-1, inhibited sodium channels and caused hyperpolarization through activation of TREK2 K+ channels. PD-L1 also potently suppressed nociceptive neuron excitability in human DRGs. Notably, blocking PD-L1 or PD-1 elicited spontaneous pain and allodynia in melanoma-bearing mice. Our findings identify a previously unrecognized role of PD-L1 as an endogenous pain inhibitor and a neuromodulator.


Asunto(s)
Analgesia , Antígeno B7-H1/fisiología , Cultivo Primario de Células , Receptor de Muerte Celular Programada 1/fisiología , Animales , Antígeno B7-H1/sangre , Antígeno B7-H1/farmacología , Células Cultivadas , Cricetinae , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ganglios Espinales/fisiología , Humanos , Hiperalgesia/inducido químicamente , Masculino , Melanoma/sangre , Melanoma/fisiopatología , Ratones , Ratones Noqueados , Neuralgia/fisiopatología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Fosforilación , Canales de Potasio de Dominio Poro en Tándem/fisiología , Receptor de Muerte Celular Programada 1/biosíntesis , Receptor de Muerte Celular Programada 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Ratas , Canales de Sodio/fisiología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
5.
Brain Behav Immun ; 55: 70-81, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26472019

RESUMEN

Previous studies have shown that activation of p38 mitogen-activating kinase (MAPK) in spinal microglia participates in the generation of inflammatory and neuropathic pain in various rodent models. However, these studies focused on male mice to avoid confounding effects of the estrous cycle of females. Recent studies have shown that some spinal pro-inflammatory signaling such as Toll-like receptor 4-mediated signaling contributes to pain hypersensitivity only in male mice. In this study we investigated the distinct role of spinal p38 in inflammatory and neuropathic pain using a highly selective p38 inhibitor skepinone. Intrathecal injection of skepinone prevented formalin induced inflammatory pain in male but not female mice. Furthermore, intrathecal skepinone reduced chronic constriction injury (CCI) induced neuropathic pain (mechanical allodynia) in male mice on CCI-day 7 but not CCI-day 21. This male-dependent inhibition of neuropathic pain also occurred in rats following intrathecal skepinone. Nerve injury induced spinal p38 activation (phosphorylation) in CX3CR1-GFP(+) microglia on CCI-day 7, and this activation was more prominent in male mice. In contrast, CCI induced comparable microgliosis and expression of the microglial markers CX3CR1 and IBA-1 in both sexes. Notably, intraperitoneal or local perineural administration of skepinone inhibited CCI-induced mechanical allodynia in both sexes of mice. Finally, skepinone only reduced the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) in lamina IIo neurons of spinal cord slices of males 7days post CCI. Therefore, the sex-specific p38 activation and signaling is confined to the spinal cord in inflammatory and neuropathic pain conditions.


Asunto(s)
Inflamación/tratamiento farmacológico , Microglía/metabolismo , Neuralgia/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Médula Espinal/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Dibenzocicloheptenos/administración & dosificación , Dibenzocicloheptenos/farmacología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidores de Proteínas Quinasas/administración & dosificación , Factores Sexuales
6.
Sci Rep ; 5: 16713, 2015 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-26577374

RESUMEN

Cervical radiculopathy represents aberrant mechanical hypersensitivity. Primary sensory neuron's ability to sense mechanical force forms mechanotransduction. However, whether this property undergoes activity-dependent plastic changes and underlies mechanical hypersensitivity associated with cervical radiculopathic pain (CRP) is not clear. Here we show a new CRP model producing stable mechanical compression of dorsal root ganglion (DRG), which induces dramatic behavioral mechanical hypersensitivity. Amongst nociceptive DRG neurons, a mechanically sensitive neuron, isolectin B4 negative Aδ-type (IB4(-) Aδ) DRG neuron displays spontaneous activity with hyperexcitability after chronic compression of cervical DRGs. Focal mechanical stimulation on somata of IB4(-) Aδ neuron induces abnormal hypersensitivity. Upregulated HCN1 and HCN3 channels and increased Ih current on this subset of primary nociceptors underlies the spontaneous activity together with neuronal mechanical hypersensitivity, which further contributes to the behavioral mechanical hypersensitivity associated with CRP. This study sheds new light on the functional plasticity of a specific subset of nociceptive DRG neurons to mechanical stimulation and reveals a novel mechanism that could underlie the mechanical hypersensitivity associated with cervical radiculopathy.


Asunto(s)
Ganglios Espinales/metabolismo , Ganglios Espinales/fisiopatología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Mecanotransducción Celular , Nociceptores/metabolismo , Radiculopatía/genética , Radiculopatía/fisiopatología , Animales , Modelos Animales de Enfermedad , Expresión Génica , Genes fos , Hiperalgesia/etiología , Potenciales de la Membrana , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fibras Nerviosas Mielínicas/metabolismo , Neuralgia/etiología , Fosforilación , Radiculopatía/complicaciones , Radiculopatía/etiología , Ratas , Regulación hacia Arriba
7.
PLoS Biol ; 10(3): e1001283, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22427743

RESUMEN

Synaptic long-term potentiation (LTP) at spinal neurons directly communicating pain-specific inputs from the periphery to the brain has been proposed to serve as a trigger for pain hypersensitivity in pathological states. Previous studies have functionally implicated the NMDA receptor-NO pathway and the downstream second messenger, cGMP, in these processes. Because cGMP can broadly influence diverse ion-channels, kinases, and phosphodiesterases, pre- as well as post-synaptically, the precise identity of cGMP targets mediating spinal LTP, their mechanisms of action, and their locus in the spinal circuitry are still unclear. Here, we found that Protein Kinase G1 (PKG-I) localized presynaptically in nociceptor terminals plays an essential role in the expression of spinal LTP. Using the Cre-lox P system, we generated nociceptor-specific knockout mice lacking PKG-I specifically in presynaptic terminals of nociceptors in the spinal cord, but not in post-synaptic neurons or elsewhere (SNS-PKG-I(-/-) mice). Patch clamp recordings showed that activity-induced LTP at identified synapses between nociceptors and spinal neurons projecting to the periaqueductal grey (PAG) was completely abolished in SNS-PKG-I(-/-) mice, although basal synaptic transmission was not affected. Analyses of synaptic failure rates and paired-pulse ratios indicated a role for presynaptic PKG-I in regulating the probability of neurotransmitter release. Inositol 1,4,5-triphosphate receptor 1 and myosin light chain kinase were recruited as key phosphorylation targets of presynaptic PKG-I in nociceptive neurons. Finally, behavioural analyses in vivo showed marked defects in SNS-PKG-I(-/-) mice in several models of activity-induced nociceptive hypersensitivity, and pharmacological studies identified a clear contribution of PKG-I expressed in spinal terminals of nociceptors. Our results thus indicate that presynaptic mechanisms involving an increase in release probability from nociceptors are operational in the expression of synaptic LTP on spinal-PAG projection neurons and that PKG-I localized in presynaptic nociceptor terminals plays an essential role in this process to regulate pain sensitivity.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Potenciación a Largo Plazo , Nociceptores/metabolismo , Dolor/patología , Aminoquinolinas/farmacología , Animales , Conducta Animal , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Activación Enzimática , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Eliminación de Gen , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Fibras Nerviosas/metabolismo , Fibras Nerviosas/patología , Nociceptores/efectos de los fármacos , Nociceptores/patología , Dolor/metabolismo , Técnicas de Placa-Clamp , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Transducción de Señal , Especificidad por Sustrato , Transmisión Sináptica
8.
Neurosci Bull ; 25(4): 221-7, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19633705

RESUMEN

The transient receptor potential vanilloid subfamily member 1 (TRPV1) is a protein mainly expressed in sensory neurons and fibers, such as in trigeminal ganglion and dorsal root ganglion, and has been indicated to be involved in several physiological and pathological processes. Studies on thermal activation have revealed that phosphorylation is involved in TRPV1 activation and 2 putative phosphorylation sites, Ser residues 502 (Ser-502) and Ser residues 800 (Ser-800), have been recently confirmed to possess the capability of resensitizing TRPV1. In addition to acidification, alkalization has also been proved to be a highly effective stimulator for TRPV1. TRPV1 could be regulated by various physical and chemical modulators, as well as the chronic pain. TRPV1 plays a crucial role in the transmission of pain signals, especially under inflammation and the neoplasm conditions, and it can also modulate nociceptive afferents by reinforcing morphine tolerance. The present review mainly focused on the structural and functional complexities of TRPV1, together with its activation and modulation by a wide variety of physical and chemical stimuli. Its pharmacological manipulation (sensitization/desensitization) and therapeutical targets were also discussed.


Asunto(s)
Vías Aferentes/citología , Sensación/fisiología , Células Receptoras Sensoriales/metabolismo , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Animales , Humanos , Fosforilación/fisiología , Estimulación Física , Serina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...