Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Pain ; 18: 17448069221107781, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35647699

RESUMO

Spinal neuroinflammation plays a critical role in the genesis of neuropathic pain. Accumulating data suggest that abscisic acid (ABA), a phytohormone, regulates inflammatory processes in mammals. In this study, we found that reduction of the LANCL2 receptor protein but not the agonist ABA in the spinal cord is associated with the genesis of neuropathic pain. Systemic or intrathecal administration of ABA ameliorates the development and pre-existence of mechanical allodynia and heat hyperalgesia in animals with partial sciatic nerve ligation (pSNL). LANCL2 is expressed only in microglia in the spinal dorsal horn. Pre-emptive treatment with ABA attenuates activation of microglia and astrocytes, ERK activity, and TNFα protein abundance in the dorsal horn in rats with pSNL. These are accompanied by restoration of spinal LANCL2 protein abundance. Spinal knockdown of LANCL2 gene with siRNA recapitulates the behavioral and spinal molecular changes induced by pSNL. Activation of spinal toll-like receptor 4 (TLR4) with lipopolysaccharide leads to activation of microglia, and over production of TNFα, which are concurrently accompanied by suppression of protein levels of LANCL2 and peroxisome proliferator activated-receptor γ. These changes are ameliorated when ABA is added with LPS. The anti-inflammatory effects induced by ABA do not requires Gi protein activity. Our study reveals that the ABA/LANCL2 system is a powerful endogenous system regulating spinal neuroinflammation and nociceptive processing, suggesting the potential utility of ABA as the management of neuropathic pain.


Assuntos
Ácido Abscísico , Neuralgia , Ácido Abscísico/metabolismo , Ácido Abscísico/farmacologia , Animais , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Lipopolissacarídeos/farmacologia , Mamíferos , Proteínas de Membrana/metabolismo , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Reguladores de Crescimento de Plantas/metabolismo , Reguladores de Crescimento de Plantas/farmacologia , Ratos , Medula Espinal/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
2.
Glia ; 67(3): 482-497, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30578561

RESUMO

Patients receiving paclitaxel for cancer treatment often develop an acute pain syndrome (paclitaxel-associated acute pain syndrome, P-APS), which occurs immediately after paclitaxel treatment. Mechanisms underlying P-APS remain largely unknown. We recently reported that rodents receiving paclitaxel develop acute pain and activation of spinal microglial toll like receptor 4 (TLR4) by paclitaxel penetrating into the spinal cord is a critical event in the genesis of P-APS. Our current study dissected cellular and molecular mechanisms underlying the P-APS. We demonstrated that bath-perfusion of paclitaxel, at a concentration similar to that found in the cerebral spinal fluid in animals receiving i.v. paclitaxel (2 mg/kg), resulted in increased calcium activity in microglia instantly, and in astrocytes with 6 min delay. TLR4 activation in microglia by paclitaxel caused microglia to rapidly release interleukin-1ß (IL-1ß) but not tumor necrosis factor α, IL-6, or interferon-γ. IL-1ß release from microglia depended on capthepsin B. IL-1ß acted on astrocytes, leading to elevated calcium activity and suppressed glutamate uptake. IL-1ß also acted on neurons to increase presynaptic glutamate release and postsynaptic AMPA receptor activity in the spinal dorsal horn. Knockout of IL-1 receptors prevented the development of acute pain induced by paclitaxel in mice. Our study indicates that IL-1ß is a crucial molecule used by microglia to alter functions in astrocytes and neurons upon activation of TLR4 in the genesis of P-APS, and targeting the signaling pathways regulating the production and function of IL-1ß from microglia is a potential avenue for the development of analgesics for the treatment of P-APS.


Assuntos
Antineoplásicos/efeitos adversos , Ácido Glutâmico/metabolismo , Interleucina-1beta/metabolismo , Microglia/metabolismo , Paclitaxel/efeitos adversos , Dor/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Animais , Cálcio/metabolismo , Potenciais Pós-Sinápticos Excitadores/fisiologia , Masculino , Camundongos , Camundongos Knockout , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Dor/induzido quimicamente , Medição da Dor , Ratos
3.
J Neurochem ; 140(6): 963-976, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28072466

RESUMO

Systemic lupus erythematosus (SLE) is a multi-organ disease of unknown etiology in which the normal immune responses are directed against the body's own healthy tissues. Patients with SLE often suffer from chronic pain. Currently, no animal studies have been reported about the mechanisms underlying pain in SLE. In this study, the development of chronic pain in MRL lupus-prone (MRL/lpr) mice, a well-established lupus mouse model, was characterized for the first time. We found that female MRL/lpr mice developed thermal hyperalgesia at the age of 13 weeks, and mechanical allodynia at the age of 16 weeks. MRL/lpr mice with chronic pain had activation of microglia and astrocytes, over-expression of macrophage colony-stimulating factor-1 (CSF-1) and interleukin-1 beta (IL-1ß), as well as suppression of glial glutamate transport function in the spinal cord. Intrathecal injection of either the CSF-1 blocker or IL-1 inhibitor attenuated thermal hyperalgesia in MRL/lpr mice. We provide evidence that the suppressed activity of glial glutamate transporters in the spinal dorsal horn in MRL/lpr mice is caused by activation of the CSF-1 and IL-1ß signaling pathways. Our findings suggest that targeting the CSF-1 and IL-1ß signaling pathways or the glial glutamate transporter in the spinal cord is an effective approach for the management of chronic pain caused by SLE.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/fisiologia , Dor Crônica/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Neuroglia/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Animais , Anisóis/farmacologia , Anisóis/uso terapêutico , Dor Crônica/tratamento farmacológico , Dor Crônica/genética , Feminino , Lúpus Eritematoso Sistêmico/genética , Camundongos , Camundongos Transgênicos , Neuroglia/efeitos dos fármacos , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Receptor de Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores
4.
Neuroscience ; 326: 158-169, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27058143

RESUMO

Emerging studies have shown that pharmacological activation of adenosine monophosphate-activated protein kinase (AMPK) produces potent analgesic effects in different animal pain models. Currently, the spinal molecular and synaptic mechanism by which AMPK regulates the pain signaling system remains unclear. To address this issue, we utilized the Cre-LoxP system to conditionally knockout the AMPKα1 gene in the nervous system of mice. We demonstrated that AMPKα1 is imperative for maintaining normal nociception, and mice deficient for AMPKα1 exhibit mechanical allodynia. This is concomitantly associated with increased glutamatergic synaptic activities in neurons located in the superficial spinal dorsal horn, which results from the increased glutamate release from presynaptic terminals and function of ligand-gated glutamate receptors at the postsynaptic neurons. Additionally, AMPKα1 knockout mice have increased activities of extracellular signal-regulated kinases (ERK) and p38 mitogen-activated protein kinases (p38), as well as elevated levels of interleukin-1ß (IL-1ß), reactive oxygen species (ROS), and heme oxygenase 1 (HO-1) in the spinal dorsal horn. Systemic administration of a non-specific ROS scavenger (phenyl-N-tert-butylnitrone, PBN) or a HO-1 activator (Cobalt protoporphyrin IX, CoPP) attenuated allodynia in AMPKα1 knockout mice. Bath-perfusion of the ROS scavenger or HO-1 activator effectively attenuated the increased ROS levels and glutamatergic synaptic activities in the spinal dorsal horn. Our findings suggest that ROS are the key down-stream signaling molecules mediating the behavioral hypersensitivity in AMPKα1 knockout mice. Thus, targeting AMPKα1 may represent an effective approach for the treatment of pathological pain conditions associated with neuroinflammation at the spinal dorsal horn.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Ácido Glutâmico/fisiologia , Hiperalgesia/metabolismo , Nociceptividade/fisiologia , Terminações Pré-Sinápticas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/fisiologia , Animais , Óxidos N-Cíclicos/farmacologia , Encefalite/metabolismo , Potenciais Pós-Sinápticos Excitadores , Feminino , Sequestradores de Radicais Livres/farmacologia , Heme Oxigenase-1/metabolismo , Interleucina-1beta/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Knockout
5.
Mol Pain ; 11: 10, 2015 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-25868824

RESUMO

Paclitaxel, a powerful anti-neoplastic drug, often causes pathological pain, which significantly reduces the quality of life in patients. Paclitaxel-induced pain includes pain that occurs immediately after paclitaxel treatment (paclitaxel-associated acute pain syndrome, P-APS) and pain that persists for weeks to years after cessation of paclitaxel treatment (paclitaxel induced chronic neuropathic pain). Mechanisms underlying P-APS remain unknown. In this study, we found that paclitaxel causes acute pain in rodents in a dose-dependent manner. The paclitaxel-induced acute pain occurs within 2 hrs after a single intravenous injection of paclitaxel. This is accompanied by low levels of paclitaxel penetrating into the cerebral spinal fluid and spinal dorsal horn. We demonstrated that an intrathecal injection of paclitaxel induces mechanical allodynia in a dose-dependent manner. Paclitaxel causes activation of toll like receptor 4 (TLR4) in the spinal dorsal horn and dorsal root ganglions. Through activating TLR4, paclitaxel increases glutamatergic synaptic activities and reduces glial glutamate transporter activities in the dorsal horn. Activations of TLR4 are necessary in the genesis of paclitaxel-induced acute pain. The cellular and molecular signaling pathways revealed in this study could provide rationales for the development of analgesics and management strategies for P-APS in patients.


Assuntos
Dor Aguda/induzido quimicamente , Antineoplásicos Fitogênicos/toxicidade , Neuralgia/induzido quimicamente , Paclitaxel/toxicidade , Receptor 4 Toll-Like/metabolismo , Animais , Gânglios Espinais/efeitos dos fármacos , Masculino , Neuralgia/tratamento farmacológico , Medição da Dor/métodos , Limiar da Dor/efeitos dos fármacos , Ratos Sprague-Dawley
6.
J Neurochem ; 133(6): 857-69, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25827582

RESUMO

Paclitaxel is a chemotherapeutic agent widely used for treating carcinomas. Patients receiving paclitaxel often develop neuropathic pain and have a reduced quality of life which hinders the use of this life-saving drug. In this study, we determined the role of GABA transporters in the genesis of paclitaxel-induced neuropathic pain using behavioral tests, electrophysiology, and biochemical techniques. We found that tonic GABA receptor activities in the spinal dorsal horn were reduced in rats with neuropathic pain induced by paclitaxel. In normal controls, tonic GABA receptor activities were mainly controlled by the GABA transporter GAT-1 but not GAT-3. In the spinal dorsal horn, GAT-1 was expressed at presynaptic terminals and astrocytes while GAT-3 was only expressed in astrocytes. In rats with paclitaxel-induced neuropathic pain, the protein expression of GAT-1 was increased while GAT-3 was decreased. This was concurrently associated with an increase in global GABA uptake. The paclitaxel-induced attenuation of GABAergic tonic inhibition was ameliorated by blocking GAT-1 but not GAT-3 transporters. Paclitaxel-induced neuropathic pain was significantly attenuated by the intrathecal injection of a GAT-1 inhibitor. These findings suggest that targeting GAT-1 transporters for reversing disinhibition in the spinal dorsal horn may be a useful approach for treating paclitaxel-induced neuropathic pain. Patients receiving paclitaxel for cancer therapy often develop neuropathic pain and have a reduced quality of life. In this study, we demonstrated that animals treated with paclitaxel develop neuropathic pain, have enhancements of GABA transporter-1 protein expression and global GABA uptake, as well as suppression of GABAergic tonic inhibition in the spinal dorsal horn. Pharmacological inhibition of GABA transporter-1 ameliorates the paclitaxel-induced suppression of GABAergic tonic inhibition and neuropathic pain. Thus, targeting GAT-1 transporters for reversing GABAergic disinhibition in the spinal dorsal horn could be a useful approach for treating paclitaxel-induced neuropathic pain.


Assuntos
Antineoplásicos Fitogênicos/toxicidade , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Neuralgia/metabolismo , Paclitaxel/toxicidade , Corno Dorsal da Medula Espinal/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Western Blotting , Modelos Animais de Doenças , Imuno-Histoquímica , Masculino , Neuralgia/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Corno Dorsal da Medula Espinal/efeitos dos fármacos
7.
Anesthesiology ; 122(6): 1401-13, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25710409

RESUMO

BACKGROUND: Neuroinflammation and dysfunctional glial glutamate transporters (GTs) in the spinal dorsal horn are implicated in the genesis of neuropathic pain. The authors determined whether adenosine monophosphate-activated protein kinase (AMPK) in the spinal dorsal horn regulates these processes in rodents with neuropathic pain. METHODS: Hind paw withdrawal responses to radiant heat and mechanical stimuli were used to assess nociceptive behaviors. Spinal markers related to neuroinflammation and glial GTs were determined by Western blotting. AMPK activities were manipulated pharmacologically and genetically. Regulation of glial GTs was determined by measuring protein expression and activities of glial GTs. RESULTS: AMPK activities were reduced in the spinal dorsal horn of rats (n = 5) with thermal hyperalgesia induced by nerve injury, which were accompanied with the activation of astrocytes, increased production of interleukin-1ß and activities of glycogen synthase kinase 3ß, and suppressed protein expression of glial glutamate transporter-1. Thermal hyperalgesia was reversed by spinal activation of AMPK in neuropathic rats (n = 10) and induced by inhibiting spinal AMPK in naive rats (n = 7 to 8). Spinal AMPKα knockdown (n = 6) and AMPKα1 conditional knockout (n = 6) induced thermal hyperalgesia and mechanical allodynia. These genetic alterations mimicked the changes of molecular markers induced by nerve injury. Pharmacological activation of AMPK enhanced glial GT activity in mice with neuropathic pain (n = 8) and attenuated glial glutamate transporter-1 internalization induced by interleukin-1ß (n = 4). CONCLUSIONS: These findings suggest that enhancing spinal AMPK activities could be an effective approach for the treatment of neuropathic pain.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Interleucina-1beta/biossíntese , Neuralgia/metabolismo , Neuroglia/metabolismo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Animais , Comportamento Animal , Genótipo , Hiperalgesia/induzido quimicamente , Hiperalgesia/psicologia , Injeções Espinhais , Masculino , Camundongos , Camundongos Knockout , Neuroglia/efeitos dos fármacos , Limiar da Dor , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Neuropatia Ciática/metabolismo , Succinato Desidrogenase/metabolismo
8.
Exp Neurol ; 252: 18-27, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24275526

RESUMO

Dysfunctional glial glutamate transporters and over production of pro-inflammatory cytokines (including interleukin-1ß, IL-1ß) are two prominent mechanisms by which glial cells enhance neuronal activities in the spinal dorsal horn in neuropathic pain conditions. Endogenous molecules regulating production of IL-1ß and glial glutamate functions remain poorly understood. In this study, we revealed a dynamic alteration of GSK3ß activities and its role in regulating glial glutamate transporter 1 (GLT-1) protein expression in the spinal dorsal horn and nociceptive behaviors following the nerve injury. Specifically, GSK3ß was expressed in both neurons and astrocytes in the spinal dorsal horn. GSK3ß activities were suppressed on day 3 but increased on day 10 following the nerve injury. In parallel, protein expression of GLT-1 in the spinal dorsal horn was enhanced on day 3 but reduced on day 10. In contrast to these time-dependent changes, the activation of astrocytes and over-production of IL-1ß were found on both day 3 and day 10. Meanwhile, thermal hyperalgesia was observed from day 2 through day 10 and mechanical allodynia from day 4 through day 10. Pre-emptive pharmacological inhibition of GSK3ß activities significantly ameliorated thermal hyperalgesia and mechanical allodynia at the late stage but did not have effects at the early stage. These were accompanied with the suppression of GSK3ß activities, prevention of decreased GLT-1 protein expression, inhibition of astrocytic activation, and reduction of IL-1ß in the spinal dorsal horn on day 10. These data indicate that the increased GSK3ß activity in the spinal dorsal horn is attributable to the downregulation of GLT-1 protein expression in neuropathic rats at the late stage. Further, we also demonstrated that the nerve-injury-induced thermal hyperalgesia on day 10 was transiently suppressed by pharmacological inhibition of GSK3ß. Our study suggests that GSK3ß may be a potential target for the development of analgesics for chronic neuropathic pain.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Regulação da Expressão Gênica/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Ciática/patologia , Medula Espinal/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Modelos Animais de Doenças , Vias de Administração de Medicamentos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Transportador 2 de Aminoácido Excitatório/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Interleucina-1beta/metabolismo , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteína Oncogênica v-akt/metabolismo , Medição da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Serina/metabolismo , Tiazóis/uso terapêutico , Fatores de Tempo , Ureia/análogos & derivados , Ureia/uso terapêutico
9.
Arthritis Rheum ; 64(2): 584-93, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21905019

RESUMO

OBJECTIVE: Fibromyalgia (FM) represents a complex disorder that is characterized by widespread pain and tenderness and is frequently accompanied by additional somatic and cognitive/affective symptoms. Genetic risk factors are known to contribute to the etiology of the syndrome. The aim of this study was to examine >350 genes for association with FM, using a large-scale candidate gene approach. METHODS: The study group comprised 496 patients with FM (cases) and 348 individuals with no chronic pain (controls). Genotyping was performed using a dedicated gene array chip, the Pain Research Panel, which assays variants characterizing >350 genes known to be involved in the biologic pathways relevant to nociception, inflammation, and mood. Association testing was performed using logistic regression. RESULTS: Significant differences in allele frequencies between cases and controls were observed for 3 genes: GABRB3 (rs4906902; P = 3.65 × 10(-6)), TAAR1 (rs8192619; P = 1.11 × 10(-5)), and GBP1 (rs7911; P = 1.06 × 10(-4)). These 3 genes and 7 other genes with suggestive evidence for association were examined in a second, independent cohort of patients with FM and control subjects who were genotyped using the Perlegen 600K platform. Evidence of association in the replication cohort was observed for TAAR1, RGS4, CNR1, and GRIA4. CONCLUSION: Variation in these 4 replicated genes may serve as a basis for development of new diagnostic approaches, and the products of these genes may contribute to the pathophysiology of FM and represent potential targets for therapeutic action.


Assuntos
Fibromialgia/genética , Predisposição Genética para Doença , Polimorfismo de Nucleotídeo Único , Adulto , Idoso , Alelos , Estudos de Casos e Controles , Feminino , Proteínas de Ligação ao GTP/genética , Frequência do Gene , Estudos de Associação Genética , Genótipo , Humanos , Pessoa de Meia-Idade , Receptores Acoplados a Proteínas G/genética , Receptores de GABA-B/genética
10.
J Pain ; 12(11 Suppl): T92-101, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22074755

RESUMO

UNLABELLED: Genetic factors play a role in the etiology of persistent pain conditions, putatively by modulating underlying processes such as nociceptive sensitivity, psychological well-being, inflammation, and autonomic response. However, to date, only a few genes have been associated with temporomandibular disorders (TMD). This study evaluated 358 genes involved in pain processes, comparing allelic frequencies between 166 cases with chronic TMD and 1,442 controls enrolled in the OPPERA (Orofacial Pain: Prospective Evaluation and Risk Assessment) study cooperative agreement. To enhance statistical power, 182 TMD cases and 170 controls from a similar study were included in the analysis. Genotyping was performed using the Pain Research Panel, an Affymetrix gene chip representing 3,295 single nucleotide polymorphisms, including ancestry-informative markers that were used to adjust for population stratification. Adjusted associations between genetic markers and TMD case status were evaluated using logistic regression. The OPPERA findings provided evidence supporting previously reported associations between TMD and 2 genes: HTR2A and COMT. Other genes were revealed as potential new genetic risk factors for TMD, including NR3C1, CAMK4, CHRM2, IFRD1, and GRK5. While these findings need to be replicated in independent cohorts, the genes potentially represent important markers of risk for TMD, and they identify potential targets for therapeutic intervention. PERSPECTIVE: Genetic risk factors for TMD pain were explored in the case-control component of the OPPERA cooperative agreement, a large population-based prospective cohort study. Over 350 candidate pain genes were assessed using a candidate gene panel, with several genes displaying preliminary evidence for association with TMD status.


Assuntos
Dor Crônica/genética , Transtornos da Articulação Temporomandibular/genética , Adolescente , Adulto , Idoso , Estudos de Casos e Controles , Dor Crônica/etiologia , Estudos de Coortes , Feminino , Estudos de Associação Genética/métodos , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Multicêntricos como Assunto/métodos , Estudos Prospectivos , Fatores de Risco , Transtornos da Articulação Temporomandibular/etiologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...