Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
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 Exp Med ; 215(5): 1301-1313, 2018 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-29703731

RESUMO

The development of chemotherapy-induced painful peripheral neuropathy is a major dose-limiting side effect of many chemotherapeutics, including bortezomib, but the mechanisms remain poorly understood. We now report that bortezomib causes the dysregulation of de novo sphingolipid metabolism in the spinal cord dorsal horn to increase the levels of sphingosine-1-phosphate (S1P) receptor 1 (S1PR1) ligands, S1P and dihydro-S1P. Accordingly, genetic and pharmacological disruption of S1PR1 with multiple S1PR1 antagonists, including FTY720, blocked and reversed neuropathic pain. Mice with astrocyte-specific alterations of S1pr1 did not develop neuropathic pain and lost their ability to respond to S1PR1 inhibition, strongly implicating astrocytes as a primary cellular substrate for S1PR1 activity. At the molecular level, S1PR1 engaged astrocyte-driven neuroinflammation and altered glutamatergic homeostasis, processes blocked by S1PR1 antagonism. Our findings establish S1PR1 as a target for therapeutic intervention and provide insight into cellular and molecular pathways. As FTY720 also shows promising anticancer potential and is FDA approved, rapid clinical translation of our findings is anticipated.


Assuntos
Bortezomib/efeitos adversos , Neuralgia/induzido quimicamente , Neuralgia/metabolismo , Esfingolipídeos/metabolismo , Administração Oral , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Ceramidas/biossíntese , Cloridrato de Fingolimode/administração & dosagem , Cloridrato de Fingolimode/farmacologia , Glutamatos/metabolismo , Masculino , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Ratos Sprague-Dawley , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia
4.
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
5.
Mol Pharmacol ; 91(3): 197-207, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28031332

RESUMO

RGS10 has emerged as a key regulator of proinflammatory cytokine production in microglia, functioning as an important neuroprotective factor. Although RGS10 is normally expressed in microglia at high levels, expression is silenced in vitro following activation of TLR4 receptor. Given the ability of RGS10 to regulate inflammatory signaling, dynamic regulation of RGS10 levels in microglia may be an important mechanism to tune inflammatory responses. The goals of the current study were to confirm that RGS10 is suppressed in an in vivo inflammatory model of microglial activation and to determine the mechanism for activation-dependent silencing of Rgs10 expression in microglia. We demonstrate that endogenous RGS10 is present in spinal cord microglia, and RGS10 protein levels are suppressed in the spinal cord in a nerve injury-induced neuropathic pain mouse model. We show that the histone deacetylase (HDAC) enzyme inhibitor trichostatin A blocks the ability of lipopolysaccharide (LPS) to suppress Rgs10 transcription in BV-2 and primary microglia, demonstrating that HDAC enzymes are required for LPS silencing of Rgs10 Furthermore, we used chromatin immunoprecipitation to demonstrate that H3 histones at the Rgs10 proximal promoter are deacetylated in BV-2 microglia following LPS activation, and HDAC1 association at the Rgs10 promoter is enhanced following LPS stimulation. Finally, we have shown that sphingosine 1-phosphate, an endogenous microglial signaling mediator that inhibits HDAC activity, enhances basal Rgs10 expression in BV-2 microglia, suggesting that Rgs10 expression is dynamically regulated in microglia in response to multiple signals.


Assuntos
Inativação Gênica , Histona Desacetilases/metabolismo , Microglia/metabolismo , Proteínas RGS/genética , Transcrição Gênica , Acetilação/efeitos dos fármacos , Animais , Azacitidina/farmacologia , Linhagem Celular , Quimiocina CXCL2/metabolismo , Modelos Animais de Doenças , Inativação Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Inflamação/patologia , Lipopolissacarídeos/farmacologia , Lisofosfolipídeos/farmacologia , Metiltransferases/metabolismo , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética , Proteínas RGS/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
6.
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
7.
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
8.
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
9.
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
10.
J Neuroinflammation ; 12: 222, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25571780

RESUMO

Toll like receptor 4 (TLR4) is an innate immune pattern recognition receptor, expressed predominantly on microglia in the CNS. Activation of spinal TLR4 plays a critical role in the genesis of pathological pain induced by nerve injury, bone cancer, and tissue inflammation. Currently, it remains unknown how synaptic activities in the spinal dorsal horn are regulated by TLR4 receptors. Through recording GABAergic currents in neurons and glial glutamate transporter currents in astrocytes in rodent spinal slices, we determined whether and how TLR4 modulates GABAergic synaptic activities in the superficial spinal dorsal horn. We found that activation of TLR4 by lipopolysaccharide (LPS) reduces GABAergic synaptic activities through both presynaptic and postsynaptic mechanisms. Specifically, LPS causes the release of IL-1ß from microglia. IL-1ß in turn suppresses GABA receptor activities at the postsynaptic site through activating protein kinase C (PKC) in neurons. GABA synthesis at the presynaptic site is reduced upon activation of TLR4. Glial glutamate transporter activities are suppressed by IL-1ß and PKC activation induced by LPS. The suppression of glial glutamate transporter activities leads to a deficiency of glutamine supply, which results in an attenuation of the glutamate-glutamine cycle-dependent GABA synthesis. These findings shed light on understanding synaptic plasticity induced by activation of TLR4 under neuroinflammation and identify GABA receptors, glial glutamate transporters, IL-1ß and PKC as therapeutic targets to abrogate abnormal neuronal activities following activation of TLR4 in pathological pain conditions.


Assuntos
Interleucina-1beta/metabolismo , Receptores de GABA/metabolismo , Corno Dorsal da Medula Espinal , Receptor 4 Toll-Like/metabolismo , Ácido gama-Aminobutírico/metabolismo , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Inibidores Enzimáticos/farmacologia , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , GABAérgicos/farmacologia , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Minociclina/farmacologia , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Corno Dorsal da Medula Espinal/citologia , Corno Dorsal da Medula Espinal/efeitos dos fármacos , Corno Dorsal da Medula Espinal/metabolismo
11.
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
12.
Rapid Commun Mass Spectrom ; 27(19): 2127-34, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23996385

RESUMO

RATIONALE: Paclitaxel, an antitumor agent for the treatment of several types of cancers, has recently been reported to cause impaired cognitive function and neuropathic pain in humans. To assess the effects of paclitaxel on the central nervous system, a sensitive and accurate method is required to quantify paclitaxel concentrations in plasma and brain tissue obtained from rodents receiving paclitaxel. METHODS: The biological samples were prepared by liquid-liquid extraction and separated by a 3.5 min reversed-phase liquid chromatography (RPLC) method using a BDS Hypersil C8 column under isocratic conditions. Paclitaxel was quantified using multiple reaction monitoring (MRM) with a triple quadrupole tandem mass spectrometer working in the positive electrospray ionization (ESI+) mode. A stable isotope labeled analogue of paclitaxel was used as the internal standard (IS). RESULTS: The method was validated to be precise and accurate within the dynamic range of 0.5-100 ng/mL based on 100 µL plasma and 1.5-300 ng/g based on 33 mg of brain tissue in homogenate. This method was applied to samples from 2 mg/kg intravenously dosed rats. The plasma concentrations were observed to be 26.62 ± 8.93 ng/mL and brain concentrations 11.08 ± 4.18 ng/g when measured 4 h post-dose. CONCLUSIONS: This rapid LC/MS/MS method was validated to be sensitive, specific, precise and accurate for the quantification of paclitaxel in rat plasma and brain tissue homogenate. Application of the method to study samples provided sufficient proof of blood-brain barrier penetration of paclitaxel, allowing further investigation of its influence on the central nervous system.


Assuntos
Química Encefálica , Cromatografia Líquida de Alta Pressão/métodos , Paclitaxel/análise , Espectrometria de Massas em Tandem/métodos , Animais , Estabilidade de Medicamentos , Modelos Lineares , Paclitaxel/sangue , Paclitaxel/química , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
13.
Brain Res ; 1230: 91-8, 2008 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-18657527

RESUMO

Neuropathy is the chief dose-limiting side effect associated with the major classes of frontline cancer therapy drugs. Here the changes in behavioral responses of rats to cutaneous mechanical and thermal stimuli occurring following treatment with cisplatin and the changes in spinal neurophysiology accompanying the development of chemotherapy-induced hyperalgesia were explored. Systemic treatment with cisplatin induced changes in both mechanical and thermal cutaneous sensory withdrawal thresholds of Sprague-Dawley rats. High doses of chemotherapy produced hypoalgesia whereas lower doses produced hyperalgesia. Follow-up neurophysiological studies in rats with chemotherapy-induced hyperalgesia revealed that deep spinal lamina wide dynamic range neurons had significantly higher spontaneous activity and longer afterdischarges to noxious mechanical stimuli than wide dynamic range neurons in control rats; cisplatin administration was also associated with longer afterdischarges and abnormal wind-up to transcutaneous electrical stimuli. The hyperexcitability observed during cisplatin-induced hyperalgesia is very similar to that observed in rats with hyperalgesia produced following treatment with other very diverse types of chemotherapeutic agents and similar to that observed following specific types of direct nerve injury.


Assuntos
Antineoplásicos/toxicidade , Comportamento Animal/efeitos dos fármacos , Cisplatino/toxicidade , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/psicologia , Animais , Peso Corporal/efeitos dos fármacos , Estimulação Elétrica , Eletrofisiologia , Temperatura Alta , Hiperalgesia/induzido quimicamente , Hiperalgesia/patologia , Masculino , Limiar da Dor/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/patologia , Estimulação Física , Células do Corno Posterior/efeitos dos fármacos , Células do Corno Posterior/patologia , Ratos , Ratos Sprague-Dawley
14.
Brain Res ; 1229: 100-10, 2008 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-18652810

RESUMO

Chemotherapy-induced pain is the most common treatment-limiting complication encountered by cancer patients receiving taxane-, vinca alkaloid- or platin-based chemotherapy. Several lines of evidence indicate that activation of pro-inflammatory cascades involving the release of cytokines including tumor necrosis factor-alpha (TNF-alpha), interleukin-1 beta (IL-1beta) and interleukin-6 (IL-6) as well as various growth factors are key events in the pathogenesis of many types of nerve-injury related pain. Similar mechanisms might also be involved in the etiology of chemotherapy-induced pain. Thalidomide and minocycline have profound immunomodulatory actions in addition to their originally intended pharmacological actions. These compounds were evaluated here for effects in preventing the development of taxol-induced mechanical and thermal hyperalgesia in rats. Thalidomide (50.0 mg/kg) reduced taxol-induced mechanical allodynia and hyperalgesia whereas minocycline (20.0 mg/kg) reduced taxol-induced mechanical hyperalgesia and allodynia as well as taxol-induced thermal hyperalgesia. These results suggest that immunomodulatory agents may provide a treatment option for the protection or reversal of chemotherapy-related pain.


Assuntos
Antibacterianos/uso terapêutico , Hiperalgesia/tratamento farmacológico , Imunossupressores/uso terapêutico , Minociclina/uso terapêutico , Talidomida/uso terapêutico , Animais , Comportamento Animal/efeitos dos fármacos , Hiperalgesia/induzido quimicamente , Hiperalgesia/classificação , Locomoção/efeitos dos fármacos , Masculino , Paclitaxel , Medição da Dor , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Teste de Desempenho do Rota-Rod/métodos , Estatísticas não Paramétricas
15.
Ren Fail ; 29(4): 399-407, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17497460

RESUMO

Recently, we demonstrated that furosemide improves renal hemodynamics and attenuates ischemia/reperfusion (I/R)-associated changes in angiogenesis-related gene expression. However, the effect of furosemide on I/R-induced apoptosis is not known. We utilized a rat model of acute ischemic nephropathy to test the hypothesis that furosemide attenuates I/R-induced apoptosis. Male Sprague-Dawley rats anesthetized with urethane (50 mg/kg) were randomly allocated into four groups (n = 6 each): sham operated saline infusion, sham operated with furosemide (30 microg/kg/hr) infusion, unilateral renal ischemia (1 hr) followed by six hours of reperfusion, and I/R with furosemide infusion. Apoptosis was measured in kidney samples and compared between groups using ANOVA with Bonferroni correction. Apoptosis-related gene expression was assessed using microarray analysis and validated with RT-PCR. Phosphorylation of Akt was analyzed using ELISA, and data were compared between groups using the Mann Whitney U test. Compared to the control group, I/R significantly (p < 0.001) induced apoptosis in both the cortex and medulla. Similarly, microarray analysis revealed that I/R induced (< or = two-fold increase compared to control group) 73 apoptosis-related genes. Phosphorylation of Akt was significantly (p < 0.05) downregulated after I/R. Treatment with furosemide significantly (p < 0.001) reduced I/R-induced apoptosis in both the cortex and medulla and attenuated the expression of 72 I/R-induced apoptosis-related genes. Compared to the I/R group, furosemide significantly (p < 0.01) upregulated the phosphorylation of Akt. These data suggest that a low dose furosemide infusion may attenuate I/R-induced apoptosis and associated gene transcription, and imply a possible novel molecular basis for the mechanism of action of furosemide in acute renal failure.


Assuntos
Injúria Renal Aguda/fisiopatologia , Apoptose/efeitos dos fármacos , Diuréticos/farmacologia , Furosemida/farmacologia , Injúria Renal Aguda/genética , Animais , Fragmentação do DNA , Modelos Animais de Doenças , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/fisiopatologia
16.
J Pain Symptom Manage ; 33(2): 166-79, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17280922

RESUMO

Vincristine is one of the frontline chemotherapy drugs for the treatment of numerous lymphoid neoplasias. The main dose-limiting complication of vincristine is the development of painful peripheral neuropathy. Although clinical reports have appeared in the literature detailing the symptoms of vincristine neuropathy, quantitative sensory testing data that might yield insight to dysfunction in subsets of primary afferents are lacking. In this report, pain descriptors and anatomical distributions of sensory abnormalities were collected in each patient. Touch detection threshold, sharpness detection threshold, the thresholds for the detection of skin warming, heat pain, skin cooling, and the perception of cooling-induced pain were measured in patients with chronic vincristine-induced pain in each area of sensory abnormality and in skin perceived as outside the affected areas. Elevated touch detection thresholds were observed both within and outside areas affected by pain and sensory abnormality. Elevated sharpness and warm detection thresholds were noted only in areas affected by pain. These data suggest that chronic vincristine-induced pain is associated with dysfunction in Abeta, Adelta, and C caliber primary afferent fibers. Deficits in Abeta fibers appear to precede and presage deficits in the other fiber types, whereas deficits in Adelta- and C-fiber function appear to be specifically associated with the generation of pain.


Assuntos
Antineoplásicos Fitogênicos/efeitos adversos , Neoplasias/tratamento farmacológico , Dor/induzido quimicamente , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Vincristina/efeitos adversos , Adulto , Vias Aferentes/efeitos dos fármacos , Idoso , Doença Crônica , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Dor/diagnóstico , Medição da Dor , Doenças do Sistema Nervoso Periférico/diagnóstico
17.
J Pain ; 8(4): 296-306, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17175202

RESUMO

UNLABELLED: Bortezomib (PS-341) is a newly developed proteosome inhibitor that shows extremely promising antineoplastic effects against a variety of neoplasias. Neuropathic pain is emerging as a major complication of bortezomib. Although clinical reports have appeared in the literature describing the general symptoms of bortezomib chemoneuropathy, specific quantitative sensory data that detail the sensory deficits that might yield insight to the primary afferent dysfunction contributing to this pain is lacking. In this report, it is shown that patients with bortezomib-induced neuropathic pain have significantly elevated touch detection threshold and slotted peg board time, impaired sharpness detection, and elevated thresholds for the detection of skin warming and heat pain. Patients also had increased reports of cold pain. These data indicate that bortezomib-induced neuropathy is associated with deficits in Abeta, Adelta, and C caliber primary afferent fibers. PERSPECTIVE: This work demonstrates that pain induced by the chemotherapy drug bortezomib is accompanied by dysfunction in all fiber types in sensory nerves. Impaired Abeta and C sensory function also extends into areas of skin that are not perceived as affected by pain.


Assuntos
Antineoplásicos/efeitos adversos , Ácidos Borônicos/efeitos adversos , Dor/induzido quimicamente , Dor/fisiopatologia , Pirazinas/efeitos adversos , Idoso , Antineoplásicos/uso terapêutico , Ácidos Borônicos/uso terapêutico , Bortezomib , Doença Crônica , Temperatura Baixa , Feminino , Temperatura Alta , Humanos , Linfoma de Células B/complicações , Linfoma de Células B/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/complicações , Mieloma Múltiplo/tratamento farmacológico , Medição da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Desempenho Psicomotor/efeitos dos fármacos , Desempenho Psicomotor/fisiologia , Pirazinas/uso terapêutico , Sensação/efeitos dos fármacos , Sensação/fisiologia , Transtornos de Sensação/induzido quimicamente , Transtornos de Sensação/fisiopatologia , Sensação Térmica/efeitos dos fármacos , Tato/efeitos dos fármacos
18.
Neurosci Lett ; 386(1): 18-22, 2005 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-15975716

RESUMO

Changes in the expression of glial glutamate transporters (GLAST and GLT-1) were examined in the spinal cord of rats with chemotherapy (taxol)-induced mechanical hyperalgesia. Immunohistochemical studies show that the expression of both GLAST and GLT-1 in the L4-L5 spinal dorsal horn is decreased by 24% (P<0.001) and 23% (P<0.001), respectively, in rats with taxol-induced hyperalgesia as compared with those in control rats. These changes were further confirmed using an enzyme-linked immunosorbent assay that confirmed downregulation of GLAST by 36% (P<0.05) and GLT-1 by 18% (P<0.05) in the L4-L5 spinal cord of taxol-treated rats. These data indicate that downregulation of glutamate transporters may contribute to the development of hyperalgesia induced by taxol and suggest that glutamate transporters may be a new target for treatment of pain.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Hiperalgesia/metabolismo , Paclitaxel , Doenças do Sistema Nervoso Periférico/metabolismo , Células do Corno Posterior/metabolismo , Simportadores/metabolismo , Sistema X-AG de Transporte de Aminoácidos/efeitos dos fármacos , Animais , Antineoplásicos Fitogênicos , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/fisiologia , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Transportador 1 de Aminoácido Excitatório , Transportador 2 de Aminoácido Excitatório/efeitos dos fármacos , Proteínas de Transporte de Glutamato da Membrana Plasmática , Ácido Glutâmico/metabolismo , Hiperalgesia/induzido quimicamente , Hiperalgesia/fisiopatologia , Masculino , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Nociceptores/efeitos dos fármacos , Nociceptores/metabolismo , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/fisiopatologia , Células do Corno Posterior/efeitos dos fármacos , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Ratos , Ratos Sprague-Dawley , Simportadores/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
19.
Pain ; 109(1-2): 132-42, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15082135

RESUMO

Taxol produces neuropathic pain with three distinct zones of involvement in the extremities. Most distally is an area of on-going pain and proximal to this is a zone of sensory disturbance but not overt pain. These two areas were confined in all but one case to the glabrous skin of the hands and/or feet. More proximal is an area not recognized by the patients as involved with pain or sensory disturbance yet wherein quantitative sensory tests nevertheless reveal altered sensibility. Impairment of perception to light touch, normally conveyed by myelinated fibers, was dramatically altered in all three areas, being approximately 50-fold greater than normal in areas of pain and sensory disturbance as well as in areas of skin perceived by the patients as not affected. Impairment of perception to sharpness, normally conveyed by small myelinated fibers, was most pronounced in areas of on-going pain, intermediate in areas of sensory disturbance and near baseline in more proximal skin of chemotherapy patients. In contrast to mechanical sensibility, thermal thresholds for warm and heat pain detection were normal throughout. Finally, chemotherapy patients showed paradoxical burning pain to skin cooling that was most pronounced in proximal areas of skin thought to be unaffected by the patients, intermediate in the border zone of altered sensibility and least pronounced in areas of on-going pain. These data suggest that taxol produces a neuropathy characterized by pronounced impairment of function in A-beta myelinated fibers, intermediate impairment of A-delta myelinated fibers, and a relative sparing of C-fibers.


Assuntos
Fibras Nervosas Mielinizadas/efeitos dos fármacos , Nociceptores/fisiopatologia , Paclitaxel/efeitos adversos , Transtornos de Sensação/induzido quimicamente , Limiar Sensorial/efeitos dos fármacos , Adulto , Idoso , Antineoplásicos Fitogênicos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/tratamento farmacológico , Fibras Nervosas Mielinizadas/fisiologia , Paclitaxel/uso terapêutico , Dor/etiologia , Dor/fisiopatologia , Limiar da Dor/efeitos dos fármacos , Desempenho Psicomotor/efeitos dos fármacos , Pele/inervação , Fatores de Tempo
20.
J Pain Symptom Manage ; 27(1): 72-8, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14711471

RESUMO

We present two patients with chemotherapy-induced painful neuropathy that had been poorly controlled with medications but successfully treated with spinal cord stimulation (SCS). A trial period of SCS provided effective pain relief in both patients who subsequently underwent permanent stimulator implantation. Psychophysical tests were performed before and after the implantation of trial and permanent stimulators. SCS improved pain scores and facilitated a reduction of medications. Both patients reported improved gait and one of them also reported an increase in leg flexibility. Psychophysical tests demonstrated an improvement in touch and sharpness detection thresholds. In summary, SCS offers a therapeutic option for patients with chemotherapy-induced peripheral neuropathy who have poor pain relief with standard medical treatment.


Assuntos
Antineoplásicos/efeitos adversos , Terapia por Estimulação Elétrica , Manejo da Dor , Dor/induzido quimicamente , Medula Espinal , Idoso , Humanos , Masculino , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA