Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 29(44): 14015-25, 2009 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-19890011

RESUMO

Previous studies of peripheral immune cells have documented that activation of adenosine 2A receptors (A(2A)Rs) decrease proinflammatory cytokine release and increase release of the potent anti-inflammatory cytokine, interleukin-10 (IL-10). Given the growing literature supporting that glial proinflammatory cytokines importantly contribute to neuropathic pain and that IL-10 can suppress such pain, we evaluated the effects of intrathecally administered A(2A)R agonists on neuropathic pain using the chronic constriction injury (CCI) model. A single intrathecal injection of the A(2A)R agonists 4-(3-(6-amino-9-(5-cyclopropylcarbamoyl-3,4-dihydroxytetrahydrofuran-2-yl)-9H-purin-2-yl)prop-2-ynyl)piperidine-1-carboxylic acid methyl ester (ATL313) or 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamido adenosine HCl (CGS21680), 10-14 d after CCI versus sham surgery, produced a long-duration reversal of mechanical allodynia and thermal hyperalgesia for at least 4 weeks. Neither drug altered the nociceptive responses of sham-operated controls. An A(2A)R antagonist [ZM241385 (4-(2-[7-amino-2-(2-furyl)(1,2,4)triazolo(2,3-a)(1,3,5)triazin-5-ylamino]ethyl)phenol)] coadministered intrathecally with ATL313 abolished the action of ATL313 in rats with neuropathy-induced allodynia but had no effect on allodynia in the absence of the A(2A)R agonist. ATL313 attenuated CCI-induced upregulation of spinal cord activation markers for microglia and astrocytes in the L4-L6 spinal cord segments both 1 and 4 weeks after a single intrathecal ATL313 administration. Neutralizing IL-10 antibodies administered intrathecally transiently abolished the effect of ATL313 on neuropathic pain. In addition, IL-10 mRNA was significantly elevated in the CSF cells collected from the lumbar region. Activation of A(2A)Rs after intrathecal administration may be a novel, therapeutic approach for the treatment of neuropathic pain by increasing IL-10 in the immunocompetent cells of the CNS.


Assuntos
Agonistas do Receptor A2 de Adenosina , Neuralgia/tratamento farmacológico , Piperidinas/administração & dosagem , Receptor A2A de Adenosina/fisiologia , Animais , Injeções Espinhais , Masculino , Neuralgia/fisiopatologia , Dor/tratamento farmacológico , Dor/fisiopatologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley
2.
Pharm Res ; 27(5): 841-54, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20224990

RESUMO

PURPOSE: Interleukin-10 (IL-10) is an anti-inflammatory molecule that has achieved interest as a therapeutic for neuropathic pain. In this work, the potential of plasmid DNA-encoding IL-10 (pDNA-IL-10) slowly released from biodegradable microparticles to provide long-term pain relief in an animal model of neuropathic pain was investigated. METHODS: PLGA microparticles encapsulating pDNA-IL-10 were developed and assessed both in vitro and in vivo. RESULTS: In vitro, pDNA containing microparticles activated macrophages, enhanced the production of nitric oxide, and increased the production of IL-10 protein relative to levels achieved with unencapsulated pDNA-IL-10. In vivo, intrathecally administered microparticles embedded in meningeal tissue, induced phagocytic cell recruitment to the cerebrospinal fluid, and relieved neuropathic pain for greater than 74 days following a single intrathecal administration, a feat not achieved with unencapsulated pDNA. Therapeutic effects of microparticle-delivered pDNA-IL-10 were blocked in the presence of IL-10-neutralizing antibody, and elevated levels of plasmid-derived IL-10 were detected in tissues for a prolonged time period post-injection (>28 days), demonstrating that therapeutic effects are dependent on IL-10 protein production. CONCLUSIONS: These studies demonstrate that microparticle encapsulation significantly enhances the potency of intrathecally administered pDNA, which may be extended to treat other disorders that require intrathecal gene therapy.


Assuntos
DNA/administração & dosagem , DNA/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Interleucina-10/genética , Doenças do Sistema Nervoso Periférico/terapia , Plasmídeos/genética , Animais , Comportamento Animal/fisiologia , Células Cultivadas , Imuno-Histoquímica , Injeções Espinhais , Interleucina-10/biossíntese , Ácido Láctico , Macrófagos/metabolismo , Masculino , Nanopartículas , Óxido Nítrico/metabolismo , Tamanho da Partícula , Doenças do Sistema Nervoso Periférico/líquido cefalorraquidiano , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
Mol Pain ; 1: 9, 2005 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-15813997

RESUMO

Despite many decades of drug development, effective therapies for neuropathic pain remain elusive. The recent recognition of spinal cord glia and glial pro-inflammatory cytokines as important contributors to neuropathic pain suggests an alternative therapeutic strategy; that is, targeting glial activation or its downstream consequences. While several glial-selective drugs have been successful in controlling neuropathic pain in animal models, none are optimal for human use. Thus the aim of the present studies was to explore a novel approach for controlling neuropathic pain. Here, an adeno-associated viral (serotype II; AAV2) vector was created that encodes the anti-inflammatory cytokine, interleukin-10 (IL-10). This anti-inflammatory cytokine is known to suppress the production of pro-inflammatory cytokines. Upon intrathecal administration, this novel AAV2-IL-10 vector was successful in transiently preventing and reversing neuropathic pain. Intrathecal administration of an AAV2 vector encoding beta-galactosidase revealed that AAV2 preferentially infects meningeal cells surrounding the CSF space. Taken together, these data provide initial support that intrathecal gene therapy to drive the production of IL-10 may prove to be an efficacious treatment for neuropathic pain.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Mediadores da Inflamação/fisiologia , Interleucina-10/biossíntese , Interleucina-10/genética , Nervo Isquiático/fisiopatologia , Ciática/prevenção & controle , Ciática/fisiopatologia , Animais , Dependovirus/fisiologia , Modelos Animais de Doenças , Vetores Genéticos/administração & dosagem , Vetores Genéticos/uso terapêutico , Humanos , Inflamação/metabolismo , Inflamação/prevenção & controle , Inflamação/virologia , Injeções Espinhais , Interleucina-10/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Ciática/metabolismo
4.
Pain ; 115(1-2): 71-83, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15836971

RESUMO

Activated glial cells (microglia and astroglia) in the spinal cord play a major role in mediating enhanced pain states by releasing proinflammatory cytokines and other substances thought to facilitate pain transmission. In the present study, we report that intrathecal administration of minocycline, a selective inhibitor of microglial cell activation, inhibits low threshold mechanical allodynia, as measured by the von Frey test, in two models of pain facilitation. In a rat model of neuropathic pain induced by sciatic nerve inflammation (sciatic inflammatory neuropathy, SIN), minocycline delayed the induction of allodynia in both acute and persistent paradigms. Moreover, minocycline was able to attenuate established SIN-induced allodynia 1 day, but not 1 week later, suggesting a limited role of microglial activation in more perseverative pain states. Our data are consistent with a crucial role for microglial cells in initiating, rather than maintaining, enhanced pain responses. In a model of spinal immune activation by intrathecal HIV-1 gp120, we show that the anti-allodynic effects of minocycline are associated with decreased microglial activation, attenuated mRNA expression of interleukin-1beta (IL-1beta), tumor necrosis factor-alpha (TNF-alpha), IL-1beta-converting enzyme, TNF-alpha-converting enzyme, IL-1 receptor antagonist and IL-10 in lumbar dorsal spinal cord, and reduced IL-1beta and TNF-alpha levels in the CSF. In contrast, no significant effects of minocycline were observed on gp120-induced IL-6 and cyclooxygenase-2 expression in spinal cord or CSF IL-6 levels. Taken together these data highlight the importance of microglial activation in the development of exaggerated pain states.


Assuntos
Citocinas/metabolismo , Hiperestesia/tratamento farmacológico , Hiperestesia/metabolismo , Microglia/metabolismo , Minociclina/administração & dosagem , Doenças do Sistema Nervoso Periférico/metabolismo , Medula Espinal/metabolismo , Animais , Relação Dose-Resposta a Droga , Hiperestesia/imunologia , Injeções Espinhais , Masculino , Microglia/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Doenças do Sistema Nervoso Periférico/imunologia , Ratos , Ratos Sprague-Dawley , Medula Espinal/efeitos dos fármacos
5.
J Pain ; 6(3): 174-83, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15772911

RESUMO

The present series of experiments examined whether the complement cascade might play a key role in the expression of mechanical allodynia. Soluble complement receptor 1 (sCR1) was used to block the activation of the membrane attack pathway of the complement cascade. In doing so, sCR1 prevents the formation of the biologically active end products C3a, C5a, and membrane attack complexes (MACs). Intrathecal sCR1 had no effect on the behavioral responses of control groups. In contrast, blockade of this pathway abolished the expression of mechanical allodynia induced by peripheral nerve inflammation (sciatic inflammatory neuropathy model), partial sciatic nerve injury (chronic constriction injury model), and intrathecal injection of human immunodeficiency virus type 1 gp120, a viral envelope protein that activates glia. The fact that enhanced nociception was prevented or reversed in all 3 paradigms suggests that complement might be broadly involved in spinally mediated pain enhancement. The mechanisms whereby complement activation might potentially affect the functioning of microglia, astrocytes, and neurons are discussed. The complement cascade has not been previously implicated in spinal sensitization. These data suggest that complement activation within the spinal cord might contribute to enhanced pain states and provide additional evidence for immune regulation of pain transmission.


Assuntos
Proteínas do Sistema Complemento/imunologia , Receptores de Complemento , Neuropatia Ciática/imunologia , Neuropatia Ciática/fisiopatologia , Medula Espinal/fisiopatologia , Animais , Doença Crônica , Complemento C3a/imunologia , Complemento C5a/imunologia , Modelos Animais de Doenças , Proteína gp120 do Envelope de HIV/farmacologia , Injeções Espinhais , Masculino , Nociceptores/imunologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Neuropatia Ciática/tratamento farmacológico , Medula Espinal/imunologia
6.
Pain ; 110(1-2): 299-309, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15275780

RESUMO

In inflammatory neuropathy, immune activation near intact peripheral nerves induces mechanical allodynia. The identity of the peripheral immune product(s) that lead to these changes in pain behavior is unknown. The present series of studies utilized the sciatic inflammatory neuropathy (SIN) model to examine this question. Here, inflammatory neuropathy is created by injecting an immune activator (zymosan) around one sciatic nerve via an indwelling catheter. Our prior studies demonstrated that peri-sciatic zymosan activated macrophages and neutrophils to release proinflammatory cytokines and reactive oxygen species (ROS). In addition, zymosan is a classical activator of the complement cascade. Thus the present series of experiments examined whether any of these inflammatory mediators are involved in the initial induction of SIN-induced ipsilateral or bilateral allodynias. Peri-sciatic injection of selective inhibitors/antagonists revealed that a number of immune products are early mediators of the resultant allodynias, including proinflammatory cytokines (tumor necrosis factor, interleukin-1, and interleukin-6), ROS, and complement. Thus these immune-derived substances can markedly alter sensory nerve function at mid-axon.


Assuntos
Proteínas do Sistema Complemento , Citocinas/metabolismo , Neuralgia/imunologia , Espécies Reativas de Oxigênio/metabolismo , Neuropatia Ciática/metabolismo , Animais , Anticorpos/administração & dosagem , Comportamento Animal , Proteínas de Transporte/uso terapêutico , Catalase/administração & dosagem , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Interações Medicamentosas , Lateralidade Funcional , Hiperalgesia/induzido quimicamente , Hiperalgesia/imunologia , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-6/imunologia , Masculino , Neuralgia/tratamento farmacológico , Neuralgia/etiologia , Medição da Dor , Estimulação Física , Ratos , Ratos Sprague-Dawley , Receptores de Complemento/administração & dosagem , Receptores do Fator de Necrose Tumoral/uso terapêutico , Receptores Tipo I de Fatores de Necrose Tumoral , Nervo Isquiático/imunologia , Nervo Isquiático/patologia , Neuropatia Ciática/induzido quimicamente , Neuropatia Ciática/imunologia , Sialoglicoproteínas/administração & dosagem , Superóxido Dismutase/administração & dosagem , Receptores Chamariz do Fator de Necrose Tumoral , Zimosan/toxicidade
7.
J Pain ; 5(7): 392-405, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15501197

RESUMO

UNLABELLED: Glia are now recognized as important contributors in pathological pain creation and maintenance. Spinal cord glia exhibit extensive gap junctional connectivity, raising the possibility that glia are involved in the contralateral spread of excitation resulting in mirror image pain. In the present experiments, the gap junction decoupler carbenoxolone was administered intrathecally after induction of neuropathic pain in response to sciatic nerve inflammation (sciatic inflammatory neuropathy) or partial nerve injury (chronic constriction injury). In both neuropathic pain models, a low dose of carbenoxolone reversed mirror image mechanical allodynia, while leaving ipsilateral mechanical allodynia unaffected. Ipsilateral thermal hyperalgesia was briefly attenuated. Critically, blockade of mechanical allodynia and thermal hyperalgesia was not observed in response to intrathecal glycyrrhizic acid, a compound similar to carbenoxolone in all respects but it does not decouple gap junctions. Thus, blockade of mechanical allodynia and thermal hyperalgesia by carbenoxolone does appear to reflect an effect on gap junctions. Examination of carbenoxolone's effects on intrathecal human immunodeficiency virus type 1 gp120 showed that blockade of pain facilitation might result, at least in part, via suppression of interleukin-1 and, in turn, interleukin-6. These data provide the first suggestion that spread of excitation via gap junctions might contribute importantly to inflammatory and traumatic neuropathic pain. PERSPECTIVE: The current studies provide evidence for involvement of gap junctions in spinal cord pain facilitation. Intrathecal carbenoxolone, a gap junction decoupler, reversed neuropathy-induced mirror image pain and intrathecal gp120-induced allodynia. In addition, it decreased gp120-induced proinflammatory cytokines. This suggests gap junction activation might lead to proinflammatory cytokine release by distantly activated glia.


Assuntos
Junções Comunicantes/fisiologia , Dor/fisiopatologia , Células do Corno Posterior/fisiologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Antiulcerosos/farmacologia , Carbenoxolona/farmacologia , Doença Crônica , Junções Comunicantes/efeitos dos fármacos , Ácido Glicirrízico/farmacologia , Proteína gp120 do Envelope de HIV/farmacologia , Hiperalgesia/fisiopatologia , Injeções Espinhais , Interleucina-1/metabolismo , Masculino , Síndromes de Compressão Nervosa/fisiopatologia , Ratos , Ratos Sprague-Dawley , Neuropatia Ciática/fisiopatologia , Organismos Livres de Patógenos Específicos
8.
J Biomed Mater Res A ; 91(3): 719-29, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19048635

RESUMO

Brain-derived neurotrophic factor (BDNF) was covalently attached to polyethylene glycol (PEG) in order to enhance delivery to the spinal cord via the cerebrospinal fluid (intrathecal administration). By varying reaction conditions, mixtures of BDNF covalently attached to one (primary), two (secondary), three (tertiary), or more (higher order) PEG molecules were produced. The biological activity of each resulting conjugate mixture was assessed with the goal of identifying a relationship between the number of PEG molecules attached to BDNF and biological activity. A high degree of in vitro biological activity was maintained in mixtures enriched in primary and secondary conjugate products, while a substantial reduction in biological activity was observed in mixtures with tertiary and higher order conjugates. When a biologically active mixture of PEG-BDNF was administered intrathecally, it displayed a significantly improved half-life in the cerebrospinal fluid and an enhanced penetration into spinal cord tissue relative to native BDNF. Results from these studies suggest a PEGylation strategy that preserves the biological activity of the protein while also improving the half-life of the protein in vivo. Furthermore, PEGylation may be a promising approach for enhancing intrathecal delivery of therapeutic proteins with potential for treating disease and injury in the spinal cord.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/química , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Neurônios/metabolismo , Polietilenoglicóis/química , Medula Espinal/efeitos dos fármacos , Aldeídos/química , Animais , Sistemas de Liberação de Medicamentos , Ésteres/química , Injeções Espinhais , Masculino , Espectrometria de Massas/métodos , Microscopia Confocal/métodos , Células PC12 , Ratos , Ratos Sprague-Dawley
9.
Brain Behav Immun ; 21(5): 686-98, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17174526

RESUMO

Paclitaxel is a commonly used cancer chemotherapy drug that frequently causes painful peripheral neuropathies. The mechanisms underlying this dose-limiting side effect are poorly understood. Growing evidence supports that proinflammatory cytokines, such as interleukin-1 (IL-1) and tumor necrosis factor (TNF), released by activated spinal glial cells and within the dorsal root ganglia (DRG) are critical in enhancing pain in various animal models of neuropathic pain. Whether these cytokines are involved in paclitaxel-induced neuropathy is unknown. Here, using a rat neuropathic pain model induced by repeated systemic paclitaxel injections, we examined whether paclitaxel upregulates proinflammatory cytokine gene expression, and whether these changes and paclitaxel-induced mechanical allodynia can be attenuated by intrathecal IL-1 receptor antagonist (IL-1ra) or intrathecal delivery of plasmid DNA encoding the anti-inflammatory cytokine, interleukin-10 (IL-10). The data show that paclitaxel treatment induces mRNA expression of IL-1, TNF, and immune cell markers in lumbar DRG. Intrathecal IL-1ra reversed paclitaxel-induced allodynia and intrathecal IL-10 gene therapy both prevented, and progressively reversed, this allodynic state. Moreover, IL-10 gene therapy resulted in increased IL-10 mRNA levels in lumbar DRG and meninges, measured 2 weeks after initiation of therapy, whereas paclitaxel-induced expression of IL-1, TNF, and CD11b mRNA in lumbar DRG was markedly decreased. Taken together, these data support that paclitaxel-induced neuropathic pain is mediated by proinflammatory cytokines, possibly released by activated immune cells in the DRG. We propose that targeting the production of proinflammatory cytokines by intrathecal IL-10 gene therapy may be a promising therapeutic strategy for the relief of paclitaxel-induced neuropathic pain.


Assuntos
Antineoplásicos Fitogênicos/efeitos adversos , Gânglios Espinais/efeitos dos fármacos , Hiperalgesia/prevenção & controle , Interleucina-10/fisiologia , Paclitaxel/efeitos adversos , Doenças do Sistema Nervoso Periférico/prevenção & controle , Animais , Antígeno CD11b/efeitos dos fármacos , Antígeno CD11b/metabolismo , Citocinas/efeitos dos fármacos , Citocinas/imunologia , Modelos Animais de Doenças , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Terapia Genética/métodos , Hiperalgesia/induzido quimicamente , Hiperalgesia/etiologia , Injeções Espinhais , Interleucina-10/administração & dosagem , Interleucina-10/genética , Interleucina-1beta/efeitos dos fármacos , Interleucina-1beta/metabolismo , Masculino , Meninges/efeitos dos fármacos , Meninges/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/complicações , Plasmídeos/administração & dosagem , Plasmídeos/genética , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores de Interleucina-1/antagonistas & inibidores , Receptores de Interleucina-1/fisiologia , Medula Espinal/citologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
10.
Neuron Glia Biol ; 2(4): 293-308, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18079973

RESUMO

Research on communication between glia and neurons has increased in the past decade. The onset of neuropathic pain, a major clinical problem that is not resolved by available therapeutics, involves activation of spinal cord glia through the release of proinflammatory cytokines in acute animal models of neuropathic pain. Here, we demonstrate for the first time that the spinal action of the proinflammatory cytokine, interleukin 1 (IL-1) is involved in maintaining persistent (2 months) allodynia induced by chronic-constriction injury (CCI). The anti-inflammatory cytokine IL-10 can suppress proinflammatory cytokines and spinal cord glial amplification of pain. Given that IL-1 is a key mediator of neuropathic pain, developing a clinically viable means of long-term delivery of IL-10 to the spinal cord is desirable. High doses of intrathecal IL-10-gene therapy using naked plasmid DNA (free pDNA-IL-10) is effective, but the dose required limits its potential clinical utility. Here we show that intrathecal gene therapy for neuropathic pain is improved sufficiently using two, distinct synthetic polymers, poly(lactic-co-glycolic) and polyethylenimine, that substantially lower doses of pDNA-IL-10 are effective. In conclusion, synthetic polymers used as i.t. gene-delivery systems are well-tolerated and improve the long-duration efficacy of pDNA-IL-10 gene therapy.

11.
Pain ; 126(1-3): 294-308, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16949747

RESUMO

Neuropathic pain is a major clinical problem unresolved by available therapeutics. Spinal cord glia play a pivotal role in neuropathic pain, via the release of proinflammatory cytokines. Anti-inflammatory cytokines, like interleukin-10 (IL-10), suppress proinflammatory cytokines. Thus, IL-10 may provide a means for controlling glial amplification of pain. We recently documented that intrathecal IL-10 protein resolves neuropathic pain, albeit briefly (approximately 2-3 h), given its short half-life. Intrathecal gene therapy using viruses encoding IL-10 can also resolve neuropathic pain, but for only approximately 2 weeks. Here, we report a novel approach that dramatically increases the efficacy of intrathecal IL-10 gene therapy. Repeated intrathecal delivery of plasmid DNA vectors encoding IL-10 (pDNA-IL-10) abolished neuropathic pain for greater than 40 days. Naked pDNA-IL-10 reversed chronic constriction injury (CCI)-induced allodynia both shortly after nerve injury as well as 2 months later. This supports that spinal proinflammatory cytokines are important in both the initiation and maintenance of neuropathic pain. Importantly, pDNA-IL-10 gene therapy reversed mechanical allodynia induced by CCI, returning rats to normal pain responsiveness, without additional analgesia. Together, these data suggest that intrathecal IL-10 gene therapy may provide a novel approach for prolonged clinical pain control.


Assuntos
DNA/administração & dosagem , Terapia Genética , Interleucina-10/genética , Neuralgia/fisiopatologia , Neuralgia/terapia , Plasmídeos/administração & dosagem , Animais , DNA/líquido cefalorraquidiano , DNA/farmacocinética , DNA/uso terapêutico , Esquema de Medicação , Humanos , Hiperestesia/etiologia , Hiperestesia/fisiopatologia , Hiperestesia/terapia , Injeções Espinhais , Ligadura , Masculino , Microinjeções , Plasmídeos/líquido cefalorraquidiano , Plasmídeos/farmacocinética , Plasmídeos/uso terapêutico , Ratos , Ratos Sprague-Dawley , Nervo Isquiático , Medula Espinal/metabolismo , Fatores de Tempo , Distribuição Tecidual
12.
Eur J Neurosci ; 21(8): 2136-48, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15869510

RESUMO

Gene therapy for the control of pain has, to date, targeted neurons. However, recent evidence supports that spinal cord glia are critical to the creation and maintenance of pain facilitation through the release of proinflammatory cytokines. Because of the ability of interleukin-10 (IL-10) to suppress proinflammatory cytokines, we tested whether an adenoviral vector encoding human IL-10 (AD-h-IL10) would block and reverse pain facilitation. Three pain models were examined, all of which are mediated by spinal pro-inflammatory cytokines. Acute intrathecal administration of rat IL-10 protein itself briefly reversed chronic constriction injury-induced mechanical allodynia and thermal hyperalgesia. The transient reversal caused by IL-10 protein paralleled the half-life of human IL-10 protein in the intrathecal space (t(1/2) approximately 2 h). IL-10 gene therapy both prevented and reversed thermal hyperalgesia and mechanical allodynia, without affecting basal responses to thermal or mechanical stimuli. Extra-territorial, as well as territorial, pain changes were reversed by this treatment. Intrathecal AD-h-IL10 injected over lumbosacral spinal cord led to elevated lumbosacral cerebrospinal fluid (CSF) levels of human IL-10, with far less human IL-10 observed in cervical CSF. In keeping with IL-10's known anti-inflammatory actions, AD-h-IL10 lowered CSF levels of IL-1, relative to control AD. These studies support that this gene therapy approach provides an alternative to neuronally focused drug and gene therapies for clinical pain control.


Assuntos
Terapia Genética/métodos , Interleucina-10/uso terapêutico , Manejo da Dor , Adenoviridae/genética , Animais , Comportamento Animal , Relação Dose-Resposta a Droga , Lateralidade Funcional/fisiologia , Vetores Genéticos , Membro Posterior/efeitos dos fármacos , Membro Posterior/inervação , Membro Posterior/fisiopatologia , Humanos , Injeções Espinhais/métodos , Interleucina-1/biossíntese , Interleucina-1/uso terapêutico , Interleucina-10/biossíntese , Interleucina-10/líquido cefalorraquidiano , Interleucina-10/genética , Masculino , Microinjeções/métodos , Dor/classificação , Dor/etiologia , Medição da Dor/métodos , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Interleucina-1/administração & dosagem , Receptores Tipo I de Interleucina-1 , Fatores de Tempo , Zimosan/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa