Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Genes Brain Behav ; 23(2): e12894, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38597363

RESUMEN

Opioid use disorder (OUD) is an ongoing public health concern in the United States, and relatively little work has addressed how genetic background contributes to OUD. Understanding the genetic contributions to oxycodone-induced analgesia could provide insight into the early stages of OUD development. Here, we present findings from a behavioral phenotyping protocol using several inbred strains from the Hybrid Rat Diversity Panel. Our behavioral protocol included a modified "up-down" von Frey procedure to measure inherent strain differences in the sensitivity to a mechanical stimulus on the hindpaw. We also performed the tail immersion assay, which measures the latency to display tail withdrawal in response to a hot water bath. Initial withdrawal thresholds were taken in drug-naïve animals to record baseline thermal sensitivity across the strains. Oxycodone-induced analgesia was measured after administration of oxycodone over the course of 2 h. Both mechanical and thermal sensitivity are shaped by genetic factors and display moderate heritability (h2 = 0.23-0.40). All strains displayed oxycodone-induced analgesia that peaked at 15-30 min and returned to baseline by 2 h. There were significant differences between the strains in the magnitude and duration of their analgesic response to oxycodone, although the heritability estimates were quite modest (h2 = 0.10-0.15). These data demonstrate that genetic background confers differences in mechanical sensitivity, thermal sensitivity, and oxycodone-induced analgesia.


Asunto(s)
Analgesia , Trastornos Relacionados con Opioides , Ratas , Animales , Oxicodona/farmacología , Analgésicos Opioides/farmacología
2.
Brain Behav Immun ; 115: 157-168, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37838078

RESUMEN

Females represent a majority of chronic pain patients and show greater inflammatory immune responses in human chronic pain patient populations as well as in animal models of neuropathic pain. Recent discoveries in chronic pain research have revealed sex differences in inflammatory signaling, a key component of sensory pathology in chronic neuropathic pain, inviting more research into the nuances of these sex differences. Here we use the chronic constriction injury (CCI) model to explore similarities and differences in expression and production of Inflammatory cytokine IL-1beta in the lumbar spinal cord, as well as its role in chronic pain. We have discovered that intrathecal IL-1 receptor antagonist reverses established pain in both sexes, and increased gene expression of inflammasome NLRP3 is specific to microglia and astrocytes rather than neurons, while IL-1beta is specific to microglia in both sexes. We report several sex differences in the expression level of the genes coding for IL-1beta, as well as the four inflammasomes responsible for IL-1beta release: NLRP3, AIM2, NLRP1, and NLRC4 in the spinal cord. Total mRNA, but not protein expression of IL-1beta is greater in females than males after CCI. Also, while CCI increases all four inflammasomes in both sexes, there are sex differences in relative levels of inflammasome expression. NLRP3 and AIM2 are more highly expressed in females, whereas NLRP1 expression is greater in males.


Asunto(s)
Dolor Crónico , Inflamasomas , Interleucina-1beta , Neuralgia , Animales , Femenino , Humanos , Masculino , Ratas , Dolor Crónico/metabolismo , Constricción , Proteínas de Unión al ADN/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Neuralgia/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Médula Espinal/metabolismo
3.
Front Pain Res (Lausanne) ; 3: 932530, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36176709

RESUMEN

Up to 92% of patients suffering from multiple sclerosis (MS) experience pain, most without adequate treatment, and many report pain long before motor symptoms associated with MS diagnosis. In the most commonly studied rodent model of MS, experimental autoimmune encephalomyelitis (EAE), motor impairments/disabilities caused by EAE can interfere with pain testing. In this study, we characterize a novel low-dose myelin-oligodendrocyte-glycoprotein (MOG)-induced Sprague-Dawley (SD) model of EAE-related pain in male rats, optimized to minimize motor impairments/disabilities. Adult male SD rats were treated with increasing doses of intradermal myelin-oligodendrocyte-glycoprotein (MOG1-125) (0, 4, 8, and 16 µg) in incomplete Freund's adjuvant (IFA) vehicle to induce mild EAE. Von Frey testing and motor assessments were conducted prior to EAE induction and then weekly thereafter to assess EAE-induced pain and motor impairment. Results from these studies demonstrated that doses of 8 and 16 µg MOG1-125 were sufficient to produce stable mechanical allodynia for up to 1 month in the absence of hindpaw motor impairments/disabilities. In the follow-up studies, these doses of MOG1-125, were administered to create allodynia in the absence of confounded motor impairments. Then, 2 weeks later, rats began daily subcutaneous injections of the Toll-like receptor 2 and 4 (TLR2-TLR4) antagonist (+)-naltrexone [(+)-NTX] or saline for an additional 13 days. We found that (+)-NTX also reverses EAE-induced mechanical allodynia in the MOG-induced SD rat model of EAE, supporting parallels between models, but now allowing a protracted timecourse to be examined completely free of motor confounds. Exploring further mechanisms, we demonstrated that both spinal NOD-like receptor protein 3 (NLRP3) and interleukin-17 (IL-17) are necessary for EAE-induced pain, as intrathecal injections of NLRP3 antagonist MCC950 and IL-17 neutralizing antibody both acutely reversed EAE-induced pain. Finally, we show that spinal glial immunoreactivity induced by EAE is reversed by (+)-NTX, and that spinal demyelination correlates with the severity of motor impairments/disabilities. These findings characterize an optimized MOG-induced SD rat model of EAE for the study of pain with minimal motor impairments/disabilities. Finally, these studies support the role of TLR2-TLR4 antagonists as a potential treatment for MS-related pain and other pain and inflammatory-related disorders.

4.
Pain ; 163(10): 1939-1951, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35486864

RESUMEN

ABSTRACT: Animal and human studies have shown that exercise prior to nerve injury prevents later chronic pain, but the mechanisms of such preconditioning remain elusive. Given that exercise acutely increases the formation of free radicals, triggering antioxidant compensation, we hypothesized that voluntary running preconditioning would attenuate neuropathic pain by supporting redox homeostasis after sciatic nerve injury in male and female rats. We show that 6 weeks of voluntary wheel running suppresses neuropathic pain development induced by chronic constriction injury across both sexes. This attenuation was associated with reduced nitrotyrosine immunoreactivity-a marker for peroxynitrite-at the sciatic nerve injury site. Our data suggest that prior voluntary wheel running does not reduce the production of peroxynitrite precursors, as expression levels of inducible nitric oxide synthase and NADPH oxidase 2 were unchanged. Instead, voluntary wheel running increased superoxide scavenging by elevating expression of superoxide dismutases 1 and 2. Prevention of neuropathic pain was further associated with the activation of the master transcriptional regulator of the antioxidant response, nuclear factor E2-related factor 2 (Nrf2). Six weeks of prior voluntary wheel running increased Nrf2 nuclear translocation at the sciatic nerve injury site; in contrast, 3 weeks of prior wheel running, which failed to prevent neuropathic pain, had no effect on Nrf2 nuclear translocation. The protective effects of prior voluntary wheel running were mediated by Nrf2, as suppression was abolished across both sexes when Nrf2 activation was blocked during the 6-week running phase. This study provides insight into the mechanisms by which physical activity may prevent neuropathic pain. Preconditioning by voluntary wheel running, terminated prior to nerve injury, suppresses later neuropathic pain in both sexes, and it is modulated through the activation of Nrf2-antioxidant signaling.


Asunto(s)
Neuralgia , Traumatismos de los Nervios Periféricos , Neuropatía Ciática , Animales , Antioxidantes , Femenino , Hiperalgesia/prevención & control , Masculino , Actividad Motora/fisiología , NADPH Oxidasa 2/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neuralgia/metabolismo , Neuralgia/prevención & control , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ácido Peroxinitroso/metabolismo , Ratas , Ratas Sprague-Dawley , Neuropatía Ciática/prevención & control , Superóxidos/metabolismo
5.
Int Rev Immunol ; 41(5): 475-516, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34152881

RESUMEN

Insights into T cell form, function, and dysfunction are rapidly evolving. T cells have remarkably varied effector functions including protecting the host from infection, activating cells of the innate immune system, releasing cytokines and chemokines, and heavily contributing to immunological memory. Under healthy conditions, T cells orchestrate a finely tuned attack on invading pathogens while minimizing damage to the host. The dark side of T cells is that they also exhibit autoreactivity and inflict harm to host cells, creating autoimmunity. The mechanisms of T cell autoreactivity are complex and dynamic. Emerging research is elucidating the mechanisms leading T cells to become autoreactive and how such responses cause or contribute to diverse disease states, both peripherally and within the central nervous system. This review provides foundational information on T cell development, differentiation, and functions. Key T cell subtypes, cytokines that create their effector roles, and sex differences are highlighted. Pathological T cell contributions to diverse peripheral and central disease states, arising from errors in reactivity, are highlighted, with a focus on multiple sclerosis, rheumatoid arthritis, osteoarthritis, neuropathic pain, and type 1 diabetes.


Asunto(s)
Artritis Reumatoide , Linfocitos T , Autoinmunidad , Quimiocinas/metabolismo , Citocinas/metabolismo , Femenino , Humanos , Masculino
6.
Brain Behav Immun ; 93: 80-95, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33358978

RESUMEN

Neuropathic pain is a major symptom of multiple sclerosis (MS) with up to 92% of patients reporting bodily pain, and 85% reporting pain severe enough to cause functional disability. None of the available therapeutics target MS pain. Toll-like receptors 2 and 4 (TLR2/TLR4) have emerged as targets for treating a wide array of autoimmune disorders, including MS, as well as having demonstrated success at suppressing pain in diverse animal models. The current series of studies tested systemic TLR2/TLR4 antagonists in males and females in a low-dose Myelin oligodendrocyte glycoprotein (MOG) experimental autoimmune encephalomyelitis (EAE) model, with reduced motor dysfunction to allow unconfounded testing of allodynia through 50+ days post-MOG. The data demonstrated that blocking TLR2/TLR4 suppressed EAE-related pain, equally in males and females; upregulation of dorsal spinal cord proinflammatory gene expression for TLR2, TLR4, NLRP3, interleukin-1ß, IkBα, TNF-α and interleukin-17; and upregulation of dorsal spinal cord expression of glial immunoreactivity markers. In support of these results, intrathecal interleukin-1 receptor antagonist reversed EAE-induced allodynia, both early and late after EAE induction. In contrast, blocking TLR2/TLR4 did not suppress EAE-induced motor disturbances induced by a higher MOG dose. These data suggest that blocking TLR2/TLR4 prevents the production of proinflammatory factors involved in low dose EAE pathology. Moreover, in this EAE model, TLR2/TLR4 antagonists were highly effective in reducing pain, whereas motor impairment, as seen in high dose MOG EAE, is not affected.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Manejo del Dolor , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple , Glicoproteína Mielina-Oligodendrócito , Dolor , Médula Espinal
7.
Behav Brain Res ; 396: 112896, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32905811

RESUMEN

Multiple sclerosis (MS) is associated with burdensome memory impairments and preclinical literature suggests that these impairments are linked to neuroinflammation. Previously, we have shown that toll-like receptor 4 (TLR4) antagonists, such as (+)-naltrexone [(+)-NTX], block neuropathic pain and associated spinal inflammation in rats. Here we extend these findings to first demonstrate that (+)-NTX blocks TLR2 in addition to TLR4. Additionally, we examined in two rat strains whether (+)-NTX could attenuate learning and memory disturbances and associated neuroinflammation using a low-dose experimental autoimmune encephalomyelitis (EAE) model of MS. EAE is the most commonly used experimental model for the human inflammatory demyelinating disease, MS. This low-dose model avoided motor impairments that would confound learning and memory measurements. Fourteen days later, daily subcutaneous (+)-NTX or saline injections began and continued throughout the study. Contextual and auditory-fear conditioning were conducted at day 21 to assess hippocampal and amygdalar function. With this low-dose model, EAE impaired long-term, but not short-term, contextual fear memory; both long-term and short-term auditory-cued fear memory were spared. This was associated with increased mRNA for hippocampal interleukin-1ß (IL-1ß), TLR2, TLR4, NLRP3, and IL-17 and elevated expression of the microglial marker Iba1 in CA1 and DG regions of the hippocampus, confirming the neuroinflammation observed in higher-dose EAE models. Importantly, (+)-NTX completely prevented the EAE-induced memory impairments and robustly attenuated the associated proinflammatory effects. These findings suggest that (+)-NTX may exert therapeutic effects on memory function by dampening the neuroinflammatory response in the hippocampus through blockade of TLR2/TLR4. This study suggests that TLR2 and TLR4 antagonists may be effective at treating MS-related memory deficits.


Asunto(s)
Encefalomielitis Autoinmune Experimental/complicaciones , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Inflamación/etiología , Inflamación/prevención & control , Trastornos de la Memoria/etiología , Trastornos de la Memoria/prevención & control , Esclerosis Múltiple/complicaciones , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Células Cultivadas , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Clásico/fisiología , Miedo/efectos de los fármacos , Miedo/fisiología , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/inmunología , Naltrexona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 4/antagonistas & inhibidores
8.
Behav Brain Res ; 380: 112369, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31743731

RESUMEN

It is well accepted that opioids promote feeding for reward. Some studies suggest a potential involvement in hunger-driven intake, but they suffer from the scarcity of methodologies differentiating between factors that intersect eating for pleasure versus energy. Here, we used a unique food deprivation discrimination paradigm to test a hypothesis that, since opioids appear to control feeding reward, injection of opioid agonists would not produce effects akin to 22 h of food deprivation. We trained rats to discriminate between 22 h and 2 h food deprivation in a two-lever, operant discrimination procedure. We tested whether opioid agonists at orexigenic doses produce discriminative stimulus effects similar to 22 h deprivation. We injected DAMGO, DSLET, or orphanin FQ in the paraventricular hypothalamic nucleus (PVN), a site regulating hunger/satiety, and butorphanol subcutaneously (to produce maximum consumption). We assessed the ability of the opioid antagonist, naltrexone, to reduce the discriminative stimulus effects of 22 h deprivation and of the 22 h deprivation-like discriminative stimulus effects of PVN-injected hunger mediator, neuropeptide Y (NPY). In contrast to PVN NPY, centrally or peripherally injected opioid agonists failed to induce discriminative stimuli similar to those of 22 h deprivation. In line with that, naltrexone did not reduce the hunger discriminative stimuli induced by either 22 h deprivation or NPY administration in 2 h food-restricted subjects, even though doses used therein were sufficient to decrease deprivation-induced feeding in a non-operant setting in animals familiar with consequences of 2 h and 22 h deprivation. We conclude that opioids promote feeding for reward rather than in order to replenish lacking energy.


Asunto(s)
Analgésicos Opioides/farmacología , Conducta Animal/efectos de los fármacos , Aprendizaje Discriminativo/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Privación de Alimentos , Hambre/efectos de los fármacos , Antagonistas de Narcóticos/farmacología , Neuropéptido Y/efectos de los fármacos , Saciedad/efectos de los fármacos , Percepción del Tiempo/efectos de los fármacos , Analgésicos Opioides/administración & dosificación , Animales , Condicionamiento Operante/efectos de los fármacos , Masculino , Naltrexona/farmacología , Antagonistas de Narcóticos/administración & dosificación , Ratas , Ratas Sprague-Dawley , Recompensa
9.
Pain ; 160(11): 2634-2640, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31299018

RESUMEN

Opioids are widely prescribed for chronic pain, including neuropathic pain, despite growing evidence of long-term harm. Previous preclinical studies have documented exacerbation of nociceptive hypersensitivity, including that induced by peripheral nerve injury, by morphine. The present series of behavioral studies sought to replicate and extend our prior research, which demonstrated a multimonth exacerbation of nociceptive hypersensitivity by a 5-day course of morphine initiated 10 days after nerve injury. The current studies demonstrate that enduring exacerbation of nociceptive hypersensitivity is not restricted to morphine, but rather is also created by the clinically relevant opioids fentanyl and oxycodone when these are likewise administered for 5 days beginning 10 days after nerve injury. Furthermore, enduring exacerbation of nociceptive hypersensitivity is also observed when the same dosing regimen for either morphine, fentanyl, or oxycodone begins 1 month after nerve injury. Finally, a striking result from these studies is that no such exacerbation of nociceptive hypersensitivity occurs when either morphine, fentanyl, or oxycodone dosing begins at the time of nerve injury. These results extend our previous findings that morphine exacerbates nociceptive hypersensitivity to the clinically relevant opioids fentanyl and oxycodone when administered after the development of nociceptive hypersensitivity, while also providing possible clinically relevant insight into when these opioids can be safely administered and not exacerbate neuropathic pain.


Asunto(s)
Fentanilo/farmacología , Morfina/farmacología , Neuralgia/tratamiento farmacológico , Oxicodona/farmacología , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacología , Animales , Dolor Crónico/tratamiento farmacológico , Modelos Animales de Enfermedad , Fentanilo/administración & dosificación , Masculino , Morfina/administración & dosificación , Oxicodona/administración & dosificación , Dimensión del Dolor/métodos , Ratas Sprague-Dawley
10.
FASEB J ; 33(8): 9577-9587, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31162938

RESUMEN

Deregulation of innate immune TLR4 signaling contributes to various diseases including neuropathic pain and drug addiction. Naltrexone is one of the rare TLR4 antagonists with good blood-brain barrier permeability and showing no stereoselectivity for TLR4. By linking 2 naltrexone units through a rigid pyrrole spacer, the bivalent ligand norbinaltorphimine was formed. Interestingly, (+)-norbinaltorphimine [(+)-1] showed ∼25 times better TLR4 antagonist activity than naltrexone in microglial BV-2 cell line, whereas (-)-norbinaltorphimine [(-)-1] lost TLR4 activity. The enantioselectivity of norbinaltorphimine was further confirmed in primary microglia, astrocytes, and macrophages. The activities of meso isomer of norbinaltorphimine and the molecular dynamic simulation results demonstrate that the stereochemistry of (+)-1 is derived from the (+)-naltrexone pharmacophore. Moreover, (+)-1 significantly increased and prolonged morphine analgesia in vivo. The efficacy of (+)-1 is long lasting. This is the first report showing enantioselective modulation of the innate immune TLR signaling.-Zhang, X., Peng, Y., Grace, P. M., Metcalf, M. D., Kwilasz, A. J., Wang, Y., Zhang, T., Wu, S., Selfridge, B. R., Portoghese, P. S., Rice, K. C., Watkins, L. R., Hutchinson, M. R., Wang, X. Stereochemistry and innate immune recognition: (+)-norbinaltorphimine targets myeloid differentiation protein 2 and inhibits toll-like receptor 4 signaling.


Asunto(s)
Antígeno 96 de los Linfocitos/metabolismo , Naltrexona/análogos & derivados , Receptor Toll-Like 4/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Interleucina-1beta/metabolismo , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Naltrexona/química , Naltrexona/farmacología , Estructura Secundaria de Proteína , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Estereoisomerismo , Relación Estructura-Actividad , Receptor Toll-Like 4/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
11.
Brain Behav Immun ; 76: 116-125, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30453021

RESUMEN

Neuropathic pain is a widespread problem which remains poorly managed by currently available therapeutics. Peripheral nerve injury and inflammation leads to changes at the nerve injury site, including activation of resident and recruited peripheral immune cells, that lead to neuronal central sensitization and pain amplification. The present series of studies tested the effects of peri-sciatic nerve delivery of single doses of adenosine 2A receptor (A2aR) agonists on pain and neuroinflammation. The data provide converging lines of evidence supportive that A2aR agonism at the site of peripheral nerve injury and inflammation is effective in suppressing ongoing neuropathic pain. After A2aR agonism resolved neuropathic pain, a return of pain enhancement (allodynia) was observed in response to peri-sciatic injection of H-89, which can inhibit protein kinase A, and by peri-sciatic injection of neutralizing antibody against the potent anti-inflammatory cytokine interleukin-10. A2aR agonist actions at the nerve injury site suppress neuroinflammation, as reflected by decreased release of interleukin-1ß and nitric oxide, as well as decreased sciatic expression of markers of monocytes/macrophages and inducible nitric oxide synthase. Taken together, the data are supportive that A2aR agonists, acting at the level of peripheral nerve injury, may be of therapeutic value in treating chronic pain of neuroinflammatory origin.


Asunto(s)
Piperidinas/farmacología , Nervio Ciático/efectos de los fármacos , Neuropatía Ciática/tratamiento farmacológico , Animales , Citocinas/metabolismo , Hiperalgesia/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Inyecciones Espinales/métodos , Masculino , Neuralgia/tratamiento farmacológico , Traumatismos de los Nervios Periféricos , Piperidinas/metabolismo , Agonistas del Receptor Purinérgico P1/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P1/metabolismo , Nervio Ciático/lesiones
12.
Brain Behav Immun ; 69: 470-479, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29366930

RESUMEN

Central neuropathic pain is a debilitating outcome of spinal cord injury (SCI) and current treatments to alleviate this pain condition are ineffective. A growing body of literature suggests that activating adenosine A2A receptors (A2ARs) decreases the production of proinflammatory cytokines and increases the production of anti-inflammatory cytokines. Here, the effect of administering intrathecal A2AR agonists on central neuropathic pain was measured using hindpaw mechanical allodynia in a rat model of SCI termed spinal neuropathic avulsion pain (SNAP). Other models of SCI cause extensive damage to the spinal cord, resulting in paralysis and health problems. SNAP rats with unilateral low thoracic (T13)/high lumbar (L1) dorsal root avulsion develop below-level bilateral allodynia, without concomitant motor or health problems. A single intrathecal injection of the A2AR agonist 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamido adenosine HCl (CGS21680) reversed SCI-induced allodynia for at least 6 weeks. The reversal is likely in part mediated by interleukin (IL)-10, as intrathecally administering neutralizing IL-10 antibodies 1 week after CGS21680 abolished the anti-allodynic effect of CGS21680. Dorsal spinal cord tissue from the ipsilateral site of SCI (T13/L1) was assayed 1 and 6 weeks after CGS21680 for IL-10, CD11b, and tumor necrosis factor (TNF) gene expression. CGS21680 treatment did not change IL-10 gene expression but did significantly decrease CD11b and TNF gene expression at both timepoints. A second A2AR agonist, 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), was also able to significantly prevent and reverse SCI-induced allodynia for several weeks after a single intrathecal injection, providing converging lines of evidence of A2AR involvement. The enduring pain reversal after a single intrathecal injection of A2AR agonists suggests that A2AR agonists could be exciting new candidates for treating SCI-induced central neuropathic pain.


Asunto(s)
Agonistas del Receptor de Adenosina A2/uso terapéutico , Adenosina/análogos & derivados , Hiperalgesia/tratamiento farmacológico , Neuralgia/tratamiento farmacológico , Fenetilaminas/uso terapéutico , Traumatismos de la Médula Espinal/complicaciones , Adenosina/uso terapéutico , Animales , Anticuerpos Neutralizantes/farmacología , Hiperalgesia/etiología , Hiperalgesia/fisiopatología , Interleucina-10/inmunología , Masculino , Neuralgia/etiología , Neuralgia/fisiopatología , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/fisiopatología
13.
Pain ; 157(9): 2012-23, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27355182

RESUMEN

Exercise is known to exert a systemic anti-inflammatory influence, but whether its effects are sufficient to protect against subsequent neuropathic pain is underinvestigated. We report that 6 weeks of voluntary wheel running terminating before chronic constriction injury (CCI) prevented the full development of allodynia for the ∼3-month duration of the injury. Neuroimmune signaling was assessed at 3 and 14 days after CCI. Prior exercise normalized ipsilateral dorsal spinal cord expression of neuroexcitatory interleukin (IL)-1ß production and the attendant glutamate transporter GLT-1 decrease, as well as expression of the disinhibitory P2X4R-BDNF axis. The expression of the macrophage marker Iba1 and the chemokine CCL2 (MCP-1), and a neuronal injury marker (activating transcription factor 3), was attenuated by prior running in the ipsilateral lumbar dorsal root ganglia. Prior exercise suppressed macrophage infiltration and/or injury site proliferation, given decreased presence of macrophage markers Iba1, iNOS (M1), and Arg-1 (M2; expression was time dependent). Chronic constriction injury-driven increases in serum proinflammatory chemokines were suppressed by prior running, whereas IL-10 was increased. Peripheral blood mononuclear cells were also stimulated with lipopolysaccharide ex vivo, wherein CCI-induced increases in IL-1ß, nitrite, and IL-10 were suppressed by prior exercise. Last, unrestricted voluntary wheel running, beginning either the day of, or 2 weeks after, CCI, progressively reversed neuropathic pain. This study is the first to investigate the behavioral and neuroimmune consequences of regular exercise terminating before nerve injury. This study suggests that chronic pain should be considered a component of "the diseasome of physical inactivity," and that an active lifestyle may prevent neuropathic pain.


Asunto(s)
Técnicas de Ejercicio con Movimientos/métodos , Neuralgia/prevención & control , Factor de Transcripción Activador 3/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Constricción Patológica/complicaciones , Citocinas/metabolismo , Modelos Animales de Enfermedad , Transportador 2 de Aminoácidos Excitadores/metabolismo , Lateralidad Funcional , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Hiperalgesia/rehabilitación , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Masculino , Proteínas de Microfilamentos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuralgia/etiología , Neuralgia/patología , Nitritos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2X5/metabolismo , Neuropatía Ciática/prevención & control , Quinasas p21 Activadas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Behav Pharmacol ; 25(2): 119-29, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24583930

RESUMEN

Cannabinoid receptor (CBR) agonists produce antinociception in conventional preclinical assays of pain-stimulated behavior but are not effective in preclinical assays of pain-depressed behavior. Fatty acid amide hydrolase (FAAH) inhibitors increase physiological levels of the endocannabinoid anandamide, which may confer improved efficacy and safety relative to direct CBR agonists. To further evaluate FAAH inhibitors as candidate analgesics, this study assessed the effects of the FAAH inhibitor URB597 in assays of acute pain-stimulated and pain-depressed behavior in male Sprague-Dawley rats. Intraperitoneal injection of dilute lactic acid served as a noxious stimulus to stimulate a stretching response or depress positively reinforced operant behavior (intracranial self-stimulation), and URB597 was tested 1 and 4 h after administration. Consistent with FAAH inhibitor effects in other assays of pain-stimulated behavior, URB597 (1-10 mg/kg intraperitoneally) produced dose-related and CB1R-mediated decreases in acid-stimulated stretching. Conversely, in the assay of acid-depressed intracranial self-stimulation, URB597 produced a delayed, partial and non-CBR-mediated antinociceptive effect. The antinociceptive dose of URB597 (10 mg/kg) increased plasma and brain anandamide levels. These results suggest that URB597 produces antinociception in these models of 'pain stimulated' and 'pain depressed' behavior, but with different rates of onset and differential involvement of CBRs.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Analgésicos/farmacología , Benzamidas/farmacología , Carbamatos/farmacología , Depresión/tratamiento farmacológico , Dolor/tratamiento farmacológico , Animales , Ácidos Araquidónicos/sangre , Ácidos Araquidónicos/metabolismo , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Depresión/etiología , Depresión/metabolismo , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Endocannabinoides/sangre , Endocannabinoides/metabolismo , Inhibidores Enzimáticos/farmacología , Neuroestimuladores Implantables , Inyecciones Intraperitoneales , Ácido Láctico , Masculino , Dolor/complicaciones , Dolor/metabolismo , Alcamidas Poliinsaturadas/sangre , Alcamidas Poliinsaturadas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Cannabinoides/metabolismo , Autoadministración
15.
J Pharmacol Exp Ther ; 343(2): 389-400, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22892341

RESUMEN

Cannabinoid receptor agonists produce reliable antinociception in most preclinical pain assays but have inconsistent analgesic efficacy in humans. This disparity suggests that conventional preclinical assays of nociception are not sufficient for the prediction of cannabinoid effects related to clinical analgesia. To extend the range of preclinical cannabinoid assessment, this study compared the effects of the marijuana constituent and low-efficacy cannabinoid agonist Δ9-tetrahydrocannabinol (THC) and the high-efficacy synthetic cannabinoid agonist 3-(2-hydroxy-4-(1,1-dimethylheptyl)phenyl)-4-(3-hydroxypropyl)cyclohexanol (CP55940) in assays of pain-stimulated and pain-depressed behavior. Intraperitoneal injection of dilute lactic acid (1.8% in 1 ml/kg) stimulated a stretching response or depressed intracranial self-stimulation (ICSS) in separate groups of male Sprague-Dawley rats. THC (0.1-10 mg/kg) and CP55940 (0.0032-0.32 mg/kg) dose-dependently blocked acid- stimulated stretching but only exacerbated acid-induced depression of ICSS at doses that also decreased control ICSS in the absence of a noxious stimulus. Repeated THC produced tolerance to sedative rate-decreasing effects of THC on control ICSS in the absence of the noxious stimulus but failed to unmask antinociception in the presence of the noxious stimulus. THC and CP55940 also failed to block pain-related depression of feeding in rats, although THC did attenuate satiation-related depression of feeding. In contrast to the effects of the cannabinoid agonists, the clinically effective analgesic and nonsteroidal anti-inflammatory drug ketoprofen (1 mg/kg) blocked acid-stimulated stretching and acid-induced depression of both ICSS and feeding. The poor efficacy of THC and CP55940 to block acute pain-related depression of behavior in rats agrees with the poor efficacy of cannabinoids to treat acute pain in humans.


Asunto(s)
Analgésicos/farmacología , Conducta Animal/efectos de los fármacos , Agonistas de Receptores de Cannabinoides/farmacología , Ciclohexanoles/farmacología , Dronabinol/farmacología , Dolor/psicología , Animales , Antiinflamatorios no Esteroideos/farmacología , Encéfalo/fisiología , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Electrodos Implantados , Cetoprofeno/farmacología , Ácido Láctico/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Autoestimulación
16.
Pharmacol Biochem Behav ; 94(1): 114-8, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19660491

RESUMEN

The introduction of transgenic and knockout mice has shaped new interest in developing novel and modified behavioral methods for mice that evaluate the various manifestations of nicotine withdrawal syndromes. This study assessed the disruption of operant baselines during drug withdrawal, an established rat model of nicotine dependence, in mice. Subjects were trained to lever press for food reinforcement during daily operant sessions. After stable operant baselines were established, mice were implanted with osmotic minipumps containing 0 (saline), 6, 12, 24, or 48 mg/kg/day nicotine base. Operant responding was assessed for disruptions in daily sessions throughout the experiment. Somatic signs of withdrawal were assessed after the operant session on day 7, following administration of mecamylamine (1 mg/kg), and on days 12, 13, and 14, following spontaneous removal of nicotine. Spontaneous removal of nicotine increased somatic signs of withdrawal but did not disrupt operant responding. Mecamylamine failed to produce signs of precipitated withdrawal in either procedure. This study demonstrated nicotine dependence in mice during spontaneous removal of nicotine. Moreover, since signs of behavioral withdrawal (i.e. disruptions in operant response rates) were not observed, these findings suggest the importance of considering differences in the apparent manifestations of withdrawal syndromes while evaluating nicotine dependence.


Asunto(s)
Conducta Animal/efectos de los fármacos , Mecamilamina/farmacología , Nicotina/efectos adversos , Agonistas Nicotínicos/efectos adversos , Antagonistas Nicotínicos/farmacología , Síndrome de Abstinencia a Sustancias/diagnóstico , Animales , Condicionamiento Operante , Relación Dosis-Respuesta a Droga , Habituación Psicofisiológica , Bombas de Infusión Implantables , Infusiones Subcutáneas , Masculino , Mecamilamina/administración & dosificación , Ratones , Ratones Endogámicos ICR , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Antagonistas Nicotínicos/administración & dosificación , Refuerzo en Psicología , Síndrome de Abstinencia a Sustancias/etiología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...