Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Pharmacol ; 14: 1253901, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38152690

RESUMEN

Progesterone has been shown to have neuroprotective capabilities against a wide range of nervous system injuries, however there are negative clinical studies that have failed to demonstrate positive effects of progesterone therapy. Specifically, we looked into whether progesterone receptors or its metabolizing enzymes, cytochrome P450c17 and 5α-reductase, are involved in the effects of progesterone on neuropathic pain after chronic constriction injury (CCI) of the sciatic nerve in mice. Intrathecal progesterone administration during the induction phase of chronic pain enhanced mechanical allodynia development and spinal glial fibrillary acidic protein (GFAP) expression, and this enhancement was inhibited by administration of ketoconazole, a P450c17 inhibitor, but not finasteride, a 5α-reductase inhibitor. Furthermore, phospho-serine levels of P450c17 in the spinal cord were elevated on day 1 after CCI operation, but not on day 17. In contrast, intrathecal progesterone administration during the maintenance phase of chronic pain decreased the acquired pain and elevated GFAP expression; this inhibition was restored by finasteride administration, but not by ketoconazole. The modification of mechanical allodynia brought on by progesterone in CCI mice was unaffected by the administration of mifepristone, a progesterone receptor antagonist. Collectively, these findings imply that progesterone suppresses spinal astrocyte activation via 5α-reductase activity during the maintenance phase of chronic pain and has an analgesic impact on the mechanical allodynia associated with the growing neuropathy. Progesterone, however, stimulates spinal astrocytes during the induction stage of peripheral neuropathy and boosts the allodynic impact caused by CCI through early spinal P450c17 activation.

2.
Biomed Pharmacother ; 144: 112272, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34607109

RESUMEN

The sigma-1 receptor (Sig-1R) plays an important role in spinal pain transmission by increasing phosphorylation of the N-methyl-D-aspartate (NMDA) receptor GluN1 subunit (pGluN1). As a result Sig-1R has been suggested as a novel therapeutic target for prevention of chronic pain. Here we investigated whether interleukin-1ß (IL-1ß) modulates the expression of the Sig-1R in spinal astrocytes during the early phase of nerve injury, and whether this modulation affects spinal pGluN1 expression and the development of neuropathic pain following chronic constriction injury (CCI) of the sciatic nerve. Repeated intrathecal (i.t.) administration of IL-1ß from days 0-3 post-surgery significantly reduced the increased pGluN1 expression at the Ser896 and Ser897 sites in the ipsilateral spinal cord, as well as, the development of mechanical allodynia and thermal hyperalgesia in the ipsilateral hind paw of CCI mice, which were restored by co-administration of IL-1 receptor antagonist with IL-1ß. Sciatic nerve injury increased the expression of Sig-1R in astrocytes of the ipsilateral spinal cord, and this increase was suppressed by i.t. administration of IL-1ß. Agonistic stimulation of the Sig-1R with PRE084 restored pGluN1 expression and the development of mechanical allodynia that were originally suppressed by IL-1ß in CCI mice. Collectively these results demonstrate that IL-1ß administration during the induction phase of neuropathic pain produces an analgesic effect on neuropathic pain development by controlling the expression of Sig-1R in spinal astrocytes.


Asunto(s)
Analgésicos/administración & dosificación , Astrocitos/efectos de los fármacos , Hiperalgesia/prevención & control , Interleucina-1beta/administración & dosificación , Neuralgia/prevención & control , Umbral del Dolor/efectos de los fármacos , Receptores sigma/metabolismo , Médula Espinal/efectos de los fármacos , Animales , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Inyecciones Espinales , Masculino , Ratones Endogámicos ICR , Proteínas del Tejido Nervioso/metabolismo , Neuralgia/metabolismo , Neuralgia/fisiopatología , Fosforilación , Receptores de N-Metil-D-Aspartato/metabolismo , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Receptor Sigma-1
3.
Front Pharmacol ; 10: 1439, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31866864

RESUMEN

Research indicates that neurosteroids are locally synthesized in the central nervous system and play an important modulatory role in nociception. While the neurosteroidogenic enzyme, cytochrome P450 side-chain cleavage enzyme (P450scc), is the initiating enzyme of steroidogenesis, P450scc has not been examined under the pathophysiological conditions associated with peripheral neuropathy. Thus, we investigated whether chronic constriction injury (CCI) of the sciatic nerve increases the expression of P450scc in the spinal cord and whether this increase modulates serine racemase (Srr) expression and D-serine production contributing to the development of neuropathic pain. CCI increased the immunoreactivity of P450scc in astrocytes of the ipsilateral lumbar spinal cord dorsal horn. Intrathecal administration of the P450scc inhibitor, aminoglutethimide, during the induction phase of neuropathic pain (days 0 to 3 post-surgery) significantly suppressed the CCI-induced development of mechanical allodynia and thermal hyperalgesia, the increased expression of astrocyte Srr in both the total and cytosol levels, and the increases in D-serine immunoreactivity at day 3 post-surgery. By contrast, intrathecal administration of aminoglutethimide during the maintenance phase of pain (days 14 to 17 post-surgery) had no effect on the developed neuropathic pain nor the expression of spinal Srr and D-serine immunoreactivity at day 17 post-surgery. Intrathecal administration of exogenous D-serine during the induction phase of neuropathic pain (days 0 to 3 post-surgery) restored the development of mechanical allodynia, but not the thermal hyperalgesia, that were suppressed by aminoglutethimide administration. Collectively, these results demonstrate that spinal P450scc increases the expression of astrocyte Srr and D-serine production, ultimately contributing to the development of mechanical allodynia induced by peripheral nerve injury.

4.
Exp Neurobiol ; 28(4): 516-528, 2019 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-31495080

RESUMEN

We have previously demonstrated that the neurosteroid dehydroepiandrosterone sulfate (DHEAS) induces functional potentiation of N-methyl-D-aspartate (NMDA) receptors via increases in phosphorylation of NMDA receptor GluN1 subunit (pGluN1). However, the modulatory mechanisms responsible for the expression of the DHEA-synthesizing enzyme, cytochrome P450c17 following peripheral nerve injury have yet to be examined. Here we determined whether oxidative stress induced by the spinal activation of nitric oxide synthase type II (NOS-II) modulates the expression of P450c17 and whether this process contributes to the development of neuropathic pain in rats. Chronic constriction injury (CCI) of the sciatic nerve induced a significant increase in the expression of NOS-II in microglial cells and NO levels in the lumbar spinal cord dorsal horn at postoperative day 5. Intrathecal administration of the NOS-II inhibitor, L-NIL during the induction phase of neuropathic pain (postoperative days 0~5) significantly reduced the CCI-induced development of mechanical allodynia and thermal hyperalgesia. Sciatic nerve injury increased the expression of PKCand PKA-dependent pGluN1 as well as the mRNA and protein levels of P450c17 in the spinal cord at postoperative day 5, and these increases were suppressed by repeated administration of L-NIL. Co-administration of DHEAS together with L-NIL restored the development of neuropathic pain and pGluN1 that were originally inhibited by L-NIL administration alone. Collectively these results provide strong support for the hypothesis that activation of NOS-II increases the mRNA and protein levels of P450c17 in the spinal cord, ultimately leading to the development of central sensitization and neuropathic pain induced by peripheral nerve injury.

5.
Biomed Pharmacother ; 118: 109299, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31387001

RESUMEN

We have recently demonstrated that the neurosteroid-metabolizing enzyme, cytochrome P450c17 is increased in spinal astrocytes contributing to the development of mechanical allodynia in chronic constriction injury (CCI)-induced neuropathic mice. However, the mechanisms by which spinal P450c17 modulates pathological changes in astrocytes remain unclear. In this study we investigated whether P450c17 modulates astrocyte activation and whether this process is mediated by spinal p38 mitogen-activated protein kinase phosphorylation ultimately leading to the development of mechanical allodynia in CCI mice. Sciatic nerve injury induced a significant increase in glial fibrillary acidic protein (GFAP) expression in the superficial dorsal horn (SDH, laminae I-II) and nucleus proprius (NP, laminae III-IV) regions of the spinal cord dorsal horn. Repeated daily (from days 0-3 post-surgery) intrathecal administration of the P450c17 inhibitor, ketoconazole (10 nmol) significantly inhibited the CCI-induced increase in GFAP-immunoreactivity, but had no effect on the CCI-induced increase in Iba-1-immunoreactivity. In addition, intrathecal administration of ketoconazole significantly inhibited the CCI-induced increase in p38 phosphorylation, while the levels of ERK and JNK phosphorylation remained unchanged. The CCI-induced development of mechanical allodynia was attenuated by administration of either ketoconazole (10 nmol) or the p38 MAPK inhibitor, SB203580 (5 nmol). Administration of a sub-effective dose of SB203580 (0.5 nmol) potentiated the pharmacological effect of ketoconazole (1 nmol) on spinal GFAP-immunostaining, as well as, the development of mechanical allodynia following CCI. Collectively these data suggest that spinal P450c17 activates astrocytes via p38 phosphorylation, ultimately leading to the development of mechanical allodynia in a model of peripheral neuropathy.


Asunto(s)
Astrocitos/enzimología , Neuralgia/enzimología , Neuralgia/patología , Médula Espinal/patología , Esteroide 17-alfa-Hidroxilasa/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Constricción Patológica , Modelos Animales de Enfermedad , Hiperalgesia/complicaciones , Hiperalgesia/patología , Imidazoles/farmacología , Cetoconazol/administración & dosificación , Cetoconazol/farmacología , Vértebras Lumbares/enzimología , Vértebras Lumbares/patología , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/patología , Fosforilación/efectos de los fármacos , Piridinas/farmacología , Asta Dorsal de la Médula Espinal/enzimología , Asta Dorsal de la Médula Espinal/patología , Esteroide 17-alfa-Hidroxilasa/metabolismo
6.
Front Mol Neurosci ; 12: 153, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31281242

RESUMEN

We have recently demonstrated that sciatic nerve injury increases the expression of spinal cytochrome P450c17, a key neurosteroidogenic enzyme, which plays a critical role in the development of peripheral neuropathic pain. However, the modulatory mechanisms responsible for the expression of spinal P450c17 have yet to be examined. Here we investigated the possible involvement of interleukin-1ß (IL-1ß) in altering P450c17 expression during the induction phase of neuropathic pain. Neuropathic pain was produced by chronic constriction injury (CCI) of the right sciatic nerve in mice and mechanical allodynia was evaluated in the hind paws using a von-Frey filament (0.16 g). Western blotting and immunohistochemistry were performed to assess the expression of spinal IL-1ß, interleukin-1 receptor type 1 (IL-1R1), P450c17, and GFAP. Spinal IL-1ß was significantly increased on day 1 post-surgery and its receptor, IL-1R1 was expressed in GFAP-positive astrocytes. Intrathecal administration of the recombinant interleukin-1 receptor antagonist (IL-1ra, 20 ng) on days 0 and 1 post-surgery enhanced GFAP expression on day 1 post-surgery and induced an early increase in P450c17 expression in astrocytes, but not in neurons. Administration of IL-1ß (10 ng) on days 0 and 1 post-surgery blocked the enhancement of both spinal P450c17 and GFAP expression induced by IL-1ra (20 ng) administration. Intrathecal administration of IL-1ra (20 ng) on days 0 to 3 post-surgery also facilitated the CCI-induced development of mechanical allodynia, and this early developed pain was dose-dependently attenuated by the administration of the P450c17 inhibitor, ketoconazole (1, 3, or 10 nmol) or the astrocyte metabolic inhibitor, fluorocitrate (0.01, 0.03, or 0.1 nmol). These results demonstrate that early increases in spinal IL-1ß temporally inhibit astrocyte P450c17 expression and astrocyte activation ultimately controlling the development of mechanical allodynia induced by peripheral nerve injury. These findings imply that spinal IL-1ß plays an important role as an early, but transient, control mechanism in the development of peripheral neuropathic pain via the inhibition of astrocyte P450c17 expression and astrocyte activation.

7.
Neurosci Lett ; 703: 156-161, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-30926374

RESUMEN

It has been suggested that interactions of neuronal nitric oxide synthase (nNOS) with postsynaptic density 95 (PSD95) play important roles in the development of chronic neuropathic pain. Here we examine the possible role of nNOS-PSD95 interactions in central sensitization as represented by phosphorylation of the NMDA receptor GluN1 subunit (pGluN1) in mice with chronic constriction injury (CCI) of the sciatic nerve. Intrathecal administration of the nNOS-PSD95 interactions inhibitor, IC87201 on post-operative days 0-3 significantly reduced the CCI-induced increase in total NO levels in the lumbar spinal cord dorsal horn. IC87201 administration on post-operative days 0-3 also attenuated the CCI-induced development of mechanical allodynia (MA) and PKC-dependent (Ser896) pGluN1. Sciatic nerve injury elicited a significant translocation of the PKC-ε isoform from the cytosol to the membrane fraction in the lumbar spinal cord dorsal horn on day 3 post-CCI surgery. Administration of IC87201 significantly inhibited this translocation of PKC-ε, while the expression of PKC-α and -ξ in the cytosol and membrane fractions was unaffected by sciatic nerve injury or injection of IC87201. Furthermore, administration of the PKC-ε inhibitor, εV1-2 on post-operative days 0-3 attenuated the CCI-induced development of MA and pGluN1. Collectively these results demonstrate that spinal nNOS-PSD95 interactions play an important role in PKC-dependent GluN1 phosphorylation via activation of the PKC-ε isoform, and ultimately contributes to the development of MA in peripheral neuropathy.


Asunto(s)
Homólogo 4 de la Proteína Discs Large/metabolismo , Hiperalgesia/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuralgia/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Activación Enzimática , Isoenzimas/metabolismo , Masculino , Ratones Endogámicos ICR , Fosforilación , Estimulación Física , Nervio Ciático/lesiones , Tacto
8.
Neuropharmacology ; 149: 169-180, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30797030

RESUMEN

While evidence indicates that sigma-1 receptors (Sig-1Rs) play an important role in the induction of peripheral neuropathic pain, there is limited understanding of the role that the neurosteroidogenic enzymes, which produce Sig-1R endogenous ligands, play during the development of neuropathic pain. We examined whether sciatic nerve injury upregulates the neurosteroidogenic enzymes, cytochrome P450c17 and 3ß-hydroxysteroid dehydrogenase (3ß-HSD), which modulate the expression and/or activation of Sig-1Rs leading to the development of peripheral neuropathic pain. Chronic constriction injury (CCI) of the sciatic nerve induced a significant increase in the expression of P450c17, but not 3ß-HSD, in the ipsilateral lumbar spinal cord dorsal horn at postoperative day 3. Intrathecal administration of the P450c17 inhibitor, ketoconazole during the induction phase of neuropathic pain (day 0 to day 3 post-surgery) significantly reduced the development of mechanical allodynia and thermal hyperalgesia in the ipsilateral hind paw. However, administration of the 3ß-HSD inhibitor, trilostane had no effect on the development of neuropathic pain. Sciatic nerve injury increased astrocyte Sig-1R expression as well as dissociation of Sig-1Rs from BiP in the spinal cord. These increases were suppressed by administration of ketoconazole, but not by administration of trilostane. Co-administration of the Sig-1R agonist, PRE084 restored the development of mechanical allodynia originally suppressed by the ketoconazole administration. However, ketoconazole-induced inhibition of thermal hyperalgesia was not affected by co-administration of PRE084. Collectively these results demonstrate that early activation of P450c17 modulates the expression and activation of astrocyte Sig-1Rs, ultimately contributing to the development of mechanical allodynia induced by peripheral nerve injury.


Asunto(s)
Hiperalgesia/metabolismo , Neuralgia/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Receptores sigma/metabolismo , Médula Espinal/enzimología , Esteroide 17-alfa-Hidroxilasa/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Animales , Astrocitos , Dihidrotestosterona/análogos & derivados , Dihidrotestosterona/farmacología , Modelos Animales de Enfermedad , Hiperalgesia/inducido químicamente , Hiperalgesia/enzimología , Hiperalgesia/prevención & control , Cetoconazol/farmacología , Masculino , Ratones , Ratones Endogámicos ICR , Neuralgia/enzimología , Neuroesteroides/metabolismo , Traumatismos de los Nervios Periféricos/inducido químicamente , Traumatismos de los Nervios Periféricos/enzimología , Enfermedades del Sistema Nervioso Periférico/metabolismo , Enfermedades del Sistema Nervioso Periférico/patología , Receptores sigma/agonistas , Nervio Ciático/enzimología , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Nervio Ciático/patología , Médula Espinal/efectos de los fármacos , Asta Dorsal de la Médula Espinal/metabolismo , Receptor Sigma-1
9.
Neuroscience ; 372: 181-191, 2018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29289721

RESUMEN

Aromatase is a key enzyme responsible for the biosynthesis of estrogen from testosterone. Although recent evidence indicates that spinal cord aromatase participates in nociceptive processing, the mechanisms underlying its regulation and its involvement in nociception remain unclear. The present study focuses on the potential role of astrocyte aromatase in formalin-induced acute pain and begins to uncover one mechanism by which spinal aromatase activation is controlled. Following intraplantar formalin injection, nociceptive responses were quantified and immunohistochemistry/co-immunoprecipitation assays were used to investigate the changes in spinal Fos expression and the phospho-serine levels of spinal aromatase. Intrathecal (i.t.) injection of letrozole (an aromatase inhibitor) mitigated both the late phase formalin-induced nociceptive responses and formalin-induced spinal Fos expression. Furthermore, formalin-injected mice showed significantly reduced phospho-serine levels of aromatase, which is associated with the rapid activation of this enzyme. However, sigma-1 receptor inhibition with i.t. BD1047 blocked the dephosphorylation of aromatase and potentiated the pharmacological effect of letrozole on formalin-induced nociceptive responses. In addition, i.t. administration of a sub-effective dose of BD1047 potentiated the pharmacological effect of cyclosporin A (a calcineurin inhibitor) on both the formalin-induced reduction in phospho-serine levels of aromatase and nociceptive behavior. These results suggest that dephosphorylation is an important regulatory mechanism involved in the rapid activation of aromatase and that spinal sigma-1 receptors mediate this dephosphorylation of aromatase through an intrinsic calcineurin pathway.


Asunto(s)
Aromatasa/metabolismo , Astrocitos/metabolismo , Inflamación/metabolismo , Dolor Nociceptivo/metabolismo , Médula Espinal/metabolismo , Animales , Inhibidores de la Aromatasa/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/patología , Calcineurina/metabolismo , Formaldehído , Proteína Ácida Fibrilar de la Glía/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/patología , Letrozol , Masculino , Ratones Endogámicos ICR , Nitrilos/farmacología , Dolor Nociceptivo/tratamiento farmacológico , Dolor Nociceptivo/patología , Proteínas Oncogénicas v-fos/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Receptores sigma/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Triazoles/farmacología , Receptor Sigma-1
10.
Biol Pharm Bull ; 41(2): 172-181, 2018 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-29187670

RESUMEN

Despite the relatively high prevalence of migraine or headache, the pathophysiological mechanisms triggering headache-associated peripheral hypersensitivities, are unknown. Since nitric oxide (NO) is well known as a causative factor in the pathogenesis of migraine or migraine-associated hypersensitivities, a mouse model has been established using systemic administration of the NO donor, nitroglycerin (NTG). Here we tried to investigate the time course development of facial or hindpaw hypersensitivity after repetitive NTG injection. NTG (10 mg/kg) was administrated to mice every other day for nine days. Two hours post-injection, NTG produced acute mechanical and heat hypersensitivity in the hind paws. By contrast, cold allodynia, but not mechanical hypersensitivity, occurred in the facial region. Moreover, this hindpaws mechanical hypersensitivity and the facial cold allodynia was progressive and long-lasting. We subsequently examined whether the depletion of capsaicin-sensitive primary afferents (CSPAs) with resiniferatoxin (RTX, 0.02 mg/kg) altered these peripheral hypersensitivities in NTG-treated mice. RTX pretreatment did not affect the NTG-induced mechanical allodynia in the hind paws nor the cold allodynia in the facial region, but it did inhibit the development of hind paw heat hyperalgesia. Similarly, NTG injection produced significant hindpaw mechanical allodynia or facial cold allodynia, but not heat hyperalgesia in transient receptor potential type V1 (TRPV1) knockout mice. These findings demonstrate that different peripheral hypersensitivities develop in the face versus hindpaw regions in a mouse model of repetitive NTG-induced migraine, and that these hindpaw mechanical hypersensitivity and facial cold allodynia are not mediated by the activation of CSPAs.


Asunto(s)
Enfermedades del Nervio Facial/fisiopatología , Hiperalgesia/fisiopatología , Trastornos Migrañosos/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Neuronas Aferentes/efectos de los fármacos , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Canales Catiónicos TRPV/metabolismo , Animales , Capsaicina/farmacología , Frío/efectos adversos , Diterpenos/toxicidad , Resistencia a Medicamentos , Enfermedades del Nervio Facial/inducido químicamente , Enfermedades del Nervio Facial/metabolismo , Enfermedades del Nervio Facial/patología , Miembro Posterior , Calor/efectos adversos , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Hiperalgesia/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos Migrañosos/metabolismo , Trastornos Migrañosos/patología , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Neuronas Aferentes/metabolismo , Neuronas Aferentes/patología , Neurotoxinas/toxicidad , Donantes de Óxido Nítrico/toxicidad , Nitroglicerina/toxicidad , Especificidad de Órganos , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/metabolismo , Enfermedades del Sistema Nervioso Periférico/patología , Fármacos del Sistema Sensorial/farmacología , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética
11.
Br J Pharmacol ; 175(3): 558-572, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29172248

RESUMEN

BACKGROUND AND PURPOSE: Although we have recently demonstrated that spinal astrocyte gap junctions mediate the development of mirror-image pain (MIP), it is still unclear which astrocyte-derived factor is responsible for the development of MIP and how its production is controlled. In the present study, we focused on the role of ipsilateral versus contralateral D-serine in the development of MIP and investigated the possible involvement of σ1 receptors and gap junctions in astrocyte D-serine production. EXPERIMENTAL APPROACH: Following carrageenan injection, mechanical allodynia was tested at various time points to examine the effect of individual drugs. Immunohistochemistry and Western blot analyses were performed to clarify the expression levels of spinal D-serine, serine racemase, σ1 receptors and connexin 43. KEY RESULTS: The expression of ipsilateral D-serine was up-regulated during the early phase of inflammation, while contralateral D-serine increased during the later phase of inflammation. The pharmacological inhibition of D-serine during the early phase blocked the development of both ipsilateral and contralateral mechanical allodynia. However, the inhibition of D-serine during the later phase of inflammation blocked contralateral, but not ipsilateral mechanical allodynia. Furthermore, the inhibition of σ1 receptors during the earlier phase of inflammation inhibited the increase in ipsilateral D-serine. Conversely, the blockade of astrocyte gap junctions suppressed the up-regulation of contralateral D-serine during the later phase of inflammation. CONCLUSION AND IMPLICATIONS: Spinal astrocyte D-serine plays an important role in the development of mirror-image pain. Furthermore, σ1 receptors and astrocyte gap junction signalling mediate ipsilateral and contralateral D-serine production respectively.


Asunto(s)
Astrocitos/fisiología , Carragenina/toxicidad , Uniones Comunicantes/fisiología , Dolor/tratamiento farmacológico , Receptores sigma/fisiología , Serina/administración & dosificación , Animales , Astrocitos/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Uniones Comunicantes/efectos de los fármacos , Inyecciones Espinales , Masculino , Dolor/inducido químicamente , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley , Médula Espinal/efectos de los fármacos , Médula Espinal/fisiología , Receptor Sigma-1
12.
Brain Res Bull ; 134: 47-54, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28652168

RESUMEN

Most acute and chronic animal models of pain rely heavily on reflexive assays for evaluating levels of nociception, which involves removing the animal from its normal social environment. Here, we examine and characterize the influence of social interactions on inflammatory pain-evoked changes in movement in two different mouse strains. To produce inflammatory nociception, we injected CFA bilaterally into the hind paws of Balb/c and C3H mice and then recorded exploratory locomotor activity using an automated detector system to first evaluate the effects of social behavior on nociception. Secondly, we determined if carprofen administration altered the effects of social behavior on nociceptive-evoked movement. This methodology was expanded to create a novel thermal activity assay to objectively measure the effect of heat and cold on CFA-evoked animal movement in paired animals. Paired Balb/c and C3H mice exhibited significant hyper-locomotion that lasted for 3h post-injection in Balb/c, but only 1h post-injection in C3H. Single Balb/c mice only showed increased activity for 1h post-injection, while single C3H mice showed no increase. This CFA-induced increase in activity in paired animals was highly inversely correlated with mechanical allodynia as measured using standard Von Frey filaments. Carprofen administration completely blocked this CFA-induced hyperlocomotor activity. Both heat and cold induced a significant increase in locomotor activity in paired mice injected with CFA, while having no effect on activity in control mice injected with saline. The results presented here indicate that social interactions greatly influence inflammatory pain-induced changes in locomotor activity and indicate that the use of movement-based assays to evaluate nociception in paired mice may provide an alternative and more sensitive method to quantify nociception and characterize novel analgesic effects over time in the context of social interactions in rodent models of pain.


Asunto(s)
Inflamación/psicología , Actividad Motora , Dolor Nociceptivo/psicología , Conducta Social , Animales , Antiinflamatorios no Esteroideos/farmacología , Carbazoles/farmacología , Modelos Animales de Enfermedad , Adyuvante de Freund , Calor , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/psicología , Inflamación/tratamiento farmacológico , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Actividad Motora/efectos de los fármacos , Pruebas Neuropsicológicas , Dolor Nociceptivo/tratamiento farmacológico , Especificidad de la Especie , Tacto
13.
Mol Pain ; 13: 1744806916688902, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28326932

RESUMEN

Background Self-injurious behaviors (SIBs) are devastating traits in autism spectrum disorder (ASD). Although deficits in pain sensation might be one of the contributing factors underlying the development of SIBs, the mechanisms have yet to be addressed. Recently, the Shank2 synaptic protein has been considered to be a key component in ASD, and mutations of SHANK2 gene induce the dysfunction of N-methyl-D-aspartate (NMDA) receptors, suggesting a link between Shank2 and NMDA receptors in ASD. Given that spinal NMDA receptors play a pivotal role in pain hypersensitivity, we investigated the possible role of Shank2 in nociceptive hypersensitivity by examining changes in spontaneous pain following intrathecal NMDA injection in S hank2-/- ( Shank2 knock-out, KO) mice. Results Intrathecal NMDA injection evoked spontaneous nociceptive behaviors. These NMDA-induced nociceptive responses were significantly reduced in Shank2 KO mice. We also observed a significant decrease of NMDA currents in the spinal dorsal horn of Shank2 KO mice. Subsequently, we examined whether mitogen-activated protein kinase or AKT signaling is involved in this reduced pain behavior in Shank2 KO mice because the NMDA receptor is closely related to these signaling molecules. Western blotting and immunohistochemistry revealed that spinally administered NMDA increased the expression of a phosphorylated form of extracellular signal-regulated kinase (p-ERK) which was significantly reduced in Shank2 KO mice. However, p38, JNK, or AKT were not changed by NMDA administration. The ERK inhibitor, PD98059, decreased NMDA-induced spontaneous pain behaviors in a dose-dependent manner in wild-type mice. Moreover, it was found that the NMDA-induced increase in p-ERK was primarily colocalized with Shank2 proteins in the spinal cord dorsal horn. Conclusion Shank2 protein is involved in spinal NMDA receptor-mediated pain, and mutations of Shank2 may suppress NMDA-ERK signaling in spinal pain transmission. This study provides new clues into the mechanisms underlying pain deficits associated with SIB and deserves further study in patients with ASD.


Asunto(s)
Hiperalgesia/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Nocicepción/efectos de los fármacos , Dolor/patología , Médula Espinal/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Modelos Animales de Enfermedad , Agonistas de Aminoácidos Excitadores/toxicidad , Femenino , Flavonoides/farmacología , Hiperalgesia/inducido químicamente , Imidazoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , N-Metilaspartato/toxicidad , Proteínas del Tejido Nervioso/genética , Dolor/inducido químicamente , Dimensión del Dolor , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Médula Espinal/efectos de los fármacos
14.
Brain Res Bull ; 130: 165-172, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28153540

RESUMEN

Although interleukin-1ß (IL-1ß) is a prototypical pro-inflammatory cytokine, the specific mechanisms underlying the role of its cognate receptor, the interleukin-1 receptor (IL-1R) in peripheral sensitization remain to be investigated. Since emerging evidence in the literature indicates that IL-1ß can modulate membrane-bound receptors, we decided to examine the involvement of P2Y1 receptor (P2Y1R) in IL-1ß induced pain and the potential interaction of P2Y1Rs and IL-1Rs in both naïve and carrageenan injected rats. Intraplantar (i.pl) injection of IL-1ß dose-dependently produced mechanical and thermal hypersensitivity in naïve rats. Pre-treatment with IL-1ra (i.pl, 30 and 100ng), an endogenous IL-1R antagonist, prevented the IL-1ß induced mechanical and thermal hypersensitivity. Pre-treatment with MRS2500 (i.pl, 1 and 3nmol), a specific P2Y1R antagonist, dose-dependently reduced IL-1ß induced thermal hypersensitivity, but did not affect the development of mechanical hypersensitivity. Conversely coadministration of MRS2500 (i.pl, 0.1nmol, sub-effective dose) together with IL-1ra (10nmol, sub-effective dose) significantly reduced IL-1ß induced thermal, but not mechanical hypersensitivity. We next used immunohistochemistry to demonstrate that P2Y1 and IL-1 type I receptors co-localize predominantly in small diameter neurons in the dorsal root ganglion. We also performed experiments to examine the interaction of P2Y1Rs and IL-1Rs under the inflammatory conditions induced by 2% carrageenan. Intraplantar coadministration of MRS2500 (3nmol, sub-effective dose) and IL-1ra (30ng, sub-effective dose) significantly reduced inflammatory thermal, but not mechanical, hypersensitivity. These data indicate the involvement of P2Y1Rs in IL-1ß mediated pain in both naive and carrageenan injected rats. There is a positive interaction between peripheral P2Y1Rs and IL-1Rs in both IL-1ß and carrageenan-induced thermal hypersensitivity.


Asunto(s)
Hiperalgesia/fisiopatología , Interleucina-1beta/fisiología , Receptores de Interleucina-1/fisiología , Receptores Purinérgicos P2Y1/fisiología , Animales , Carragenina/administración & dosificación , Hiperalgesia/inducido químicamente , Interleucina-1beta/administración & dosificación , Masculino , Umbral del Dolor , Ratas Sprague-Dawley
15.
J Pain ; 18(4): 415-427, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27986591

RESUMEN

We have recently shown that spinal sigma-1 receptor (Sig-1R) activation facilitates nociception via an increase in phosphorylation of the N-methyl-D-aspartate (NMDA) receptor GluN1 subunit (pGluN1). The present study was designed to examine whether the Sig-1R-induced facilitative effect on NMDA-induced nociception is mediated by D-serine, and whether D-serine modulates spinal pGluN1 expression and the development of neuropathic pain after chronic constriction injury (CCI) of the sciatic nerve. Intrathecal administration of the D-serine degrading enzyme, D-amino acid oxidase attenuated the facilitation of NMDA-induced nociception induced by the Sig-1R agonist, 2-(4-morpholinethyl)1-phenylcyclohexane carboxylate. Exogenous D-serine increased protein kinase C (PKC)-dependent (Ser896) pGluN1 expression and facilitated NMDA-induced nociception, which was attenuated by preteatment with the PKC inhibitor, chelerythrine. In CCI mice, administration of the serine racemase inhibitor, L-serine O-sulfate potassium salt or D-amino acid oxidase on postoperative days 0 to 3 suppressed CCI-induced mechanical allodynia (MA) and pGluN1 expression on day 3 after CCI surgery. Intrathecal administration of D-serine restored MA as well as the GluN1 phosphorylation on day 3 after surgery that was suppressed by the Sig-1R antagonist, N-[2-(3,4-dichlorophenyl)ethyl]-N-methyl-2-(dimethylamino)ethylamine dihydrobromide or the astrocyte inhibitor, fluorocitrate. In contrast, D-serine had no effect on CCI-induced thermal hyperalgesia or GluN1 expression. These results indicate that spinal D-serine: 1) mediates the facilitative effect of Sig-1R on NMDA-induced nociception, 2) modulates PKC-dependent pGluN1 expression, and 3) ultimately contributes to the induction of MA after peripheral nerve injury. PERSPECTIVE: This report shows that reducing D-serine suppresses central sensitization and significantly alleviates peripheral nerve injury-induced chronic neuropathic pain and that this process is modulated by spinal Sig-1Rs. This preclinical evidence provides a strong rationale for using D-serine antagonists to treat peripheral nerve injury-induced neuropathy.


Asunto(s)
Hiperalgesia/etiología , Proteínas del Tejido Nervioso/metabolismo , Neuralgia/complicaciones , Proteína Quinasa C/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores sigma/metabolismo , Serina/farmacología , Animales , D-Aminoácido Oxidasa/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Etilenodiaminas/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR , Morfolinas/farmacología , N-Metilaspartato/farmacología , N-Metilaspartato/toxicidad , Fosforilación/efectos de los fármacos , Estimulación Física/efectos adversos , Receptores sigma/antagonistas & inhibidores , Médula Espinal/efectos de los fármacos , Receptor Sigma-1
16.
Exp Neurol ; 287(Pt 1): 1-13, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27776252

RESUMEN

Although we have recently demonstrated that carrageenan-induced inflammation upregulates the expression of spinal interleukin (IL)-1ß, which inhibits spinal astrocyte activation and results in the delayed development of Mirror-Image Pain (MIP), little is known regarding the mechanisms that underlie how spinal IL-1ß inhibits the astrocyte activation. In this study, we examined the effect of spinal IL-1ß on astrocyte gap junctions (GJ) and the development of MIP. Following unilateral carrageenan (CA) injection, mechanical allodynia (MA) was evaluated at various time points. Immunohistochemistry and Western blot analysis were used to determine changes in the expression of GFAP and connexins (Cx) in the spinal cord dorsal horn. Carrageenan rats showed a delayed onset of contralateral MA, which mimicked the temporal expression pattern of spinal Cx43 (an astrocyte gap junctional protein) and GFAP. Intrathecal administration of an interleukin-1 receptor antagonist (IL-1ra) twice-a-day on post-carrageenan injection days 0 to 3 caused a significant increase in contralateral MA and spinal Cx43 and GFAP expression. In addition, co-administration of IL-1ß with IL-1ra blocked the IL-1ra-induced increase in contralateral MA and the upregulated expression of spinal Cx43 and GFAP. Finally, co-administration of carbenoxolone (CBX; a GJ decoupler) or Gap26 (a specific Cx43 mimetic blocking peptide) with IL-1ra significantly blocked the IL-1ra-induced early development of contralateral MA and the associated upregulation of spinal Cx43 and GFAP expression. These results demonstrate that spinal IL-1ß suppresses Cx43 expression and astrocyte activation during the early phase of CA-induced inflammation resulting in the delayed onset of contralateral MA. These findings imply that spinal IL-1ß can inhibit astrocyte activation and regulate the time of induction of contralateral MA through modulation of spinal Cx43 expression.

17.
Neuropharmacology ; 111: 34-46, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27567941

RESUMEN

We have previously shown using a spinal cord injury (SCI) model that gap junctions contribute to the early spread of astrocyte activation in the lumbar spinal cord and that this astrocyte communication plays critical role in the induction of central neuropathic pain. Sigma-1 receptors (Sig-1Rs) have been implicated in spinal astrocyte activation and the development of peripheral neuropathic pain, yet their contribution to central neuropathic pain remains unknown. Thus, we investigated whether SCI upregulates spinal Sig-1Rs, which in turn increase the expression of the astrocytic gap junction protein, connexin 43 (Cx43) leading to the induction of central neuropathic pain. A thoracic spinal cord hemisection significantly increased both astrocyte activation and Cx43 expression in lumbar dorsal horn. Sig-1Rs were also increased in lumbar dorsal horn astrocytes, but not neurons or microglia. Intrathecal injection of an astrocyte metabolic inhibitor (fluorocitrate); a gap junction/hemichannel blocker (carbenoxolone); or a Cx43 mimetic peptide (43Gap26) significantly reduced SCI-induced bilateral below-level mechanical allodynia. Blockade of Sig-1Rs with BD1047 during the induction phase of pain significantly suppressed the SCI-induced development of mechanical allodynia, astrocyte activation, increased expression of Cx43 in both total and membrane levels, and increased association of Cx43 with Sig-1R. However, SCI did not change the expression of oligodendrocyte (Cx32) or neuronal (Cx36) gap junction proteins. These findings demonstrate that SCI activates astrocyte Sig-1Rs leading to increases in the expression of the gap junction protein, Cx43 and astrocyte activation in the lumbar dorsal horn, and ultimately contribute to the induction of bilateral below-level mechanical allodynia.


Asunto(s)
Astrocitos/metabolismo , Conexina 43/metabolismo , Hiperalgesia/metabolismo , Neuralgia/metabolismo , Receptores sigma/metabolismo , Asta Dorsal de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/complicaciones , Animales , Conexina 43/fisiología , Modelos Animales de Enfermedad , Etilenodiaminas/administración & dosificación , Hiperalgesia/complicaciones , Masculino , Ratones , Ratones Endogámicos ICR , Neuralgia/complicaciones , Receptores sigma/antagonistas & inhibidores , Receptor Sigma-1
18.
Brain Res Bull ; 121: 227-32, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26876754

RESUMEN

The role of peripheral neurosteroids and their related mechanisms on nociception have not been thoroughly investigated. Based on emerging evidence in the literature indicating that neurosteroids and their main target receptors, i.e., sigma-1, GABAA and NMDA, affect P2X-induced changes in neuronal activity, this study was designed to investigate the effect of peripherally injected dehydroepiandrosterone sulphate (DHEAS) and pregnenolone sulfate (PREGS) on P2X receptor-mediated mechanical allodynia in rats. Intraplantar injection of either neurosteroids alone did not produced any detectable changes in paw withdrawal frequency to the innocuous mechanical stimulation in naïve rats. However, When DHEAS or PREGS were co-injected with a sub-effective dose of αßmeATP, mechanical allodynia was developed and this was dose dependently blocked by pre-injection of the P2X antagonist, TNP-ATP. These results demonstrates that DHEAS and PREGS potentiate the activity of P2X receptors which results in the enhancement of αßmeATP-induced mechanical allodynia. In order to investigate the potential role of peripheral sigma-1, GABAA and NMDA receptors in this facilitatory action, we pretreated animals with BD-1047 (a sigma-1 antagonist), muscimol (a GABAA agonist) or MK-801 (a NMDA antagonist) prior to DHEAS or PREGS+αßmeATP injection. Only BD-1047 effectively prevented the facilitatory effects induced by neurosteroids on αßmeATP-induced mechanical allodynia. Collectively, we have shown that peripheral neurosteroids potentiate P2X-induced mechanical allodynia and that this action is mediated by sigma-1, but not by GABAA nor NMDA, receptors.


Asunto(s)
Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Receptores Purinérgicos P2X/metabolismo , Receptores sigma/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Sulfato de Deshidroepiandrosterona/toxicidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Etilenodiaminas/farmacología , Hiperalgesia/tratamiento farmacológico , Masculino , Dimensión del Dolor , Pregnenolona/toxicidad , Agonistas del Receptor Purinérgico P2X/farmacología , Ratas , Ratas Sprague-Dawley , Receptores sigma/antagonistas & inhibidores , Factores de Tiempo
19.
J Pain ; 17(3): 298-309, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26604098

RESUMEN

UNLABELLED: The chemotherapeutic agent, oxaliplatin, produces a robust painful neuropathy that results in the loss of intraepidermal nerve fibers (IENFs). We have previously reported that an acupuncture point (acupoint) injection of diluted bee venom (DBV) produces a temporary antiallodynic effect in oxaliplatin-induced neuropathic mice. Herein we show a significant long-lasting antinociceptive effect of repetitive DBV acupoint treatment on oxaliplatin-induced mechanical allodynia and a significant reduction in the loss of IENFs. DBV (0.1 mg/kg, subcutaneous) was administered once a day for 18 days beginning on day 15 after oxaliplatin injection. Immunohistochemistry for IENF was performed on the glabrous skin of the hind paw footpad using the pan-neuronal marker, protein gene product 9.5. A temporary increase in mechanical threshold was observed 60 minutes after a single DBV injection into the Zusanli acupoint, and this effect was enhanced over time with repetitive DBV treatments. The basal mechanical threshold before daily DBV injection also increased from day 7 after DBV injections, and peaked at day 14 after DBV treatment. Moreover, the oxaliplatin-induced loss of IENFs was significantly reduced in mice treated repetitively with DBV. Repetitive pretreatment with the α-2 adrenoceptor antagonist, yohimbine, (5 mg/kg, subcutaneous) completely prevented the antiallodynic effects and the increase in IENFs observed in mice treated repetitively with DBV. PERSPECTIVE: We showed that repetitive acupoint stimulation with DBV gradually and significantly reduced oxaliplatin-induced mechanical allodynia and restored the loss of IENFs in neuropathic mice via an α-2 adrenoceptor mechanism. Collectively, results of this study suggest that repetitive acupoint treatment with DBV can be a potential strategy for the management of chemotherapy-induced neuropathy.


Asunto(s)
Terapia por Acupuntura/métodos , Analgésicos/administración & dosificación , Venenos de Abeja/administración & dosificación , Hiperalgesia/terapia , Fibras Nerviosas/patología , Enfermedades del Sistema Nervioso Periférico/terapia , Puntos de Acupuntura , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , Modelos Animales de Enfermedad , Epidermis/efectos de los fármacos , Epidermis/inervación , Epidermis/patología , Pie/inervación , Pie/patología , Hiperalgesia/patología , Hiperalgesia/fisiopatología , Masculino , Ratones Endogámicos C57BL , Compuestos Organoplatinos , Oxaliplatino , Dolor/patología , Dolor/fisiopatología , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Enfermedades del Sistema Nervioso Periférico/patología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Distribución Aleatoria , Receptores Adrenérgicos alfa 2/metabolismo , Tacto , Yohimbina/farmacología
20.
Int J Cancer ; 138(10): 2466-76, 2016 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-26704560

RESUMEN

Cancer chemotherapy with platinum-based antineoplastic agents including oxaliplatin frequently results in a debilitating and painful peripheral neuropathy. We evaluated the antinociceptive effects of the alpha-2 adrenoceptor agonist, clonidine on oxaliplatin-induced neuropathic pain. Specifically, we determined if (i) the intraperitoneal (i.p.) injection of clonidine reduces mechanical allodynia in mice with an oxaliplatin-induced neuropathy and (ii) concurrent inhibition of p38 mitogen-activated protein kinase (MAPK) activity by the p38 MAPK inhibitor SB203580 enhances clonidine's antiallodynic effect. Clonidine (0.01-0.1 mg kg(-1), i.p.), with or without SB203580(1-10 nmol, intrathecal) was administered two weeks after oxaliplatin injection(10 mg kg(-1), i.p.) to mice. Mechanical withdrawal threshold, motor coordination and blood pressure were measured. Postmortem expression of p38 MAPK and ERK as well as their phosphorylated forms(p-p38 and p-ERK) were quantified 30 min or 4 hr after drug injection in the spinal cord dorsal horn of treated and control mice. Clonidine dose-dependently reduced oxaliplatin-induced mechanical allodynia and spinal p-p38 MAPK expression, but not p-ERK. At 0.1 mg kg(-1), clonidine also impaired motor coordination and decreased blood pressure. A 10 nmol dose of SB203580 alone significantly reduced mechanical allodynia and p-p38 MAPK expression, while a subeffective dose(3 nmol) potentiated the antiallodynic effect of 0.03 mg kg(-1) clonidine and reduced the increased p-p38 MAPK. Coadministration of SB203580 and 0.03 mg kg(-1) clonidine decreased allodynia similar to that of 0.10 mg kg(-1) clonidine, but without significant motor or vascular effects. These findings demonstrate that clonidine treatment reduces oxaliplatin-induced mechanical allodynia. The concurrent administration of SB203580 reduces the dosage requirements for clonidine, thereby alleviating allodynia without producing undesirable motor or cardiovascular effects.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Antineoplásicos/efectos adversos , Clonidina/farmacología , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Compuestos Organoplatinos/efectos adversos , Animales , Presión Sanguínea/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hiperalgesia/diagnóstico , Hiperalgesia/tratamiento farmacológico , Imidazoles/farmacología , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Oxaliplatino , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Asta Dorsal de la Médula Espinal/efectos de los fármacos , Asta Dorsal de la Médula Espinal/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA