Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Neuropharmacology ; 205: 108907, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34856203

RESUMO

Pioglitazone, an agonist at peroxisome proliferator-activated receptor gamma, is FDA-approved for the treatment of insulin resistance in type 2 diabetes. Numerous studies in male rodents suggest that pioglitazone inhibits inflammatory and neuropathic pain, but few included female subjects. To address this gap, we compared the effects of pioglitazone in both sexes in the intraplantar methylglyoxal model (MG) model of chemical pain and painful diabetic neuropathy (PDN), the plantar incision model (PIM) of postoperative pain, the spared nerve injury (SNI) model of traumatic nerve injury, and the ZDF rat and db/db mouse models of PDN. We administered pioglitazone by one-time intrathecal or intraperitoneal injection or by adding it to chow for 6 weeks, followed by measurement of hypersensitivity to non-noxious mechanical, noxious mechanical, heat, and/or cold stimuli. In all mouse models, injection of pioglitazone decreased pain-like behaviors with greater potency and/or efficacy in females as compared to males: heat and mechanical hypersensitivity in the MG model (0.1-10 mg/kg); mechanical hypersensitivity in the PIM model (10 µg); mechanical and cold hypersensitivity in the SNI model (100 mg/kg); and heat hypersensitivity in the db/db model (100 mg/kg). Furthermore, co-administration of low doses of morphine (1 mg/kg) and pioglitazone (10 mg/kg) decreased SNI-induced mechanical and cold hypersensitivity in female but not male mice. In the ZDF rat, pioglitazone (100 mg/kg) decreased heat and mechanical hypersensitivity with no sex difference. In the db/db model, pioglitazone had no effect when given into chow for 6 weeks at 0.3, 3 or 30 mg/kg doses. We conclude that females exhibit greater anti-hyperalgesic responses to pioglitazone in mouse models of chemical-induced nociception, postsurgical pain, neuropathic pain, and PDN. These findings set the stage for clinical trials to determine whether pioglitazone has analgesic properties across a broad spectrum of chronic pain conditions, particularly in women.


Assuntos
Analgésicos/farmacologia , Hiperalgesia/tratamento farmacológico , Neuralgia/tratamento farmacológico , Dor Nociceptiva/tratamento farmacológico , PPAR gama/agonistas , Dor Pós-Operatória/tratamento farmacológico , Pioglitazona/farmacologia , Analgésicos/administração & dosagem , Animais , Neuropatias Diabéticas/complicações , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Morfina/farmacologia , Neuralgia/etiologia , Dor Nociceptiva/induzido quimicamente , Dor Pós-Operatória/etiologia , Traumatismos dos Nervos Periféricos/complicações , Pioglitazona/administração & dosagem , Aldeído Pirúvico/farmacologia , Caracteres Sexuais
2.
Neuroscience ; 151(4): 948-54, 2008 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-18222040

RESUMO

The medial thalamus contains abundant mu-opioid receptors and is activated by acute morphine administration. However, the role of the medial thalamus in the rewarding effects of morphine is unclear. The present study examined whether mu-opioid receptors of the medial thalamus influenced the acquisition and expression of morphine-induced conditioned place preference (CPP) in rats. An unbiased apparatus and biased subject assignment were used. Administration of morphine in increasing doses (2 mg/kg, 4 mg/kg, 6 mg/kg, 10 mg/kg, s.c.) was paired with an initially non-preferred chamber and saline administration was paired with an initially preferred chamber. Conditioning trials were conducted twice daily for 4 days. Microinjection of the irreversible mu-opioid receptor antagonist, beta-funaltrexamine (5 microg/rat), into the medial thalamus 23 h prior to each morphine conditioning completely blocked the acquisition of CPP. However, microinjection of beta-funaltrexamine into the medial thalamus after morphine conditioning trials, but 23 h prior to a test session, had no effect on the expression of CPP. It is concluded that mu-opioid receptors in the rat medial thalamus are involved in the acquisition, but not expression, of morphine-induced CPP.


Assuntos
Condicionamento Operante/efeitos dos fármacos , Morfina/farmacologia , Entorpecentes/farmacologia , Receptores Opioides/fisiologia , Tálamo/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Condicionamento Operante/fisiologia , Vias de Administração de Medicamentos , Interações Medicamentosas , Masculino , Microinjeções/métodos , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Sprague-Dawley , Tálamo/fisiologia
3.
Nutrition ; 24(9): 885-91, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18725085

RESUMO

OBJECTIVE: Recent pharmacologic studies in our laboratory have suggested that the spinal neuropeptide Y (NPY) Y1 receptor contributes to pain inhibition and to the analgesic effects of NPY. To rule out off-target effects, the present study used Y1-receptor-deficient (-/-) mice to further explore the contribution of Y1 receptors to pain modulation. METHODS AND RESULTS: Y1(-/-) mice exhibited reduced latency in the hotplate test of acute pain and a longer-lasting heat allodynia in the complete Freund's adjuvant (CFA) model of inflammatory pain. Y1 deletion did not change CFA-induced inflammation. Upon targeting the spinal NPY systems with intrathecal drug delivery, NPY reduced tactile and heat allodynia in the CFA model and the partial sciatic nerve ligation model of neuropathic pain. Importantly, we show for the first time that NPY does not exert these anti-allodynic effects in Y1(-/-) mice. Furthermore, in nerve-injured CD1 mice, concomitant injection of the potent Y1 antagonist BIBO3304 prevented the anti-allodynic actions of NPY. Neither NPY nor BIBO3304 altered performance on the Rotorod test, arguing against an indirect effect of motor function. CONCLUSION: The Y1 receptor contributes to pain inhibition and to the analgesic effects of NPY.


Assuntos
Hiperalgesia/fisiopatologia , Neuropeptídeo Y/farmacologia , Dor/fisiopatologia , Receptores de Neuropeptídeo Y/fisiologia , Analgésicos/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Arginina/administração & dosagem , Arginina/análogos & derivados , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Temperatura Alta/efeitos adversos , Hiperalgesia/tratamento farmacológico , Hiperalgesia/genética , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Neuropeptídeo Y/genética , Dor/tratamento farmacológico , Dor/genética , Medição da Dor/métodos , Medição da Dor/estatística & dados numéricos , Neuropatia Ciática/tratamento farmacológico , Neuropatia Ciática/genética , Neuropatia Ciática/fisiopatologia
4.
Neuroscience ; 334: 148-159, 2016 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-27520081

RESUMO

Trigeminal neuropathic pain is described as constant excruciating facial pain. The study goal was to investigate the role of nucleus locus coeruleus (LC) in a model of chronic orofacial neuropathic pain (CCI-ION). The study examines LC's relationship to both the medullary dorsal horn receiving trigeminal nerve sensory innervation and the medial prefrontal cortex (mPFC). LC is a major source of CNS noradrenaline (NA) and a primary nucleus involved in pain modulation. Although descending inhibition of acute pain by LC is well established, contribution of the LC to facilitation of chronic neuropathic pain is also reported. In the present study, a rat orofacial pain model of trigeminal neuropathy was induced by chronic constrictive injury of the infraorbital nerve (CCI-ION). Orofacial neuropathic pain was indicated by development of whisker pad mechanical hypersensitivity. Hypersensitivity was alleviated by selective elimination of NA neurons, including LC (A6 cell group), with the neurotoxin anti-dopamine-ß-hydroxylase saporin (anti-DßH-saporin) microinjected either intracerebroventricularly (i.c.v.) or into trigeminal spinal nucleus caudalis (spVc). The GABAA receptor antagonist, bicuculline, administered directly into LC (week 8) inhibited hypersensitivity. This indicates a valence shift in which increased GABAA signaling ongoing in LC after trigeminal nerve injury paradoxically produces excitatory facilitation of the chronic pain state. Microinjection of NAα1 receptor antagonist, benoxathian, into mPFC attenuated whisker pad hypersensitivity, while NAα2 receptor antagonist, idazoxan, was ineffective. Thus, GABAA-mediated activation of NA neurons during CCI-ION can facilitate hypersensitivity through NAα1 receptors in the mPFC. These data indicate LC is a chronic pain generator.


Assuntos
Dor Crônica/metabolismo , Dor Facial/metabolismo , Locus Cerúleo/metabolismo , Neuralgia/metabolismo , Receptores de GABA-A/metabolismo , Traumatismos do Nervo Trigêmeo/metabolismo , Fator 3 Ativador da Transcrição/metabolismo , Neurônios Adrenérgicos/efeitos dos fármacos , Neurônios Adrenérgicos/metabolismo , Antagonistas Adrenérgicos alfa/farmacologia , Animais , Bicuculina/farmacologia , Dor Crônica/tratamento farmacológico , Modelos Animais de Doenças , Dor Facial/tratamento farmacológico , Antagonistas de Receptores de GABA-A/farmacologia , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Idazoxano/farmacologia , Locus Cerúleo/efeitos dos fármacos , Masculino , Neuralgia/tratamento farmacológico , Oxati-Inas/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Traumatismos do Nervo Trigêmeo/tratamento farmacológico
5.
Endocrinology ; 139(5): 2407-13, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9564852

RESUMO

Although glucocorticoids inhibit inflammation and are used to treat painful inflammatory rheumatic diseases, the contribution, if any, of endogenous pituitary-adrenocortical activity to the control of pain remains unclear. We report that injection of dilute formalin into the hindpaw not only evokes inflammation and pain-related behavior, but it also increases ACTH and corticosterone to a greater extent than restraint and saline injection alone. This difference was particularly robust during the final periods of pain-related behavior in the formalin test, when the ACTH and corticosterone (B) levels in the restraint/saline control group had returned to normal. These results indicate that formalin-evoked increases in ACTH and B reflect nociceptive input, rather than the stress associated with handling. To test the hypothesis that the formalin-induced increase in corticosterone reduces pain and inflammation, we next evaluated the effect of adrenalectomy (to prevent activation of glucocorticoid receptors) or high-dose dexamethasone (to saturate glucocorticoid receptors) on nociceptive processing in the formalin test. Neither adrenalectomy nor dexamethasone changed behavioral or cardiovascular nociceptive responses. Furthermore, the increases in blood pressure and heart rate produced by formalin may not be mediated by adrenomedullary catecholamine release. In addition, we conclude that the nociceptive component of the formalin stimulus is sufficient to activate the pituitary-adrenocortical system in the awake rat, but that the resulting release of corticosterone does not feed back and reduce nociceptive processing.


Assuntos
Córtex Suprarrenal/fisiologia , Corticosterona/fisiologia , Formaldeído/farmacologia , Nociceptores/fisiologia , Hipófise/fisiologia , Adrenalectomia , Hormônio Adrenocorticotrópico/sangue , Animais , Comportamento Animal/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Corticosterona/sangue , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Inflamação/induzido quimicamente , Masculino , Dor , Ratos , Ratos Sprague-Dawley
6.
Pain ; 69(1-2): 101-10, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9060019

RESUMO

Injection of formalin in the rat hindpaw produces two phases of nociceptive behavior. Although it is generally agreed that the first phase results from direct chemical activation of nociceptive primary afferent fibers, the factors that contribute to the second phase are not established. In the present study, we monitored the expression of the c-fos protein to evaluate whether the pattern of activity of dorsal horn neurons differs as a result of ongoing afferent activity during the two phases. To selectively block the first or second phase, we respectively used remifentanil, a potent and short acting opiate agonist, and QX-314, a quaternary derivative of lidocaine, which does not cross the blood brain barrier. We also evaluated the effect of eliminating nociceptive behavior in both phases using both remifentanil and lidocaine or a combination of local anesthetics, bupivicaine and quaternary lidocaine. In all groups, formalin (5%, 50 microliters) was injected subcutaneously into the plantar surface of the hindpaw. To assess the nociceptive behavior produced by formalin, we monitored the number of flinches. Injection of remifentanil during the first phase completely blocked the first phase formalin-evoked nociceptive behavior, and had no effect on the second phase. Injection of lidocaine during the interphase completely blocked second phase nociceptive behavior. As expected, when remifentanil was administered during the first phase and lidocaine during the second phase, all formalin-evoked nociceptive behavior was blocked. The same was true for rats that received injections of bupivicaine and lidocaine during phases 1 and 2, respectively. In laminae I-II of the L4-L5 segment, the magnitude of the decrease in Fos expression was comparable for remifentanil (26.5%) and lidocaine (27.3%); the decrease was greater when both remifentanil and lidocaine were administered (50.5%), and even greater when bupivicaine and lidocaine were used (74.2%). In laminae V-VI, remifentanil, by itself, decreased c-fos expression by 39.4%; for lidocaine alone, the decrease was 58.4%. We did not observe further significant decreases when both remifentanil and lidocaine, or bupivacaine and lidocaine were injected (69.7% and 74.6%, respectively). Our results not only provide strong evidence that activity during the second phase is necessary for maintaining the maximal expression of c-fos in the spinal cord, but also reveal significant regional differences in the central patterns of activity generated during the two phases. These results also confirm our previous reports that c-fos expression is not eliminated when the behavioral manifestation of the noxious stimulus is completely blocked.


Assuntos
Analgésicos Opioides/farmacologia , Anestésicos Locais/farmacologia , Formaldeído , Lidocaína/farmacologia , Medição da Dor/efeitos dos fármacos , Piperidinas/farmacologia , Proteínas Proto-Oncogênicas c-fos/biossíntese , Medula Espinal/metabolismo , Animais , Bupivacaína/farmacologia , Imuno-Histoquímica , Masculino , Dor/psicologia , Proteínas Proto-Oncogênicas c-fos/genética , Ratos , Ratos Sprague-Dawley , Remifentanil , Medula Espinal/efeitos dos fármacos
7.
Pain ; 69(1-2): 171-7, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9060028

RESUMO

The opioid antagonist, naloxone, produces equivocal effects on the magnitude of nociceptive responses in several animal models of persistent pain, including the formalin test. Hindpaw injection of dilute formalin produces not only inflammation but also phasic (Phase 1) and persistent (Phase 2) behavioral and cardiovascular nociceptive responses in the rat. To test the hypothesis that endogenous opioid systems contribute to the magnitude of responses to intraplantar formalin injection, we evaluated the effects of continuous naloxone administration (0.01-100 mg/kg per h, i.v.) on formalin-evoked hindpaw inflammation, on behavioral indices of pain, flinching and licking pain behavior, and on changes in mean arterial pressure and heart rate. We report that naloxone, at doses less than 100 mg/kg per h, did not change any formalin-evoked response. Although the 100 mg/kg per h dose significantly decreased these responses, it also produced muscle rigidity and profound bradycardia. We conclude that endogenous opioids do not significantly modulate the nociceptive processing induced by subcutaneous formalin.


Assuntos
Formaldeído , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Dor/fisiopatologia , Animais , Comportamento Animal/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Formaldeído/administração & dosagem , Frequência Cardíaca/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/patologia , Infusões Intravenosas , Injeções Subcutâneas , Masculino , Naloxona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Dor/patologia , Dor/psicologia , Ratos , Ratos Sprague-Dawley
8.
Pain ; 84(2-3): 263-70, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10666531

RESUMO

Hindpaw injection of dilute formalin produces brief (Phase 1) and persistent (Phase 2) nociceptive responses in the rat. We recently showed that systemically-administered remifentanil during Phase 1 interacted with peripheral opioid receptors to delay the onset and termination of Phase 2 (Taylor et al., 1997b). To test the hypothesis that opioid inhibition of proinflammatory events during Phase 1 contributed to this delay, we evaluated the effects of remifentanil on the time course of formalin-induced inflammation. We found that formalin increased paw thickness (edema), plasma extravasation and local blood flow within minutes of its injection, i.e. during Phase 1. Each of these responses was blocked during remifentanil administration (30 microg/kg i.v. bolus, followed 90 s later with a 15 microg/kg/min infusion for 13.5 min), indicating that opioids inhibit Phase 1 inflammation. Opioid blockade of the blood flow response could be reversed with a peripherally-acting opioid antagonist, naloxone methiodide, indicating that remifentanil acted upon peripheral opioid receptors. Although the administration of remifentanil during Phase 1 did not reduce the magnitude of inflammatory responses during Phase 2, it did delay the onset and termination of edema during Phase 2. As this corresponds to the effects of remifentanil on nociceptive responses during Phase 2, we suggest that opioid analgesics act upon peripheral sites to inhibit inflammation during Phase 1, leading to a delay in the temporal profile of inflammatory (and likely nociceptive) responses during Phase 2.


Assuntos
Analgésicos Opioides/farmacologia , Permeabilidade Capilar/efeitos dos fármacos , Formaldeído/farmacologia , Membro Posterior/irrigação sanguínea , Piperidinas/farmacologia , Plasma/metabolismo , Animais , Edema/induzido quimicamente , Edema/patologia , Membro Posterior/diagnóstico por imagem , Inflamação/induzido quimicamente , Inflamação/fisiopatologia , Fluxometria por Laser-Doppler , Masculino , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/efeitos dos fármacos , Remifentanil , Temperatura Cutânea/efeitos dos fármacos , Ultrassonografia
9.
Brain Res ; 797(2): 313-20, 1998 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-9666156

RESUMO

Exposure to acute stressors activates both the hypothalamic-pituitary-adrenal (HPA) and cardiovascular systems. Prior chronic stress enhances HPA responses to novel, acute stressors, but whether it alters cardiovascular responsivity to novel, acute stress is unknown. In the present study, we examined mean arterial blood pressure (MAP) and heart rate (HR) to two distinct stimuli, restraint and formalin, following prior exposure to 7 days of intermittent cold. In two sets of control and chronically stressed animals, we measured MAP and HR for 60 min following onset of 30 min restraint and MAP, HR and behavioral responses to intraplantar injection of formalin. Chronic stress raised MAP and HR under resting conditions and elevated HR during, but not following termination of, restraint. These increases in HR during restraint were due to the differences in resting levels of HR, since both control and chronically stressed animals exhibited similar increases from resting levels in HR during restraint. Conversely, chronically stressed animals exhibited lower changes in MAP and HR from resting levels following termination of restraint. Formalin produced the characteristic biphasic pattern of cardiovascular and behavioral responses. Prior chronic stress did not alter behavior, but increased MAP and HR in Interphase and only MAP in Phase 2. The increases in MAP during Interphase and Phase 2 were a result of the elevations in resting levels of MAP, but even when differences in resting levels were taken into account, HR remained elevated in the Interphase in chronically stressed animals. Together, these data demonstrate that prior chronic intermittent cold stress modifies cardiovascular function both under resting conditions and, in very specific ways, under stimulated conditions produced by restraint and formalin. We propose that these modifications are produced by brain regions that are known to regulate cardiovascular function and which are activated by chronic stress.


Assuntos
Fenômenos Fisiológicos Cardiovasculares , Dor/fisiopatologia , Estresse Fisiológico/fisiopatologia , Hormônio Adrenocorticotrópico/sangue , Animais , Comportamento Animal/fisiologia , Pressão Sanguínea/fisiologia , Doença Crônica , Formaldeído , Frequência Cardíaca/fisiologia , Masculino , Ponte/fisiologia , Ratos , Ratos Sprague-Dawley , Restrição Física , Sistema Nervoso Simpático/fisiologia
10.
Brain Res ; 755(1): 9-16, 1997 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-9163536

RESUMO

Intraplantar injection of dilute formalin evokes brief (Phase 1) and persistent (Phase 2) increases in primary afferent activity, pain behavior, and cardiovascular responses, and induces spinal cord Fos-like immunoreactivity (Fos-LI). Although previous studies demonstrated that the destruction of small diameter primary afferents with neonatal capsaicin treatment decrease formalin-evoked nociception, these studies only evaluated behavioral responses, and did not distinguish between Phase 1 and 2. To address these questions, we simultaneously evaluated formalin-evoked pain behavior (flinching of the afflicted paw), cardiovascular responses (heart rate and mean arterial pressure), and lumbar spinal cord Fos expression in control rats and in rats treated with capsaicin (100 mg/kg) one day postpartum. We found that neonatal capsaicin-treated rats, compared to controls, exhibited similar cardiovascular responses and slightly less flinching behavior during Phase 1. During Phase 2, however, capsaicin-treated rats exhibited 59% less flinching and 45% smaller heart rate responses. Also, in capsaicin-treated rats, we counted 59% fewer Fos-labeled neurons in the spinal cord. These results indicate that capsaicin-sensitive afferents contribute to formalin-evoked behavioral and cardiovascular responses and to spinal cord neuronal responses. The differential effect of neonatal capsaicin on nociception during Phase 1 and Phase 2 suggests that sensitization mechanisms during Phase 1 do not contribute to the magnitude of nociceptive responses during Phase 2.


Assuntos
Comportamento Animal/efeitos dos fármacos , Capsaicina/farmacologia , Proteínas do Tecido Nervoso/biossíntese , Dor/induzido quimicamente , Proteínas Proto-Oncogênicas c-fos/biossíntese , Medula Espinal/efeitos dos fármacos , Vias Aferentes/efeitos dos fármacos , Animais , Pressão Sanguínea/efeitos dos fármacos , Feminino , Frequência Cardíaca/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Proteínas do Tecido Nervoso/análise , Medição da Dor/métodos , Proteínas Proto-Oncogênicas c-fos/análise , Ratos , Ratos Sprague-Dawley , Medula Espinal/metabolismo
11.
J Pain ; 1(3): 218-28, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-14622621

RESUMO

Intraplantar formalin injection produces early (Phase 1, 0- to 5-minute) and late (Phase 2, 15-plus minutes after injection) nociceptive responses, including painlike behavior and activation of primary afferents and dorsal horn neurons. Although we and others have reported that opioid analgesia or local anesthesia during Phase 1 does not reduce the overall magnitude of behavioral and/or neuronal responses during Phase 2, recent studies concluded that spinal sensitization during Phase 1 significantly contributes to the magnitude of painlike behavior during Phase 2. In this article, we provide additional evidence that Phase 1 and Phase 2 behaviors are independent. We found that remifentanil analgesia during Phase 1 does not reduce Phase 2, regardless of route of administration, duration of analgesia, types of behavior assessed, formalin concentration, concomitant use of general anesthesia, or concomitant administration of an N-methyl-D-aspartate (NMDA) antagonist. We suggest that Phase 1 behaviors compared with Phase 2 behaviors in the formalin test are not an appropriate model of spinal sensitization or preemptive opioid analgesia. Instead, early opioid administration delayed the onset of edema produced by formalin. Because the antiedema effect of remifentanil was reversed with a peripherally acting opioid receptor antagonist, we suggest that opioids interact with peripheral receptors to temporarily delay the onset and offset of formalin-induced edema.

12.
Neurosci Lett ; 291(3): 139-42, 2000 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-10984626

RESUMO

Nociceptive processing is altered in individuals with inherited hypertension. Because brainstem noradrenergic (NA) neurons have been implicated in both nociceptive transmission and hypertension, we compared behavioral and cardiovascular indices of pain in spontaneously hypertensive rats (SHR) and their normotensive Wistar-Kyoto controls (WKY) after intracerebroventricular administration of an anti-DbetaH-saporin immunotoxin. In WKY rats, NA lesions decreased indices of persistent pain in the formalin test, but did not change nociceptive responses in multiple models of acute pain. In SHR rats, NA lesions did not alter persistent nociception, but decreased thresholds in the hotplate test. We conclude that coeruleospinal inhibitory pathways modulate hypoalgesia but not hyperalgesia in the SHR rat. Brainstem noradrenergic inhibition of acute nociception in the hotplate test is enhanced in the SHR rat, but brainstem noradrenergic contribution to persistent nociceptive processing in the formalin test is reduced in the SHR rat.


Assuntos
Tronco Encefálico/metabolismo , Hipertensão/metabolismo , Norepinefrina/metabolismo , Dor/metabolismo , Ratos Endogâmicos SHR/metabolismo , Análise de Variância , Animais , Anticorpos Monoclonais , Comportamento Animal/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Tronco Encefálico/efeitos dos fármacos , Formaldeído/farmacologia , Temperatura Alta , Hipertensão/genética , Imunotoxinas/administração & dosagem , Injeções Intraventriculares , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Endogâmicos WKY , Tempo de Reação , Proteínas Inativadoras de Ribossomos Tipo 1 , Saporinas , Limiar Sensorial/efeitos dos fármacos
13.
Neurosci Lett ; 201(1): 9-12, 1995 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-8830322

RESUMO

Spontaneously hypertensive rats (SHRs) are typically less responsive to phasic noxious stimuli than are their normotensive controls. Here, we used the formalin test to compare behavioral and cardiovascular responses to persistent noxious stimuli. Hindpaw formalin injection produced exaggerated flinching, arterial pressure and heart rate responses in SHRs, suggesting that abnormalities in blood pressure control systems increase nociceptive responses to persistent noxious stimuli.


Assuntos
Comportamento Animal/fisiologia , Fenômenos Fisiológicos Cardiovasculares , Hipertensão/fisiopatologia , Nociceptores/fisiologia , Medição da Dor/efeitos dos fármacos , Ratos Endogâmicos SHR/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Sistema Cardiovascular/efeitos dos fármacos , Formaldeído/administração & dosagem , Frequência Cardíaca/fisiologia , Hipertensão/genética , Hipertensão/psicologia , Injeções Subcutâneas , Masculino , Nociceptores/efeitos dos fármacos , Ratos , Ratos Endogâmicos WKY , Restrição Física
14.
Physiol Behav ; 60(3): 919-25, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8873270

RESUMO

Repeated delivery of fast rise-time acoustic stimuli elicit cardiac changes in humans that reflect startle, orienting, and defense responses. To test the hypothesis that fast rise-time stimuli produce these responses in the rat, we evaluated magnitude, latency, and habituation of cardiovascular responses to brief airpuff stimuli in normotensive rats. We also evaluated airpuff-elicited cardiovascular responses in spontaneously hypertensive rats. In addition to a robust increase in blood pressure, airpuffs produced one or more of three sequential heart-rate responses in normotensive rats--first, short-latency tachycardia (latency ).8 s), then rapidly habituating bradycardia (latency 2.2 s), then long-latency tachycardia (latency 3.5 s)--which likely reflected startle, orienting, and defense responses, respectively. Airpuffs rarely produced bradycardia in hypertensive rats, suggesting that this strain does not appropriately orient to sensory stimuli. In addition, compared to normotensive rats, hypertensive rats exhibited greater between-session habituation of long-latency tachycardia and blood pressure increases. This finding contrasts with the Folkow hypothesis, which assumes that, in subjects with a genetic predisposition to develop hypertension, sympathetic responses will remain exaggerated after repeated stimulation, thus contributing to thickening of the arterial vasculature.


Assuntos
Pressão Sanguínea/fisiologia , Habituação Psicofisiológica/fisiologia , Frequência Cardíaca/fisiologia , Hipertensão/fisiopatologia , Animais , Masculino , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY
15.
Physiol Behav ; 49(3): 527-32, 1991 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2062929

RESUMO

Tactile (air puff) or acoustic startle stimuli elicit behavioral (motor) and complex cardiovascular responses which include pressor as well as cardiac decelerative and accelerative responses. An acoustic component of the air puff stimulus (12.5 psi) was identified. Studies were conducted to separate the contributions of both stimulus modalities to the observed responses. The acoustic component was approximated with a wide-spectrum 97-dB white-noise stimulus. This acoustic stimulus failed to evoke heart rate responses but did yield motor and pressor responses. In a second approach, tympanic membrane rupture (TMR) was used to interrupt acoustic sensory stimuli. TMR fully abolished the motor and pressor responses to acoustic startle. With air puff startle, while TMR severely attenuated the motor response it only decreased slightly the pressor and cardiac accelerative responses and failed to influence the cardiac decelerative component. Our results indicate that air puff startle contains both tactile and acoustic modalities. Further, the motor response is largely driven by the acoustic modality since TMR abolished this response elicited by either acoustic or tactile stimulation. More importantly, motor and cardiovascular responses to startle may be separated through discrimination of afferent stimuli suggesting either differences in neural pathways for acoustic and tactile stimuli or a differential dependency of the various responses on stimulus characteristics.


Assuntos
Nível de Alerta , Aprendizagem por Associação , Atenção , Percepção Auditiva , Reflexo de Sobressalto , Tato , Estimulação Acústica , Animais , Pressão Sanguínea , Frequência Cardíaca , Masculino , Estimulação Física , Ratos , Ratos Endogâmicos
16.
Ann Clin Lab Sci ; 24(6): 548-54, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7847783

RESUMO

Placental steroid hormone production appears to be critical in maintaining pregnancy and possibly initiating parturition. Cytokines, produced by activated macrophages and decidua, are present during delivery, but their role in this process is not yet clear. To our knowledge, only one recent study, which used JEG-3 choriocarcinoma cells as an in vitro model, has evaluated the possibility that cytokines might affect placental steroidogenesis. Our current study reports observations on the effect of macrophage conditioned media (MCM, known to contain several cytokines) on the synthesis of progesterone and estradiol by term, normal, human placenta. Macrophage conditioned media significantly decreased progesterone (36 percent) and increased estradiol (76 percent) production by short-term placental organ cultures. These results suggest that macrophage secretory products might significantly alter placental steroidogenesis which could make them important factors in the physiology of parturition.


Assuntos
Meios de Cultivo Condicionados/farmacologia , Citocinas/farmacologia , Estradiol/biossíntese , Placenta/metabolismo , Progesterona/biossíntese , Prostaglandinas/metabolismo , Meios de Cultivo Condicionados/química , Técnicas de Cultura , Feminino , Humanos , Macrófagos , Placenta/citologia , Placenta/efeitos dos fármacos
17.
J Gen Psychol ; 104(1st Half): 103-10, 1981 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7229629

RESUMO

Physiological responses of 44 male and female undergraduates to child abuse stimuli were recorded for the purpose of making recommendations for the selection and training of child abuse hotline counselors. The Ss' heart rate, finger pulse volume, and skin resistance were measured on a dynograph while they were listening to three case histories on a tape and viewing six slides, depicting child abuse cases. Generally, the hypotheses that women have stronger reactions than men, and adults with children have stronger reactions than adults without children were supported. It was found that men tended to be less emotionally affected by exposure to child abuse stimuli and, therefore, presumably are able to deal with it with less emotional bias. A training program consisting of desensitization, mock calls, and initial supervised participation in counseling and a follow-up habituation study were suggested.


Assuntos
Maus-Tratos Infantis , Aconselhamento , Telefone , Adolescente , Adulto , Nível de Alerta , Criança , Emoções/fisiologia , Feminino , Humanos , Masculino , Seleção de Pessoal , Fatores Sexuais , Sistema Nervoso Simpático/fisiologia
18.
Curr Top Behav Neurosci ; 20: 75-97, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24590824

RESUMO

The neuropathic pain of multiple sclerosis is quite prevalent and severely impacts quality of life. A few randomized, placebo-controlled, blinded clinical trials suggest that cannabis- and anticonvulsant-based treatments provide partial pain relief, but at the expense of adverse events. An even smaller, but emerging, number of translational studies are using rodent models of experimental autoimmune encephalomyelitis (EAE), which exhibit pain-like behaviors resembling those of Multiple sclerosis (MS) patients. These studies not only support the possible effectiveness of anticonvulsants, but also compel further clinical trials with serotonin-norepinephrine reuptake inhibitors, the immunosuppressant drug rapamycin, or drugs which interfere with glutamatergic neurotransmission. Future behavioral studies in EAE models are essential toward a new pharmacotherapy of multiple sclerosis pain.

19.
Neuroscience ; 256: 178-94, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24184981

RESUMO

Neuropeptide Y (NPY) is present in the superficial laminae of the dorsal horn and inhibits spinal nociceptive processing, but the mechanisms underlying its anti-hyperalgesic actions are unclear. We hypothesized that NPY acts at neuropeptide Y1 receptors in the dorsal horn to decrease nociception by inhibiting substance P (SP) release, and that these effects are enhanced by inflammation. To evaluate SP release, we used microdialysis and neurokinin 1 receptor (NK1R) internalization in rat. NPY decreased capsaicin-evoked SP-like immunoreactivity in the microdialysate of the dorsal horn. NPY also decreased non-noxious stimulus (paw brush)-evoked NK1R internalization (as well as mechanical hyperalgesia and mechanical and cold allodynia) after intraplantar injection of carrageenan. Similarly, in rat spinal cord slices with dorsal root attached, [Leu(31), Pro(34)]-NPY inhibited dorsal root stimulus-evoked NK1R internalization. In rat dorsal root ganglion neurons, Y1 receptors colocalized extensively with calcitonin gene-related peptide (CGRP). In dorsal horn neurons, Y1 receptors were extensively expressed and this may have masked the detection of terminal co-localization with CGRP or SP. To determine whether the pain inhibitory actions of Y1 receptors are enhanced by inflammation, we administered [Leu(31), Pro(34)]-NPY after intraplantar injection of complete Freund's adjuvant (CFA) in rat. We found that [Leu(31), Pro(34)]-NPY reduced paw clamp-induced NK1R internalization in CFA rats but not uninjured controls. To determine the contribution of increased Y1 receptor-G protein coupling, we measured [(35)S]GTPγS binding simulated by [Leu(31), Pro(34)]-NPY in mouse dorsal horn. CFA inflammation increased the affinity of Y1 receptor G-protein coupling. We conclude that Y1 receptors contribute to the anti-hyperalgesic effects of NPY by mediating the inhibition of SP release, and that Y1 receptor signaling in the dorsal horn is enhanced during inflammatory nociception.


Assuntos
Hiperalgesia/etiologia , Inflamação/complicações , Neurônios Aferentes/fisiologia , Neuropeptídeo Y/metabolismo , Receptores de Neuropeptídeo Y/metabolismo , Medula Espinal/patologia , Substância P/metabolismo , Animais , Adjuvante de Freund/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Hiperalgesia/patologia , Técnicas In Vitro , Inflamação/induzido quimicamente , Inflamação/patologia , Masculino , Neurônios Aferentes/efeitos dos fármacos , Medição da Dor , Limiar da Dor/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores da Neurocinina-1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Raízes Nervosas Espinhais/fisiologia , Isótopos de Enxofre/farmacocinética
20.
Neuropharmacology ; 70: 236-46, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23415633

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARγ) is emerging as a new pharmacotherapeutic target for chronic pain. When oral (3-30 mg/kg/day in chow for 7 wk) or twice-daily intraperitoneal (1-10 mg/kg/day for 2 wk) administration began before spared nerve injury (SNI), pioglitazone, a PPARγ agonist, dose-dependently prevented multiple behavioral signs of somatosensory hypersensitivity. The highest dose of intraperitoneal pioglitazone did not produce ataxia or reductions in transient mechanical and heat nociception, indicating that inhibitory effects on hypersensitivity were not secondary to adverse drug-induced behaviors or antinociception. Inhibitory effects on hypersensitivity persisted at least one week beyond cessation of pioglitazone administration, suggestive of long-lasting effects on gene expression. Blockade of PPARγ with GW9662, an irreversible and selective PPARγ antagonist, dose-dependently reduced the inhibitory effect of pioglitazone on hypersensitivity, indicating a PPARγ-dependent action. Remarkably, a single preemptive injection of pioglitazone 15 min before SNI attenuated hypersensitivity for at least 2 weeks; this was enhanced with a second injection delivered 12 h after SNI. Pioglitazone injections beginning after SNI also reduced hypersensitivity, albeit to a lesser degree than preemptive treatment. Intraperitoneal pioglitazone significantly reduced the nerve injury-induced up-regulation of cd11b, GFAP, and p-p38 in the dorsal horn, indicating a mechanism of action involving spinal microglia and/or astrocyte activation. Oral pioglitazone significantly reduced touch stimulus-evoked phospho-extracellular signal-related kinase (p-ERK) in lamina I-II, indicating a mechanism of action involving inhibition of central sensitization. We conclude that pioglitazone reduces spinal glial and stimulus-evoked p-ERK activation and that PPARγ activation blocks the development of and reduces established neuropathic pain.


Assuntos
Neuralgia/fisiopatologia , PPAR gama/fisiologia , Tiazolidinedionas/uso terapêutico , Anilidas/farmacologia , Animais , Ataxia/induzido quimicamente , Antígeno CD11b/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Hiperalgesia/prevenção & controle , Masculino , Neuralgia/tratamento farmacológico , Neuralgia/prevenção & controle , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , Pioglitazona , Células do Corno Posterior/efeitos dos fármacos , Células do Corno Posterior/metabolismo , Ratos , Tiazolidinedionas/administração & dosagem , Tiazolidinedionas/antagonistas & inibidores , Tiazolidinedionas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa