Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 293
Filtrar
1.
Pharmacol Biochem Behav ; 180: 22-31, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30825491

RESUMO

Clinical evidence indicates that positive allosteric modulators (PAMs) of GABAA receptors have analgesic benefit in addition to efficacy in anxiety disorders. However, the utility of GABAA receptor PAMs as analgesics is compromised by the central nervous system side effects of non-selective potentiators. A selective potentiator of GABAA receptors associated with α2/3 subunits, KRM-II-81(5-(8-ethynyl-6-(pyridin-2-yl)-4H-benzo[f]imidazo[1,5-a][1,4]diazepin-3-yl)oxazole), has demonstrated anxiolytic, anticonvulsant, and antinociceptive effects in rodents with reduced motoric side effects. The present study evaluated the potential of KRM-II-81 as a novel analgesic. Oral administration of KRM-II-81 attenuated formalin-induced flinching; in contrast, diazepam was not active. KRM-II-81 attenuated nociceptive-associated behaviors engendered by chronic spinal nerve ligation (L5/L6). Diazepam decreased locomotion of rats at the dose tested in the formalin assay (10 mg/kg) whereas KRM-II-81 produced small decreases that were not dose-dependent (10-100 mg/kg). Plasma and brain levels of KRM-II-81 were used to demonstrate selectivity for α2/3- over α1-associated GABAA receptors and to define the degree of engagement of these receptors. Plasma and brain concentrations of KRM-II-81 were positively-associated with analgesic efficacy. GABA currents from isolated rat dorsal-root ganglion cultures were potentiated by KRM-II-81 with an ED50 of 32 nM. Measures of respiratory depression were reduced by alprazolam whereas KRM-II-81 was either inactive or produced effects with lower potency and efficacy. These findings add to the growing body of data supporting the idea that α2/3-selective GABAA receptor PAMs will have efficacy and tolerability as pain medications including those for neuropathic pain. Given their predicted anxiolytic effects, α2/3-selective GABAA receptor PAMs offer an additional inroad into the management of pain.


Assuntos
Analgésicos/farmacologia , Sinergismo Farmacológico , Formaldeído/farmacologia , Oxazóis/farmacologia , Medição da Dor , Receptores de GABA-A/metabolismo , Nervos Espinhais/cirurgia , Adjuvantes Anestésicos/farmacologia , Administração Oral , Alprazolam/administração & dosagem , Alprazolam/farmacologia , Analgésicos/administração & dosagem , Analgésicos/metabolismo , Analgésicos/uso terapêutico , Animais , Comportamento Animal/efeitos dos fármacos , Diazepam/farmacologia , Relação Dose-Resposta a Droga , Moduladores GABAérgicos/administração & dosagem , Moduladores GABAérgicos/farmacologia , Ligadura , Masculino , Neuralgia/tratamento farmacológico , Oxazóis/administração & dosagem , Oxazóis/metabolismo , Oxazóis/uso terapêutico , Ratos , Ratos Sprague-Dawley
2.
J Neurophysiol ; 115(1): 208-17, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26490291

RESUMO

A number of drugs, including triptans, promote migraine chronification in susceptible individuals. In rats, a period of triptan administration over 7 days can produce "latent sensitization" (14 days after discontinuation of drug) demonstrated as enhanced sensitivity to presumed migraine triggers such as environmental stress and lowered threshold for electrically induced cortical spreading depression (CSD). Here we have used fMRI to evaluate the early changes in brain networks at day 7 of sumatriptan administration that may induce latent sensitization as well as the potential response to stress. After continuous infusion of sumatriptan, rats were scanned to measure changes in resting state networks and the response to bright light environmental stress. Rats receiving sumatriptan, but not saline infusion, showed significant differences in default mode, autonomic, basal ganglia, salience, and sensorimotor networks. Bright light stress produced CSD-like responses in sumatriptan-treated but not control rats. Our data show the first brain-related changes in a rat model of medication overuse headache and suggest that this approach could be used to evaluate the multiple brain networks involved that may promote this condition.


Assuntos
Gânglios da Base/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Depressão Alastrante da Atividade Elétrica Cortical , Agonistas do Receptor 5-HT1 de Serotonina/farmacologia , Estresse Psicológico/fisiopatologia , Sumatriptana/farmacologia , Animais , Gânglios da Base/fisiologia , Córtex Cerebral/fisiologia , Luz/efeitos adversos , Masculino , Ratos , Ratos Sprague-Dawley , Estresse Psicológico/etiologia
3.
J Pharmacol Exp Ther ; 347(1): 7-19, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23860305

RESUMO

The most highly abused prescription drugs are opioids used for the treatment of pain. Physician-reported drug-seeking behavior has resulted in a significant health concern among doctors trying to adequately treat pain while limiting the misuse or diversion of pain medications. In addition to abuse liability, opioid use is associated with unwanted side effects that complicate pain management, including opioid-induced emesis and constipation. This has resulted in restricting long-term doses of opioids and inadequate treatment of both acute and chronic debilitating pain, demonstrating a compelling need for novel analgesics. Recent reports indicate that adaptations in endogenous substance P/neurokinin-1 receptor (NK1) are induced by chronic pain and sustained opioid exposure, and these changes may contribute to processes responsible for opioid abuse liability, emesis, and analgesic tolerance. Here, we describe a multifunctional mu-/delta-opioid agonist/NK1 antagonist compound [Tyr-d-Ala-Gly-Phe-Met-Pro-Leu-Trp-NH-Bn(CF3)2 (TY027)] that has a preclinical profile of excellent antinociceptive efficacy, low abuse liability, and no opioid-related emesis or constipation. In rodent models of acute and neuropathic pain, TY027 demonstrates analgesic efficacy following central or systemic administration with a plasma half-life of more than 4 hours and central nervous system penetration. These data demonstrate that an innovative opioid designed to contest the pathology created by chronic pain and sustained opioids results in antinociceptive efficacy in rodent models, with significantly fewer side effects than morphine. Such rationally designed, multitargeted compounds are a promising therapeutic approach in treating patients who suffer from acute and chronic pain.


Assuntos
Analgésicos Opioides/administração & dosagem , Medição da Dor/efeitos dos fármacos , Dor/tratamento farmacológico , Receptores da Neurocinina-1/metabolismo , Nervos Espinhais/efeitos dos fármacos , Nervos Espinhais/lesões , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/química , Animais , Furões , Injeções Intraventriculares , Injeções Espinhais , Masculino , Camundongos , Camundongos Endogâmicos ICR , Morfina/administração & dosagem , Morfina/efeitos adversos , Naloxona/administração & dosagem , Naloxona/efeitos adversos , Dor/patologia , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley , Receptores da Neurocinina-1/fisiologia , Nervos Espinhais/patologia , Resultado do Tratamento
4.
J Neurophysiol ; 110(5): 1221-6, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23785130

RESUMO

In humans, functional magnetic resonance imaging (fMRI) activity in the anterior cingulate cortex (ACC) and the nucleus accumbens (NAc) appears to reflect affective and motivational aspects of pain. The responses of this reward-aversion circuit to relief of pain, however, have not been investigated in detail. Moreover, it is not clear whether brain processing of the affective qualities of pain in animals parallels the mechanisms observed in humans. In the present study, we analyzed fMRI blood oxygen level-dependent (BOLD) activity separately in response to an onset (aversion) and offset (reward) of a noxious heat stimulus to a dorsal part of a limb in both humans and rats. We show that pain onset results in negative activity change in the NAc and pain offset produces positive activity change in the ACC and NAc. These changes were analogous in humans and rats, suggesting that translational studies of brain circuits modulated by pain are plausible and may offer an opportunity for mechanistic investigation of pain and pain relief.


Assuntos
Giro do Cíngulo/fisiologia , Núcleo Accumbens/fisiologia , Dor/fisiopatologia , Adulto , Afeto , Animais , Humanos , Imageamento por Ressonância Magnética , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
5.
Br J Pharmacol ; 166(3): 950-63, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22122547

RESUMO

BACKGROUND AND PURPOSE: The amphibian peptide Bv8 induces potent nociceptive sensitization in rodents. Its mammalian homologue, prokineticin 2 (PROK2), is strongly up-regulated in inflamed tissues and is a major determinant in triggering inflammatory pain. Bv8 and PROK2 activate two closely related GPCRs, PK(1) and PK(2) , in a relatively non-selective fashion. To characterize better the roles of the two receptors in hyperalgesia and to obtain ligands whose binding affinity and efficacy differed for the two receptors, we modified the Bv8 molecule in regions essential for receptor recognition and activation. EXPERIMENTAL APPROACH: We modified the Bv8 molecule by substituting Trp in position 24 with Ala (A-24) and compared it with Bv8 for binding and activating PK(1) and PK(2) receptors in cell preparations and in affecting nociceptive thresholds in rodents. KEY RESULTS: A-24 preferentially bound to PK(2) receptors and activated them with a lower potency (5-fold) than Bv8. When systemically injected, A-24 induced Bv8-like hyperalgesia in rats and in mice, at doses 100 times higher than Bv8. Locally and systemically injected at inactive doses, A-24 antagonized Bv8-induced hyperalgesia. In rat and mouse models of inflammatory and post-surgical pain, A-24 showed potent and long-lasting anti-hyperalgesic activity. Unlike Bv8, A-24 increased ß-endorphin levels in mouse brain. CONCLUSIONS AND IMPLICATIONS: A-24 induced its anti-hyperalgesic effect in rodents by directly blocking nociceptor PK(1) receptors and by activating the central opioid system and the descending pain control pathway through brain PK(2) receptors.


Assuntos
Proteínas de Anfíbios/química , Proteínas de Anfíbios/farmacologia , Analgésicos/química , Analgésicos/farmacologia , Neuropeptídeos/química , Neuropeptídeos/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Alanina/química , Substituição de Aminoácidos , Proteínas de Anfíbios/uso terapêutico , Analgésicos/uso terapêutico , Animais , Células CHO , Quimiotaxia/efeitos dos fármacos , Cricetinae , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Edema/tratamento farmacológico , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Ligantes , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Neuropeptídeos/uso terapêutico , Dor Pós-Operatória/tratamento farmacológico , Dor Pós-Operatória/metabolismo , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Transfecção , Triptofano/química
6.
Br J Pharmacol ; 161(5): 986-1001, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20977451

RESUMO

BACKGROUND AND PURPOSE: The use of opioids in treating pain is limited due to significant side effects including somnolence, constipation, analgesic tolerance, addiction and respiratory depression. Pre-clinical studies have shown that neurokinin 1 (NK(1) ) receptor antagonists block opioid-induced antinociceptive tolerance and may inhibit opioid-induced rewarding behaviours. Here, we have characterized a bifunctional peptide with both opioid agonist and NK(1) antagonist pharmacophores in a rodent model of neuropathic pain. EXPERIMENTAL APPROACH: Rats were evaluated for behavioural responses to both tactile and thermal stimuli in either an uninjured, sham- or nerve-injured state. TY005 (Tyr-DAla-Gly-Phe-Met-Pro-Leu-Trp-O-3,5-Bn(CF(3) )(2) ) was delivered spinally or systemically to assess the antinociceptive effects after acute exposure. Motor skills were evaluated using the rotarod test to determine potential sedative effects. Spinal TY005 was given chronically to sham- or nerve-injured animals to determine the development of tolerance. KEY RESULTS: Bolus injections of TY005 produced dose-dependent antinociception in non-injured animals and alleviated nerve injury-induced thermal and tactile hypersensitivities (i.e. antihyperalgesia) more effectively than morphine. Sedative effects were not evident from the rotarod test at doses that were antihyperalgesic, nor at doses threefold higher. Repeated administration of TY005 did not lead to the development of antihyperalgesic tolerance or alter sensory thresholds. CONCLUSIONS AND IMPLICATIONS: Collectively, the data suggest that opioid agonist/NK(1) antagonist bifunctional peptides represent a promising novel approach to the management of chronic pain without the development of tolerance, reducing the need for escalation of doses and unwanted side effects associated with opiates alone.


Assuntos
Analgésicos Opioides/farmacologia , Tolerância a Medicamentos , Neuralgia/tratamento farmacológico , Oligopeptídeos/farmacologia , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/efeitos adversos , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Injeções Intravenosas , Injeções Espinhais , Masculino , Destreza Motora/efeitos dos fármacos , Neuralgia/fisiopatologia , Antagonistas dos Receptores de Neurocinina-1 , Oligopeptídeos/administração & dosagem , Oligopeptídeos/efeitos adversos , Medição da Dor , Ratos , Ratos Sprague-Dawley , Receptores Opioides/agonistas
7.
Med Secoli ; 22(1-3): 111-28, 2010.
Artigo em Italiano | MEDLINE | ID: mdl-21563471

RESUMO

The necropolis of Castel Malnome, chronologically framed between the I and II century AD, is located in the vicinity of Ponte Galeria (Rome), nearby the via Portuense. The excavation of the funerary site has allowed the collection of 292 inhumations, referred to the lower social classes and for the most part adult males. Regarding the funerary ritual, only the 42.8% of the graves had a tiles cover, while about one third provided grave goods. The field analysis shows that almost all the burials are primaries, and is not possible to highlight a main position of the inhumated individuals. The laboratory analysis, till today carried out on 100 individuals, shows a high degree of skeletal robustness and of occupational markers due to heavy work load (inflammation, muscle-skeletal trauma, fractures, osteoarthritis, enthesopathies). The recording of oral pathologies and aspecific stresses, in order to obtain information about the health status of the population, shows a very high frequency of caries mainly related to poor dental hygiene, and of enamel hypoplasia.


Assuntos
Nível de Saúde , Estilo de Vida/história , Saúde Ocupacional/história , Adolescente , Adulto , Feminino , História Antiga , Humanos , Masculino , Cidade de Roma , Adulto Jovem
8.
Cephalalgia ; 29(12): 1277-84, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19438917

RESUMO

Medication overuse headache (MOH) is a challenging, debilitating disorder that develops from the frequent use of medications taken for the treatment of migraine headache pain. MOH affects an estimated 3-5% of the general population. The mechanisms underlying the development of MOH remain unknown. Opiates are one of the major classes of medications used for the treatment of migraine at least in some countries, including the USA. Although the effects of repeated opiate use for headache are unknown, it is possible that opiate use may contribute to increased frequency and occurrence of such headaches. Recent preclinical studies exploring the neuroadaptive changes following sustained exposure to morphine may give some insights into possible causes of MOH. Peripherally, these changes include increased expression of calcitonin gene-related peptide (CGRP) in trigeminal primary afferent neurons. Centrally, they include increased excitatory neurotransmission at the level of the dorsal horn and nucleus caudalis. Critically, these neuroadaptive changes persist for long periods of time and the evoked release of CGRP is enhanced following morphine pretreatment. Stimuli known to elicit migraine, such as nitric oxide donors or stress, produce hyperalgesia in morphine- but not in saline-pretreated rats even long after the discontinuation of the opiate. CGRP plays a prominent role in initiating vasodilation of the intracranial blood vessels and subsequent headache. Furthermore, studies have demonstrated increased excitability of the nociceptive pathway in migraine sufferers, and CGRP receptor antagonists have been shown to be efficacious in migraine pain. Thus, such persistent neuroadaptive changes may be relevant to the processes that promote MOH.


Assuntos
Analgésicos Opioides/efeitos adversos , Transtornos da Cefaleia Secundários/fisiopatologia , Transtornos de Enxaqueca/tratamento farmacológico , Morfina/efeitos adversos , Nociceptores/efeitos dos fármacos , Nervo Trigêmeo/efeitos dos fármacos , Adaptação Fisiológica/efeitos dos fármacos , Animais , Peptídeo Relacionado com Gene de Calcitonina/fisiologia , Humanos , Hiperalgesia/induzido quimicamente , Hiperalgesia/fisiopatologia , Nociceptores/fisiologia , Ratos , Nervo Trigêmeo/fisiologia
9.
Neuroscience ; 140(4): 1311-20, 2006 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-16650614

RESUMO

Nerve injury can produce hypersensitivity to noxious and normally innocuous stimulation. Injury-induced central (i.e. spinal) sensitization is thought to arise from enhanced afferent input to the spinal cord and to be critical for expression of behavioral hypersensitivity. Descending facilitatory influences from the rostral ventromedial medulla have been suggested to also be critical for the maintenance, though not the initiation, of experimental neuropathic pain. The possibility that descending facilitation from the rostral ventromedial medulla is required for the maintenance of central sensitization was examined by determining whether ablation of mu-opioid receptor-expressing cells within the rostral ventromedial medulla prevented the enhanced expression of repetitive touch-evoked FOS within the spinal cord of animals with spinal nerve ligation injury as well as nerve injury-induced behavioral hypersensitivity. Rats received a single microinjection of vehicle, saporin, dermorphin or dermorphin-saporin into the rostral ventromedial medulla and 28 days later, underwent either sham or spinal nerve ligation procedures. Animals receiving rostral ventromedial medulla pretreatment with vehicle, dermorphin or saporin that were subjected to spinal nerve ligation demonstrated both thermal and tactile hypersensitivity, and showed significantly increased expression of touch-evoked FOS in the dorsal horn ipsilateral to nerve injury compared with sham-operated controls at days 3, 5 or 10 post-spinal nerve ligation. In contrast, nerve-injured animals pretreated with dermorphin-saporin showed enhanced behaviors and touch-evoked FOS expression in the spinal dorsal horn at day 3, but not days 5 and 10, post-spinal nerve ligation when compared with sham-operated controls. These results indicate the presence of nerve injury-induced behavioral hypersensitivity associated with nerve injury-induced central sensitization. Further, the results demonstrate the novel concept that once initiated, maintenance of nerve injury-induced central sensitization in the spinal dorsal horn requires descending pain facilitation mechanisms arising from the rostral ventromedial medulla.


Assuntos
Bulbo/fisiologia , Medição da Dor/métodos , Traumatismos dos Nervos Periféricos , Nervos Periféricos/fisiologia , Tratos Piramidais/fisiologia , Animais , Masculino , Ratos , Ratos Sprague-Dawley
10.
J Pharmacol Exp Ther ; 318(1): 195-205, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16565167

RESUMO

The antinociceptive pharmacology of N-[[4-(4,5-dihydro-1H-imidazol-2-yl)phenyl]methyl]-2-[2-[[(4-methoxy-2,6-dimethylphenyl) sulfonyl]methylamino]ethoxy]-N-methylacetamide fumarate (LF22-0542), a novel nonpeptidic B1 antagonist, was characterized. LF22-0542 showed high affinity for human and mouse B1 receptors with virtually no affinity for the human B2 receptor; a selectivity index of at least 4000 times was obtained when LF22-0542 was profiled throughout binding or cell biology assays on 64 other G-protein-coupled receptor, 10 ion channels, and seven enzymes. LF22-0542 was a competitive B1 receptor antagonist and elicited significant antinociceptive actions in the mouse acetic acid-induced writhing assay, as well as in the second phases of formalin-induced nociception in mice and in both the first and second phases of the formalin response in rats. LF22-0542 was active after s.c. but not p.o. administration. In B1 receptor knockout (KO) mice, acetic acid and formalin responses were significantly reduced and LF22-0542 had no additional effects in these animals. LF22-0542 alleviated thermal hypersensitivity in both acute (carrageenan) and persistent inflammatory (complete Freund's adjuvant) pain models in rats. LF22-0542 produced a full reversal of experimental neuropathic thermal hypersensitivity but was inactive in reversing nerve injury-induced tactile hypersensitivity in rats. In agreement with this observation, B1 KO mice subjected to peripheral nerve injury did not show thermal hypersensitivity but developed nerve injury-induced tactile hypersensitivity normally. The data demonstrate the antihyperalgesic actions of a selective systemically administered B1 receptor antagonist and suggest the utility of this class of agents for the treatment of inflammatory pain states and for some aspects of neuropathic pain.


Assuntos
Acrilamidas/farmacologia , Analgésicos/farmacologia , Antagonistas de Receptor B1 da Bradicinina , Fumaratos/farmacologia , Medição da Dor/efeitos dos fármacos , Acrilamidas/química , Analgésicos/química , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Fumaratos/química , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptor B1 da Bradicinina/fisiologia
11.
Neuroscience ; 123(1): 43-52, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14667440

RESUMO

Several experimental models of peripheral neuropathy show that a significant upregulation of spinal dynorphin A and its precursor peptide, prodynorphin, is a common consequence of nerve injury. A genetically modified mouse strain lacking prodynorphin does not exhibit sustained neuropathic pain after nerve injury, supporting a pronociceptive role of elevated levels of spinal dynorphin. A null mutation of the gamma isoform of protein kinase C (PKCgamma KO [knockout]), as well as an inbred mouse strain, 129S6, also does not manifest behavioral signs of neuropathic pain following peripheral nerve injury. The objective of this study was to extend our observations to these genetic models to test the hypothesis that elevated levels of spinal dynorphin are essential for the maintenance of abnormal pain. In PKCgamma wild-type mice and the outbred mouse strain ICR, ligation of the L5 and L6 spinal nerves (SNL) elicited both tactile hypersensitivity and thermal hyperalgesia. Both strains showed a significant elevation in dynorphin in the lumbar spinal dorsal horn following SNL. Spinal administration of an anti-dynorphin A antiserum blocked the thermal and tactile hypersensitivity in both strains of mice. However, the PKCgamma KO mice and the 129S6 mice (which express PKCgamma) did not show abnormal pain after SNL; neither strain showed elevated levels of spinal dynorphin. The multiple phenotypic deficits in PKCgamma KO mice confound the interpretation of the proposed role of PKCgamma-expressing spinal neurons in neuropathic pain states. Additionally, the data show that the regulation of spinal dynorphin expression is a common critical feature of expression of neuropathic pain.


Assuntos
Dinorfinas/biossíntese , Traumatismos dos Nervos Periféricos , Doenças do Sistema Nervoso Periférico/metabolismo , Medula Espinal/metabolismo , Regulação para Cima/fisiologia , Animais , Dinorfinas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Nervos Periféricos/efeitos dos fármacos , Nervos Periféricos/metabolismo , Proteína Quinase C/biossíntese , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Especificidade da Espécie , Medula Espinal/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
12.
Neuroscience ; 121(3): 815-24, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14568039

RESUMO

Glial cell line-derived neurotrophic factor (GDNF) is necessary for the development of sensory neurons, and appears to be critical for the survival of dorsal root ganglion (DRG) cells that bind the lectin IB4. Intrathecal infusion of GDNF has been shown to prevent and reverse the behavioral expression of experimental neuropathic pain arising from injury to spinal nerves. This effect of GDNF has been attributed to a blockade of the expression of the voltage gated, tetrodotoxin-sensitive sodium channel subtype, Na(V)1.3, in the injured DRG. Here we report that GDNF given intrathecally via osmotic-pump to nerve-injured rats (L5/L6 spinal nerve ligation) prevented the changes in a variety of neurochemical markers in the DRG upon injury. They include a loss of binding of IB4, downregulation of the purinergic receptor P2X(3), upregulation of galanin and neuropeptide Y immunoreactivity in large diameter DRG cells, and expression of the transcription factor ATF3. GDNF infusion concomitantly prevented the development of spinal nerve ligation-induced tactile hypersensitivity and thermal hyperalgesia. These observations suggest that high dose, exogenous GDNF has a broad neuroprotective role in injured primary afferent. The receptor(s) that mediates these effects of GDNF is not known. GDNF's ability to block neuropathic pain states is not likely to be specific to Na(V)1.3 expression.


Assuntos
Gânglios Espinais/lesões , Fatores de Crescimento Neural/metabolismo , Neurônios/metabolismo , Dor/metabolismo , Fatores Ativadores da Transcrição , Animais , Proteínas Sanguíneas/metabolismo , Modelos Animais de Doenças , Lateralidade Funcional , Galanina/metabolismo , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Humanos , Imuno-Histoquímica , Lectinas/metabolismo , Masculino , Neuropeptídeo Y/metabolismo , Dor/patologia , Dor/prevenção & controle , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X3 , Fatores de Tempo , Fatores de Transcrição/metabolismo
13.
J Neurosci ; 23(23): 8370-9, 2003 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-12967999

RESUMO

Nerve injury-induced afferent discharge is thought to elicit spinal sensitization and consequent abnormal pain. Experimental neuropathic pain, however, also depends on central changes, including descending facilitation arising from the rostral ventromedial medulla (RVM) and upregulation of spinal dynorphin. A possible intersection of these influences at the spinal level was explored by measuring evoked, excitatory transmitter release in tissues taken from nerve-injured animals with or without previous manipulation of descending modulatory systems. Spinal nerve ligation (SNL) produced expected tactile and thermal hyperesthesias. Capsaicin-evoked calcitonin gene-related peptide (CGRP) release was markedly enhanced in lumbar spinal tissue from SNL rats when compared with sham-operated controls. Enhanced, evoked CGRP release from SNL rats was blocked by anti-dynorphin A(1-13) antiserum; this treatment did not alter evoked release in tissues from sham-operated rats. Dorsolateral funiculus lesion (DLF) or destruction of RVM neurons expressing mu-opioid receptors with dermorphin-saporin, blocked tactile and thermal hypersensitivity, as well as SNL-induced upregulation of spinal dynorphin. Spinal tissues from these DLF-lesioned or dermorphin-saporin-treated SNL rats did not exhibit enhanced capsaicin-evoked CGRP-IR release. These data demonstrate exaggerated release of excitatory transmitter from primary afferents after injury to peripheral nerves, supporting the likely importance of increased afferent input as a driving force of neuropathic pain. The data also show that modulatory influences of descending facilitation are required for enhanced evoked transmitter release after nerve injury. Thus, convergence of descending modulation, spinal plasticity, and afferent drive in the nerve-injured state reveals a mechanism by which some aspects of nerve injury-induced hyperesthesias may occur.


Assuntos
Vias Aferentes/fisiopatologia , Síndromes de Compressão Nervosa/fisiopatologia , Neuralgia/fisiopatologia , Neurotransmissores/metabolismo , Nervos Espinhais/fisiopatologia , Vias Aferentes/metabolismo , Analgésicos Opioides/farmacologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Capsaicina/farmacologia , Modelos Animais de Doenças , Dinorfinas/metabolismo , Encefalinas/metabolismo , Hiperestesia/etiologia , Hiperestesia/fisiopatologia , Ligadura , Região Lombossacral , Masculino , Bulbo/efeitos dos fármacos , Bulbo/fisiopatologia , Microinjeções , N-Glicosil Hidrolases , Síndromes de Compressão Nervosa/complicações , Neuralgia/etiologia , Plasticidade Neuronal , Oligopeptídeos/farmacologia , Peptídeos Opioides , Medição da Dor/efeitos dos fármacos , Precursores de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Inativadoras de Ribossomos Tipo 1 , Saporinas , Limiar Sensorial/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Nervos Espinhais/metabolismo
14.
Life Sci ; 73(6): 699-704, 2003 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-12801591

RESUMO

Disease states such as neuropathic pain offer special challenges in drug design due to the system changes which accompany these diseases. In this manuscript we provide an example of a new approach to drug design in which we have modified a potent and selective peptide ligand for the CCK-2 receptor to a peptide which has potent agonist binding affinity and bioactivity at delta and mu opioid receptors, and simultaneous antagonist activity at CCK receptors. De novo design based on the concept of overlapping pharmacophores was a central hypothesis of this design, and led to compounds such as H-Tyr-DPhe-Gly-DTrp-NMeNle-Asp-Phe-NH(2) (i.e., RSA 601) which have the designed properties.


Assuntos
Desenho de Fármacos , Oligopeptídeos/farmacologia , Dor/tratamento farmacológico , Receptores da Colecistocinina/antagonistas & inibidores , Receptores Opioides/agonistas , Animais , Sítios de Ligação , Cobaias , Íleo/efeitos dos fármacos , Íleo/metabolismo , Ligantes , Masculino , Camundongos , Oligopeptídeos/química , Dor/metabolismo , Receptor de Colecistocinina B , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Relação Estrutura-Atividade , Ducto Deferente/efeitos dos fármacos , Ducto Deferente/metabolismo
15.
Pain ; 100(3): 243-248, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12467995

RESUMO

Spinal antinociception produced by delta 9-tetrahydro-cannabinol (Delta(9)-THC) and other cannabinoid agonists has been suggested to be mediated by the release of dynorphin acting at the kappa opioid receptor. Alternatively, as cannabinoid receptors are distributed appropriately in the pain transmission pathway, cannabinoid agonists might act directly at the spinal level to inhibit nociception, without requiring dynorphin release. Here, these possibilities were explored using mice with a deletion of the gene encoding prodynorphin. Antinociceptive dose-response curves were constructed for spinal Delta(9)-THC and WIN 55,212-2 in prodynorphin knock-out mice and in wild-type littermates. WIN 55,212-2 and Delta(9)-THC were equipotent in the wild-type and prodynorphin knock-out mice. Spinal pretreatment with a kappa opioid receptor antagonist, nor-binaltorphimine (nor-BNI), did not alter the dose-response curves for either WIN 55,212-2 or Delta(9)-THC in prodynorphin knock-out and wild-type mice. However, the same dose of nor-BNI used blocked U50,488H-induced antinociception in both wild-type and prodynorphin knock-out mice, confirming kappa opioid receptor activity. Pretreatment with SR141716A, a cannabinoid receptor antagonist blocked the antinociceptive actions of both WIN 55,212-2 and Delta(9)-THC. These data support the conclusion that antinociception produced by spinal cannabinoids are likely to be mediated directly through activation of cannabinoid receptors without the requirement for dynorphin release or activation of kappa opioid receptors.


Assuntos
Dronabinol/administração & dosagem , Dinorfinas/deficiência , Morfolinas/administração & dosagem , Naltrexona/análogos & derivados , Naftalenos/administração & dosagem , Medição da Dor/efeitos dos fármacos , Dor/tratamento farmacológico , Medula Espinal/efeitos dos fármacos , Analgésicos/administração & dosagem , Analgésicos não Narcóticos/administração & dosagem , Animais , Benzoxazinas , Relação Dose-Resposta a Droga , Injeções Espinhais , Masculino , Camundongos , Camundongos Knockout , Valores de Referência , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Método Simples-Cego , Especificidade da Espécie
16.
Pain ; 98(1-2): 79-88, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12098619

RESUMO

Recent studies indicate that sustained opioid administration produces increased expression of spinal dynorphin, which promotes enhanced sensitivity to non-noxious and noxious stimuli. Such increased "pain" may manifest behaviorally as a decrease in spinal antinociceptive potency. Here, the possibility of similar mechanisms in the antinociception of spinal cannabinoids was explored. Response thresholds to non-noxious mechanical and noxious thermal stimuli were assessed. Antinociception was determined using the 52 degrees C tail-flick test. Mice received repeated WIN 55,212-2, its inactive enantiomer, WIN 55,212-3 or vehicle (i.th., bid, 5 days). WIN 55,212-2, but not WIN 55,212-3 or vehicle, produced a time-related increased sensitivity to non-noxious and noxious stimuli. WIN 55,212-2, but not WIN 55,212-3 or vehicle, elicited a significant increase in lumbar spinal dynorphin content at treatment day 5. Increased sensitivity to mechanical and thermal stimuli produced by WIN 55,212-2 was reversed to baseline levels by i.th. MK-801 or dynorphin antiserum; control serum had no effect. WIN 55,212-2, but not WIN 55,212-3 or vehicle, produced dose-related antinociception and repeated administration resulted in antinociceptive tolerance. While MK-801 and dynorphin antiserum did not alter acute antinociception produced by WIN 55,212-2, these substances significantly blocked antinociceptive tolerance when given immediately prior to WIN 55,212-2 challenge on day 5. Daily MK-801 pretreatments, prior to WIN 55,212-2 injection, also produced a significant block of antinociceptive tolerance. These data suggest that like opioids, repeated spinal administration of a cannabinoid CB1 agonist elicits abnormal pain, which results in increased expression of spinal dynorphin. Manipulations that block cannabinoid-induced pain also block the behavioral manifestation of cannabinoid tolerance.


Assuntos
Analgésicos/farmacologia , Canabinoides/farmacologia , Dinorfinas/administração & dosagem , Morfolinas/farmacologia , Naftalenos/farmacologia , Nociceptores/efeitos dos fármacos , Nociceptores/fisiologia , Dor/induzido quimicamente , Animais , Anticorpos/imunologia , Benzoxazinas , Reações Cruzadas , Sinergismo Farmacológico , Tolerância a Medicamentos , Dinorfinas/metabolismo , Dinorfinas/farmacologia , Temperatura Alta , Técnicas Imunoenzimáticas , Injeções Espinhais , Isomerismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Limiar da Dor/efeitos dos fármacos , Estimulação Física , Medula Espinal/metabolismo
17.
Proc Natl Acad Sci U S A ; 98(23): 13373-8, 2001 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-11698689

RESUMO

Differential expression of ion channels contributes functional diversity to sensory neuron signaling. We find nerve injury induced by the Chung model of neuropathic pain leads to striking reductions in voltage-gated K(+) (Kv) channel subunit expression in dorsal root ganglia (DRG) neurons, suggesting a potential molecular mechanism for hyperexcitability of injured nerves. Moreover, specific classes of DRG neurons express distinct Kv channel subunit combinations. Importantly, Kv1.4 is the sole Kv1 alpha subunit expressed in smaller diameter neurons, suggesting that homomeric Kv1.4 channels predominate in A delta and C fibers arising from these cells. These neurons are presumably nociceptors, because they also express the VR-1 capsaicin receptor, calcitonin gene-related peptide, and/or Na(+) channel SNS/PN3/Nav1.8. In contrast, larger diameter neurons associated with mechanoreception and proprioception express high levels of Kv1.1 and Kv1.2 without Kv1.4 or other Kv1 alpha subunits, suggesting that heteromers of these subunits predominate on large, myelinated afferent axons that extend from these cells.


Assuntos
Neurônios Aferentes/fisiologia , Dor/fisiopatologia , Canais de Potássio/fisiologia , Animais , Imunofluorescência , Gânglios Espinais/metabolismo , Gânglios Espinais/fisiologia , Masculino , Neurônios Aferentes/metabolismo , Canais de Potássio/metabolismo , Ratos , Ratos Sprague-Dawley , Frações Subcelulares/metabolismo
18.
J Neurosci ; 21(14): 5281-8, 2001 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-11438603

RESUMO

Neurons in the rostroventromedial medulla (RVM) project to spinal loci where the neurons inhibit or facilitate pain transmission. Abnormal activity of facilitatory processes may thus represent a mechanism of chronic pain. This possibility and the phenotype of RVM cells that might underlie experimental neuropathic pain were investigated. Cells expressing mu-opioid receptors were targeted with a single microinjection of saporin conjugated to the mu-opioid agonist dermorphin; unconjugated saporin and dermorphin were used as controls. RVM dermorphin-saporin, but not dermorphin or saporin, significantly decreased cells expressing mu-opioid receptor transcript. RVM dermorphin, saporin, or dermorphin-saporin did not change baseline hindpaw sensitivity to non-noxious or noxious stimuli. Spinal nerve ligation (SNL) injury in rats pretreated with RVM dermorphin-saporin failed to elicit the expected increase in sensitivity to non-noxious mechanical or noxious thermal stimuli applied to the paw. RVM dermorphin or saporin did not alter SNL-induced experimental pain, and no pretreatment affected the responses of sham-operated groups. This protective effect of dermorphin-saporin against SNL-induced pain was blocked by beta-funaltrexamine, a selective mu-opioid receptor antagonist, indicating specific interaction of dermorphin-saporin with the mu-opioid receptor. RVM microinjection of dermorphin-saporin, but not of dermorphin or saporin, in animals previously undergoing SNL showed a time-related reversal of the SNL-induced experimental pain to preinjury baseline levels. Thus, loss of RVM mu receptor-expressing cells both prevents and reverses experimental neuropathic pain. The data support the hypothesis that inappropriate tonic-descending facilitation may underlie some chronic pain states and offer new possibilities for the design of therapeutic strategies.


Assuntos
Tronco Encefálico/efeitos dos fármacos , Imunotoxinas , N-Glicosil Hidrolases , Neuralgia/tratamento farmacológico , Neurônios/efeitos dos fármacos , Receptores Opioides mu/antagonistas & inibidores , Proteínas Recombinantes de Fusão/administração & dosagem , Animais , Comportamento Animal/efeitos dos fármacos , Tronco Encefálico/citologia , Tronco Encefálico/metabolismo , Modelos Animais de Doenças , Ligadura , Masculino , Bulbo/citologia , Bulbo/efeitos dos fármacos , Bulbo/metabolismo , Microinjeções , Naltrexona/administração & dosagem , Naltrexona/análogos & derivados , Neuralgia/fisiopatologia , Neurônios/metabolismo , Oligopeptídeos/administração & dosagem , Peptídeos Opioides , Medição da Dor/efeitos dos fármacos , Estimulação Física , Proteínas de Plantas/administração & dosagem , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Receptores Opioides mu/biossíntese , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/química , Proteínas Inativadoras de Ribossomos Tipo 1 , Saporinas , Nervos Espinhais/lesões , Nervos Espinhais/fisiopatologia
19.
Anesthesiology ; 94(5): 882-7, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11388542

RESUMO

BACKGROUND: Cannabinoid receptor agonists reverse nausea and vomiting produced by chemotherapy and radiation therapy in animals and humans but have not been tested against opioid-induced emesis. This study tests the hypothesis that cannabinoid receptor agonists will prevent opioid-induced vomiting. METHODS: Twelve male ferrets were used. They weighed 1.2-1.6 kg at the beginning and 1.8-2.3 kg at the end of the experiments. All drugs were injected subcutaneously. WIN55,212-2, a mixed CB1-CB2 cannabinoid receptor agonist, was administered 25 min before morphine. Retches and vomits were counted at 5-min intervals for 30 min after morphine injection. RESULTS: Retching and vomiting responses increased with increasing morphine doses up to 1.0 mg/kg, above which the responses decreased. Previous administration of naloxone prevented morphine-induced retching and vomiting. WIN55,212-2 dose-dependently reduced retching and vomiting. The ED50 was 0.05 mg/kg for retches and 0.03 mg/kg for vomits. At 0.13 mg/kg, retching decreased by 76% and vomiting by 92%. AM251, a CB1 receptor-selective antagonist, blocked the antiemetic actions of WIN55,212-2, but AM630, a CB2 receptor-selective antagonist, did not. CONCLUSIONS: These results demonstrate that WIN55,212-2 prevents opioid-induced vomiting and suggest that the antiemetic activity of WIN55,212-2 occurs at CB1 receptors. This is consistent with findings that CB1 receptors are the predominant cannabinoid receptors in the central nervous system and that antiemetic effects of cannabinoids appear to be centrally mediated.


Assuntos
Analgésicos Opioides/toxicidade , Antieméticos/farmacologia , Morfina/toxicidade , Morfolinas/farmacologia , Naftalenos/farmacologia , Receptores de Droga/agonistas , Vômito/prevenção & controle , Animais , Benzoxazinas , Furões , Masculino , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptores de Canabinoides , Receptores de Droga/fisiologia
20.
J Pept Res ; 57(4): 257-76, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11328484

RESUMO

Replacement of Phe3 in the endogenous delta-opioid selective peptide deltorphin I with four optically pure stereoisomers of the topographically constrained, highly hydrophobic novel amino acid beta-isopropylphenylalanine (beta-iPrPhe) produced four pharmacologically different deltorphin I peptidomimetics. Radiolabeled ligand-binding assays and in vitro biological evaluation indicate that the stereoconfiguration of the iPrPhe residue plays a crucial role in determining the binding affinity, bioactivity and selectivity of [beta-iPrPhe3]deltorphin I analogs: a (2S,3R) configuration of the iPrPhe3 residue in [beta-iPrPhe3]deltorphin I provided the most desirable biological properties with binding affinity (IC50 = 2 nM), bioassay potency (IC50 = 1.23 nM in MVD assay) and exceptional selectivity for the delta-opioid receptor over the mu-opioid receptor (30 000). Further conformational studies based on two-dimensional NMR and computer-assisted molecular modeling suggested a model for the possible bioactive conformation in which the Tyr1 and (2S,3R)-beta-iPrPhe3 residues adopt trans side-chain conformations, and the linear peptide backbone favors a distorted beta-turn conformation.


Assuntos
Analgésicos Opioides/química , Oligopeptídeos/química , Analgésicos Opioides/síntese química , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Simulação por Computador , Cobaias , Técnicas In Vitro , Masculino , Camundongos , Modelos Moleculares , Estrutura Molecular , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Músculo Liso/fisiologia , Ressonância Magnética Nuclear Biomolecular , Oligopeptídeos/síntese química , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Conformação Proteica , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA