Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 293
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Neurophysiol ; 115(1): 208-17, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26490291

RESUMEN

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.


Asunto(s)
Ganglios Basales/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Depresión de Propagación Cortical , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Estrés Psicológico/fisiopatología , Sumatriptán/farmacología , Animales , Ganglios Basales/fisiología , Corteza Cerebral/fisiología , Luz/efectos adversos , Masculino , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/etiología
2.
J Neurophysiol ; 110(5): 1221-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23785130

RESUMEN

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.


Asunto(s)
Giro del Cíngulo/fisiología , Núcleo Accumbens/fisiología , Dolor/fisiopatología , Adulto , Afecto , Animales , Humanos , Imagen por Resonancia Magnética , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
3.
J Pharmacol Exp Ther ; 347(1): 7-19, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23860305

RESUMEN

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.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Dimensión del Dolor/efectos de los fármacos , Dolor/tratamiento farmacológico , Receptores de Neuroquinina-1/metabolismo , Nervios Espinales/efectos de los fármacos , Nervios Espinales/lesiones , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/química , Animales , Hurones , Inyecciones Intraventriculares , Inyecciones Espinales , Masculino , Ratones , Ratones Endogámicos ICR , Morfina/administración & dosificación , Morfina/efectos adversos , Naloxona/administración & dosificación , Naloxona/efectos adversos , Dolor/patología , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley , Receptores de Neuroquinina-1/fisiología , Nervios Espinales/patología , Resultado del Tratamiento
4.
Cephalalgia ; 29(12): 1277-84, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19438917

RESUMEN

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.


Asunto(s)
Analgésicos Opioides/efectos adversos , Cefaleas Secundarias/fisiopatología , Trastornos Migrañosos/tratamiento farmacológico , Morfina/efectos adversos , Nociceptores/efectos de los fármacos , Nervio Trigémino/efectos de los fármacos , Adaptación Fisiológica/efectos de los fármacos , Animales , Péptido Relacionado con Gen de Calcitonina/fisiología , Humanos , Hiperalgesia/inducido químicamente , Hiperalgesia/fisiopatología , Nociceptores/fisiología , Ratas , Nervio Trigémino/fisiología
5.
Pharmacol Biochem Behav ; 180: 22-31, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30825491

RESUMEN

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.


Asunto(s)
Analgésicos/farmacología , Sinergismo Farmacológico , Formaldehído/farmacología , Oxazoles/farmacología , Dimensión del Dolor , Receptores de GABA-A/metabolismo , Nervios Espinales/cirugía , Adyuvantes Anestésicos/farmacología , Administración Oral , Alprazolam/administración & dosificación , Alprazolam/farmacología , Analgésicos/administración & dosificación , Analgésicos/metabolismo , Analgésicos/uso terapéutico , Animales , Conducta Animal/efectos de los fármacos , Diazepam/farmacología , Relación Dosis-Respuesta a Droga , Moduladores del GABA/administración & dosificación , Moduladores del GABA/farmacología , Ligadura , Masculino , Neuralgia/tratamiento farmacológico , Oxazoles/administración & dosificación , Oxazoles/metabolismo , Oxazoles/uso terapéutico , Ratas , Ratas Sprague-Dawley
6.
Neuroscience ; 140(4): 1311-20, 2006 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-16650614

RESUMEN

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.


Asunto(s)
Bulbo Raquídeo/fisiología , Dimensión del Dolor/métodos , Traumatismos de los Nervios Periféricos , Nervios Periféricos/fisiología , Tractos Piramidales/fisiología , Animales , Masculino , Ratas , Ratas Sprague-Dawley
8.
J Neurosci ; 21(1): 279-86, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11150345

RESUMEN

Many clinical case reports have suggested that sustained opioid exposure can elicit unexpected, paradoxical pain. Here, we explore the possibility that (1) opioid-induced pain results from tonic activation of descending pain facilitation arising in the rostral ventromedial medulla (RVM) and (2) the presence of such pain manifests behaviorally as antinociceptive tolerance. Rats implanted subcutaneously with pellets or osmotic minipumps delivering morphine displayed time-related tactile allodynia and thermal hyperalgesia (i. e., opioid-induced "pain"); placebo pellets or saline minipumps did not change thresholds. Opioid-induced pain was observed while morphine delivery continued and while the rats were not in withdrawal. RVM lidocaine, or bilateral lesions of the dorsolateral funiculus (DLF), did not change response thresholds in placebo-pelleted rats but blocked opioid-induced pain. The intrathecal morphine antinociceptive dose-response curve (DRC) in morphine-pelleted rats was displaced to the right of that in placebo-pelleted rats, indicating antinociceptive "tolerance." RVM lidocaine or bilateral DLF lesion did not alter the intrathecal morphine DRC in placebo-pelleted rats but blocked the rightward displacement seen in morphine-pelleted animals. The subcutaneous morphine antinociceptive DRC in morphine-pelleted rats was displaced to the right of that in placebo-pelleted rats; this right shift was blocked by RVM lidocaine. The data show that (1) opioids elicit pain through tonic activation of bulbospinal facilitation from the RVM, (2) increased pain decreases spinal opioid antinociceptive potency, and (3) blockade of pain restores antinociceptive potency, revealing no change in antinociceptive signal transduction. These studies offer a mechanism for paradoxical opioid-induced pain and allow the development of approaches by which the loss of analgesic activity of opioids might be inhibited.


Asunto(s)
Analgésicos/administración & dosificación , Tolerancia a Medicamentos , Bulbo Raquídeo/fisiopatología , Narcóticos/efectos adversos , Dolor/fisiopatología , Animales , Relación Dosis-Respuesta a Droga , Implantes de Medicamentos , Calor , Hiperalgesia/inducido químicamente , Hiperalgesia/fisiopatología , Lidocaína/administración & dosificación , Masculino , Bulbo Raquídeo/efectos de los fármacos , Microinyecciones , Morfina/administración & dosificación , Morfina/efectos adversos , Narcóticos/administración & dosificación , Compresión Nerviosa , Dolor/inducido químicamente , Dimensión del Dolor/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Estimulación Física , Ratas , Tiempo de Reacción/efectos de los fármacos , Traumatismos de la Médula Espinal/fisiopatología
9.
J Neurosci ; 21(5): 1779-86, 2001 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11222667

RESUMEN

Whereas tissue injury increases spinal dynorphin expression, the functional relevance of this upregulation to persistent pain is unknown. Here, mice lacking the prodynorphin gene were studied for sensitivity to non-noxious and noxious stimuli, before and after induction of experimental neuropathic pain. Prodynorphin knock-out (KO) mice had normal responses to acute non-noxious stimuli and a mild increased sensitivity to some noxious stimuli. After spinal nerve ligation (SNL), both wild-type (WT) and KO mice demonstrated decreased thresholds to innocuous mechanical and to noxious thermal stimuli, indicating that dynorphin is not required for initiation of neuropathic pain. However, whereas neuropathic pain was sustained in WT mice, KO mice showed a return to baselines by post-SNL day 10. In WT mice, SNL upregulated lumbar dynorphin content on day 10, but not day 2, after injury. Intrathecal dynorphin antiserum reversed neuropathic pain in WT mice at post-SNL day 10 (when dynorphin was upregulated) but not on post-SNL day 2; intrathecal MK-801 reversed SNL-pain at both times. Opioid (mu, delta, and kappa) receptor density and G-protein activation were not different between WT and KO mice and were unchanged by SNL injury. The observations suggest (1) an early, dynorphin-independent phase of neuropathic pain and a later dynorphin-dependent stage, (2) that upregulated spinal dynorphin is pronociceptive and required for the maintenance of persistent neuropathic pain, and (3) that processes required for the initiation and the maintenance of the neuropathic pain state are distinct. Identification of mechanisms that maintain neuropathic pain appears important for strategies to treat neuropathic pain.


Asunto(s)
Dinorfinas/metabolismo , Neuralgia/metabolismo , Neuralgia/fisiopatología , Nervios Espinales/fisiopatología , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Maleato de Dizocilpina/administración & dosificación , Dinorfinas/antagonistas & inhibidores , Dinorfinas/farmacología , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Hiperestesia/metabolismo , Hiperestesia/fisiopatología , Sueros Inmunes/administración & dosificación , Inyecciones Espinales , Ligadura , Región Lumbosacra , Masculino , Ratones , Ratones Noqueados , Neuralgia/tratamiento farmacológico , Dimensión del Dolor/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Estimulación Física , Tiempo de Reacción/efectos de los fármacos , Receptores Opioides/análisis , Receptores Opioides/metabolismo , Médula Espinal/química , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Nervios Espinales/cirugía
10.
J Neurosci ; 23(23): 8370-9, 2003 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-12967999

RESUMEN

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.


Asunto(s)
Vías Aferentes/fisiopatología , Síndromes de Compresión Nerviosa/fisiopatología , Neuralgia/fisiopatología , Neurotransmisores/metabolismo , Nervios Espinales/fisiopatología , Vías Aferentes/metabolismo , Analgésicos Opioides/farmacología , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Capsaicina/farmacología , Modelos Animales de Enfermedad , Dinorfinas/metabolismo , Encefalinas/metabolismo , Hiperestesia/etiología , Hiperestesia/fisiopatología , Ligadura , Región Lumbosacra , Masculino , Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/fisiopatología , Microinyecciones , N-Glicosil Hidrolasas , Síndromes de Compresión Nerviosa/complicaciones , Neuralgia/etiología , Plasticidad Neuronal , Oligopéptidos/farmacología , Péptidos Opioides , Dimensión del Dolor/efectos de los fármacos , Precursores de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Proteínas Inactivadoras de Ribosomas Tipo 1 , Saporinas , Umbral Sensorial/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Nervios Espinales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA