Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Drugs Today (Barc) ; 56(1): 5-19, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32055802

RESUMEN

Migraine is a common, painful and highly disabling neurological condition that has plagued mankind for millennia, but its pathophysiology remained largely obscure until recently. The clinical success of triptans for treating migraine and the discovery that calcitonin gene-related peptide (CGRP) plays a prominent role in migraine led to increased research interest into this disease. An important improvement has been the development of monoclonal antibodies, including galcanezumab, that bind to CGRP or to its receptor, preventing its activation. Subsequent clinical trials have reported that galcanezumab is safe and well tolerated, and is effective in reducing the frequency of migraine attacks in patients with episodic or chronic migraine. At the same time, increased study of the pathophysiology of cluster headache, a relatively rare condition with excruciatingly painful headache attacks (i.e., "suicide headaches"), led to the discovery that, as in migraine, CGRP plays an important role in its pathology. Clinical trials suggest that galcanezumab is safe and effective for the prevention of episodic cluster headache, and it is under study for chronic cluster headache. Galcanezumab is approved for the prevention of migraine in the U.S., the European Union, Canada and Mexico, and was also approved for the treatment of episodic cluster headache in the U.S.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Cefalalgia Histamínica/tratamiento farmacológico , Trastornos Migrañosos/tratamiento farmacológico , Anticuerpos Monoclonales , Humanos
2.
J Clin Pharm Ther ; 43(4): 443-449, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29722031

RESUMEN

WHAT IS KNOWN AND OBJECTIVE: Although pain is one of the most common afflictions, it is often inadequately managed because the available analgesic options are relatively limited due to insufficient efficacy, unacceptable adverse effects or the potential for misuse or abuse. However, recent publications suggest that an alternative approach-indirect enhancement of endogenous pain-relieving pathways-might be desirable. We review this approach, in particular the dual enkephalinase inhibitors (DENKIs). METHODS: Published literature and Internet sources were searched for information related to the basic science and clinical data on inhibition of metabolic pathways of endogenous analgesic agents. The identified sources were reviewed, assessed and synthesized. Emphasis was placed on the benefits of the approach, as well as on the individual agents. RESULTS: Inhibition of the enzymes that degrade the endogenous opioid ligands Met- and Leu-enkephalin results in an increased synaptic concentration of the enkephalins and an analgesic effect in a variety of animal models of pain and in preliminary trials in humans. The design of compounds that inhibit both of the two major enkephalin-degrading enzymes (neprilysin and aminopeptidase N) has been found to be better than those that inhibit only one of the enzymes. These dual-acting enkephalinase inhibitors yield analgesia with less adverse effects than current opioid drugs. WHAT IS NEW AND CONCLUSION: Unlike currently available analgesics, inhibitors of the metabolic degradation of endogenous analgesic substances attempt to elicit a more "natural" and targeted analgesic effect. This indirect approach offers an opportunity for novel additions to the otherwise relatively limited choice of analgesic classes.


Asunto(s)
Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Neprilisina/antagonistas & inhibidores , Dolor/tratamiento farmacológico , Animales , Antígenos CD13/antagonistas & inhibidores , Humanos
3.
J Clin Pharm Ther ; 43(3): 437-441, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29520812

RESUMEN

WHAT IS KNOWN AND OBJECTIVE: Advances in pain research have led to an understanding that many pains are driven by more than one underlying (patho)physiologic cause (ie, they are "multimechanistic") and that better pain relief is obtained with fewer adverse effects when an analgesic is correspondingly multimechanistic. At least two of the more-modern analgesics combine opioid and non-opioid mechanisms, and have become known as "atypical opioids." Less well known is that just as Nature evolved opioids, it also evolved atypical opioids, presaging modern drug discovery efforts. COMMENT: Traditional (typical) opioids are extracts or analogs of substances derived from the poppy plant. They produce their analgesic and adverse effects primarily through a single, opioid mechanism (albeit with individual differences). Two most recent analgesics were developed to have both an opioid mechanism and, a second, non-opioid mechanism of action (inhibition of monoamine neurotransmitter reuptake). Little known is that Nature had already evolved a plant source of compounds with the same properties. WHAT IS NEW AND CONCLUSION: As debate about the use and abuse potential of kratom swirls, conflicting, often contradicting, opinions are expressed. A review of the basic pharmacology of kratom reveals the explanation for the bifurcation in viewpoints: kratom has both opioid and non-opioid properties. Fascinatingly, just as the poppy plant (Papaver) evolved the typical opioids, Mitragyna evolved the mitragynines-Nature's "atypical opioids."


Asunto(s)
Analgésicos Opioides/farmacología , Analgésicos/farmacología , Dolor/tratamiento farmacológico , Alcaloides de Triptamina Secologanina/farmacología , Analgésicos/aislamiento & purificación , Analgésicos Opioides/aislamiento & purificación , Animales , Humanos , Mitragyna/química , Extractos Vegetales/farmacología , Alcaloides de Triptamina Secologanina/aislamiento & purificación
4.
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
5.
Neuroscience ; 123(1): 43-52, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14667440

RESUMEN

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.


Asunto(s)
Dinorfinas/biosíntesis , Traumatismos de los Nervios Periféricos , Enfermedades del Sistema Nervioso Periférico/metabolismo , Médula Espinal/metabolismo , Regulación hacia Arriba/fisiología , Animales , Dinorfinas/farmacología , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Nervios Periféricos/efectos de los fármacos , Nervios Periféricos/metabolismo , Proteína Quinasa C/biosíntesis , Proteína Quinasa C/deficiencia , Proteína Quinasa C/genética , Especificidad de la Especie , Médula Espinal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
6.
Neuroscience ; 121(3): 815-24, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14568039

RESUMEN

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.


Asunto(s)
Ganglios Espinales/lesiones , Factores de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , Dolor/metabolismo , Factores de Transcripción Activadores , Animales , Proteínas Sanguíneas/metabolismo , Modelos Animales de Enfermedad , Lateralidad Funcional , Galanina/metabolismo , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Factor Neurotrófico Derivado de la Línea Celular Glial , Humanos , Inmunohistoquímica , Lectinas/metabolismo , Masculino , Neuropéptido Y/metabolismo , Dolor/patología , Dolor/prevención & control , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X3 , Factores de Tiempo , Factores de Transcripción/metabolismo
7.
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
8.
Pain ; 100(3): 243-248, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12467995

RESUMEN

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.


Asunto(s)
Dronabinol/administración & dosificación , Dinorfinas/deficiencia , Morfolinas/administración & dosificación , Naltrexona/análogos & derivados , Naftalenos/administración & dosificación , Dimensión del Dolor/efectos de los fármacos , Dolor/tratamiento farmacológico , Médula Espinal/efectos de los fármacos , Analgésicos/administración & dosificación , Analgésicos no Narcóticos/administración & dosificación , Animales , Benzoxazinas , Relación Dosis-Respuesta a Droga , Inyecciones Espinales , Masculino , Ratones , Ratones Noqueados , Valores de Referencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Método Simple Ciego , Especificidad de la Especie
9.
Pain ; 98(1-2): 79-88, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12098619

RESUMEN

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.


Asunto(s)
Analgésicos/farmacología , Cannabinoides/farmacología , Dinorfinas/administración & dosificación , Morfolinas/farmacología , Naftalenos/farmacología , Nociceptores/efectos de los fármacos , Nociceptores/fisiología , Dolor/inducido químicamente , Animales , Anticuerpos/inmunología , Benzoxazinas , Reacciones Cruzadas , Sinergismo Farmacológico , Tolerancia a Medicamentos , Dinorfinas/metabolismo , Dinorfinas/farmacología , Calor , Técnicas para Inmunoenzimas , Inyecciones Espinales , Isomerismo , Masculino , Ratones , Ratones Endogámicos ICR , Umbral del Dolor/efectos de los fármacos , Estimulación Física , Médula Espinal/metabolismo
10.
J Neurosci ; 21(14): 5281-8, 2001 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-11438603

RESUMEN

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.


Asunto(s)
Tronco Encefálico/efectos de los fármacos , Inmunotoxinas , N-Glicosil Hidrolasas , Neuralgia/tratamiento farmacológico , Neuronas/efectos de los fármacos , Receptores Opioides mu/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/administración & dosificación , Animales , Conducta Animal/efectos de los fármacos , Tronco Encefálico/citología , Tronco Encefálico/metabolismo , Modelos Animales de Enfermedad , Ligadura , Masculino , Bulbo Raquídeo/citología , Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/metabolismo , Microinyecciones , Naltrexona/administración & dosificación , Naltrexona/análogos & derivados , Neuralgia/fisiopatología , Neuronas/metabolismo , Oligopéptidos/administración & dosificación , Péptidos Opioides , Dimensión del Dolor/efectos de los fármacos , Estimulación Física , Proteínas de Plantas/administración & dosificación , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Receptores Opioides mu/biosíntesis , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Proteínas Inactivadoras de Ribosomas Tipo 1 , Saporinas , Nervios Espinales/lesiones , Nervios Espinales/fisiopatología
12.
Pain ; 90(1-2): 105-11, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11166976

RESUMEN

Peripheral nerve injury produces signs of neuropathic pain including tactile allodynia and thermal hyperalgesia, sensory modalities which may be associated with different neuronal pathways. Studies of spinally-transected, nerve-injured rats have led to suggestions that thermal hyperalgesia may be mediated predominately through local spinal circuitry whereas ascending input to supraspinal sites is critical to the manifestation of tactile allodynia. Here, the nature of ascending spinal input mediating tactile allodynia was explored using selective spinal lesions. Male Sprague-Dawley rats received L(5)/L(6) spinal nerve ligation (SNL) and ipsilateral or contralateral (relative to the SNL side) lesions including spinal hemisections and bilateral and unilateral dorsal column lesions. The rats were maintained in a sling and monitored for tactile allodynia by measuring withdrawal thresholds to probing with von Frey filaments 24 h after the hemisection. Rats receiving dorsal column lesions demonstrated no motor deficits while rats receiving spinal hemisection showed paralysis of the paw which nevertheless responded to strong noxious stimulation. Spinal hemisection ipsilateral, but not contralateral, to SNL completely abolished tactile allodynia while maintaining spinal nocifensive reflexes to noxious pinch. Bilateral and ipsilateral dorsal column lesions blocked tactile allodynia while contralateral dorsal column lesions did not. Administration of lidocaine into the nucleus gracilis ipsilateral to SNL also blocked tactile allodynia, but did not alter thermal hyperalgesia in SNL rats or increase thermal nociceptive responses in sham-operated rats. Lidocaine microinjected into the contralateral nucleus gracilis produced no changes in responses to tactile or thermal stimuli in either group. These results indicate that tactile allodynia after peripheral nerve injury is dependent upon inputs to supraspinal sites. Furthermore, it is apparent that afferent signals interpreted as tactile allodynia course through the ipsilateral dorsal columns and are relayed through the nucleus gracilis. This neuronal pathway is consistent with the interpretation that tactile allodynia pursuant to peripheral nerve injury is transmitted to the central nervous system by means of large diameter, myelinated fibers.


Asunto(s)
Hiperalgesia/fisiopatología , Umbral del Dolor/fisiología , Células del Asta Posterior/fisiología , Nervios Espinales/fisiología , Anestésicos Locales/farmacología , Animales , Calor , Lidocaína/farmacología , Masculino , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/fisiología , Umbral del Dolor/efectos de los fármacos , Células del Asta Posterior/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Nervios Espinales/efectos de los fármacos , Nervios Espinales/lesiones , Columna Vertebral , Tacto
13.
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
14.
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
15.
Mol Interv ; 1(3): 160-7, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-14993349

RESUMEN

One of the curious but common consequences of opioid administration in the clinical setting is the induction, at sites uninvolved in the original presentation of discomfort, of pain itself. The induction of pain is also a reliable, measurable phenomenon in animals receiving continuous delivery of opioid. Such pain induction is associated with the expression of spinal dynorphin, a finding that is especially intriguing in light of dynorphin's ability to recapitulate many of the characteristics of chronic, neuropathic pain when administered intrathecally (i.e., into the spine). The effective treatment of chronic pain syndromes-and of tolerance to antinociceptive therapies-may thus rest on an understanding of the biological roles of dynorphin in neurotransmission.


Asunto(s)
Tolerancia a Medicamentos , Dinorfinas/metabolismo , Dolor/fisiopatología , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Animales , Dinorfinas/antagonistas & inhibidores , Dinorfinas/farmacología , Humanos , Hiperalgesia/inducido químicamente , Inyecciones Espinales , Modelos Biológicos , Neurotransmisores/metabolismo , Dolor/etiología , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
16.
Neurosci Lett ; 292(2): 115-8, 2000 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-10998562

RESUMEN

Recent studies suggest a role of Group 1 metabotropic glutamate receptors in mediating the development of spinal hypersensitivity in some pain states. Here, the possible role of mGluR(5) receptors in experimental neuropathic pain elicited by ligation of spinal nerves (L(5)/L(6) spinal nerve ligation, SNL) was explored with SIB-1757, a selective mGluR(5) antagonist. SNL-induced tactile allodynia was detected by decreased paw withdrawal thresholds to probing with von Frey filaments and thermal hyperalgesia by decreased paw withdrawal latencies to radiant heat applied to the plantar aspect of the hindpaw. SIB-1757 was given by either intrathecal (i.th.), subcutaneous (s.c.) or intraplantar (i.pl.) injection. In SNL rats, i.th. SIB-1757 produced a partial reversal of tactile allodynia with a shallow dose-response curve ranging over three-orders of magnitude; SIB-1757 was inactive against allodynia when given systemically. SIB-1757 produced full reversal of thermal hyperalgesia in SNL rats following administration either spinally or locally to the injured paw; administration to the contralateral paw had no effect. SIB-1757 did not produce antinociception in either the SNL or sham-operated rats by any route. These data suggest a significant modulation of thermal hyperalgesia by mGluR(5) antagonists, consistent with reports that this receptor may be associated with afferent C-fibers. The less impressive effect seen on tactile allodynia, likely to be mediated by large fiber input, suggests that the observed modulation may be related to blockade of mGluR(5)-mediated spinal sensitization. These results do not support the involvement of these receptors in modulation of acute nociception but suggest the possibility of a role for Group I mGluRs in the mediation of aspects of neuropathic pain which may be associated with C-fiber inputs.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/farmacología , Hiperalgesia/tratamiento farmacológico , Fenazopiridina/farmacología , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Animales , Relación Dosis-Respuesta a Droga , Ligadura , Masculino , Fibras Nerviosas/fisiología , Nociceptores/efectos de los fármacos , Nociceptores/fisiología , Umbral del Dolor/efectos de los fármacos , Estimulación Física , Ratas , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5 , Nervios Espinales/cirugía
17.
J Neurosci ; 20(18): 7074-9, 2000 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-10995854

RESUMEN

The nonopioid actions of spinal dynorphin may promote aspects of abnormal pain after nerve injury. Mechanistic similarities have been suggested between opioid tolerance and neuropathic pain. Here, the hypothesis that spinal dynorphin might mediate effects of sustained spinal opioids was explored. Possible abnormal pain and spinal antinociceptive tolerance were evaluated after intrathecal administration of [D-Ala(2), N-Me-Phe(4), Gly-ol(5)]enkephalin (DAMGO), an opioid mu agonist. Rats infused with DAMGO, but not saline, demonstrated tactile allodynia and thermal hyperalgesia of the hindpaws (during the DAMGO infusion) and a decrease in antinociceptive potency and efficacy of spinal opioids (tolerance), signs also characteristic of nerve injury. Spinal DAMGO elicited an increase in lumbar dynorphin content and a decrease in the mu receptor immunoreactivity in the spinal dorsal horn, signs also seen in the postnerve-injury state. Intrathecal administration of dynorphin A(1-17) antiserum blocked tactile allodynia and reversed thermal hyperalgesia to above baseline levels (i.e., antinociception). Spinal dynorphin antiserum, but not control serum, also reestablished the antinociceptive potency and efficacy of spinal morphine. Neither dynorphin antiserum nor control serum administration altered baseline non-noxious or noxious thresholds or affected the intrathecal morphine antinociceptive response in saline-infused rats. These data suggest that spinal dynorphin promotes abnormal pain and acts to reduce the antinociceptive efficacy of spinal opioids (i.e., tolerance). The data also identify a possible mechanism for previously unexplained clinical observations and offer a novel approach for the development of strategies that could improve the long-term use of opioids for pain.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Tolerancia a Medicamentos , Dinorfinas/metabolismo , Hiperalgesia/metabolismo , Médula Espinal/metabolismo , Analgésicos/administración & dosificación , Animales , Dinorfinas/antagonistas & inhibidores , Dinorfinas/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Miembro Posterior , Calor , Hiperalgesia/inducido químicamente , Sueros Inmunes/administración & dosificación , Inmunohistoquímica , Inyecciones Espinales , Masculino , Morfina/administración & dosificación , Dimensión del Dolor/efectos de los fármacos , Pruebas de Precipitina , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Umbral Sensorial/efectos de los fármacos , Médula Espinal/citología , Médula Espinal/efectos de los fármacos , Tacto
18.
Neurosci Lett ; 290(2): 129-32, 2000 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-10936694

RESUMEN

Evidence exists to indicate that tactile allodynia arising from peripheral nerve injury is integrated predominately at supraspinal, rather than spinal, sites. In the present experiments, the possibility that disruption of descending pathways through the dorsolateral funiculus (DLF) might alter expression of nerve-injury induced tactile allodynia was explored. Male, Sprague-Dawley rats received L(5)/L(6) spinal nerve ligation (SNL). Lesions to the DLF were made ipsilateral or contralateral to SNL. Tactile allodynia was determined by measuring withdrawal thresholds to probing with von Frey filaments. Rats with DLF lesions presented no apparent motor deficits and did not alter sensory threshold in sham-SNL operated rats. DLF lesions made ipsilateral to SNL completely blocked tactile allodynia in SNL rats. Contralateral DLF lesions and sham surgery did not have any effect on SNL-induced allodynia. These results indicate that tactile allodynia after peripheral nerve injury is dependent upon tonic activation of net descending facilitation from supraspinal sites and support the hypothesis of tonic activation of descending facilitation as a basis for chronic pain.


Asunto(s)
Vías Eferentes/fisiopatología , Bulbo Raquídeo/fisiopatología , Compresión Nerviosa/efectos adversos , Dolor/fisiopatología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Médula Espinal/fisiopatología , Tacto/fisiología , Animales , Desnervación , Vías Eferentes/citología , Masculino , Bulbo Raquídeo/citología , Dolor/patología , Umbral del Dolor/fisiología , Enfermedades del Sistema Nervioso Periférico/patología , Estimulación Física , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología
19.
Pain ; 87(2): 193-199, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10924812

RESUMEN

The spinal activity of racemic ketoprofen and its enantiomers in models of neuropathic and tonic pain was explored in rats. Tactile allodynia and thermal hyperalgesia were induced by tight ligation of the L(5)/L(6) spinal nerves. Tonic pain was modeled by the formalin-induced flinch response. The spinal injection of (S)-ketoprofen alone or of morphine alone did not produce antiallodynic activity. A 1:1 combination of these drugs produced a robust dose-dependent antiallodynic action, consistent with previous observations where (S)-ketorolac combined with morphine also produced antiallodynia. (R)-ketoprofen given alone spinally produced a dose-dependent antiallodynia, but its activity was not augmented by spinal morphine. Conversely, (S)-ketoprofen, but not (R)-ketoprofen, blocked the second phase of the formalin-induced flinch response; neither enantiomer significantly blocked phase one of the formalin response. Again, (S)-, but not (R)-ketoprofen, interacted synergistically with spinal morphine in suppressing the phase II formalin response. These results are consistent with a spinal COX inhibitory action of (S)-ketoprofen. These results also point to a novel, as yet undefined, mechanism of action of (R)-ketoprofen against signs of neuropathic pain that does not appear to involve COX inhibition. The ability to modulate tactile allodynia is of special interest as this represents an aspect of clinical neuropathic pain that is very difficult to treat adequately.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Cetoprofeno/farmacología , Dimensión del Dolor/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Analgésicos Opioides/farmacología , Animales , Antiinflamatorios no Esteroideos/química , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Cetoprofeno/química , Masculino , Morfina/farmacología , Umbral del Dolor/fisiología , Ratas , Ratas Sprague-Dawley , Estereoisomerismo
20.
Pain ; 87(3): 265-273, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10963906

RESUMEN

Complete or partial spinal section at T(8) has been shown to block tactile allodynia but not thermal hyperalgesia following L(5)/L(6) spinal nerve ligation (SNL), suggesting the supraspinal integration of allodynia in neuropathic pain. In the present study, the possibility of mediation of nerve injury-associated pain through tonic activity of descending nociceptive facilitation arising from the rostroventromedial medulla (RVM) was investigated. Specifically, the actions of brainstem cholecystokinin and the possible importance of sustained afferent input from injured nerve fibers were determined using pharmacological and physiological approaches in rats with SNL. Lidocaine given bilaterally into the RVM blocked tactile allodynia and thermal hyperalgesia in SNL rats and was inactive in sham-operated rats. Bilateral injection of L365,260 (CCK(B) receptor antagonist) into the RVM also reversed both tactile allodynia and thermal hyperalgesia. Microinjection of CCK-8 (s) into the RVM of naive rats produced a robust tactile allodynic effect and a more modest hyperalgesia. CCK immunoreactivity was not significantly different between SNL and sham-operated rats. The anti-nociceptive effect of morphine given into the ventrolateral periaqueductal gray region (PAG) was substantially reduced by SNL. The injection of L365,260 into the RVM or of bupivacaine at the site of nerve injury restored the potency and efficacy of PAG morphine in SNL rats. These results suggest that changes in supraspinal processing are likely to contribute to the observed poor efficacy of opioids in clinical states of neuropathic pain. These data also indicate that the activation of descending nociceptive facilitatory pathways is important in the maintenance of neuropathic pain, appears to be dependent on CCK release, and may be driven from sustained afferent input from injured nerves to brainstem sites. Collectively, these data support the hypothesis that abnormal tonic activity of descending facilitation mechanisms may underlie chronic pain from peripheral nerve injury.


Asunto(s)
Colecistoquinina/fisiología , Hiperalgesia/fisiopatología , Bulbo Raquídeo/fisiología , Dimensión del Dolor , Dolor/fisiopatología , Anestésicos Locales/farmacología , Anestésicos Locales/uso terapéutico , Animales , Benzodiazepinonas/farmacología , Benzodiazepinonas/uso terapéutico , Colecistoquinina/efectos adversos , Calor/efectos adversos , Hiperalgesia/inducido químicamente , Lidocaína/farmacología , Lidocaína/uso terapéutico , Ligadura , Masculino , Bulbo Raquídeo/efectos de los fármacos , Dolor/tratamiento farmacológico , Dimensión del Dolor/efectos de los fármacos , Sustancia Gris Periacueductal/efectos de los fármacos , Sustancia Gris Periacueductal/fisiopatología , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores de Colecistoquinina/antagonistas & inhibidores , Médula Espinal/efectos de los fármacos , Médula Espinal/fisiopatología , Nervios Espinales/lesiones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA