Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Eur J Neurosci ; 29(4): 727-37, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19200067

RESUMEN

The clinically important opioid fentanyl, administered acutely, enhances mechanical hypersensitivity in a model of surgical pain induced by plantar incision. Activity of neurokinin-1 (NK-1) receptor-expressing ascending spinal neurons, descending pathways originating in the rostral ventromedial medulla (RVM), and spinal dynorphin are necessary for the development and maintenance of hyperalgesia during sustained morphine exposure, suggesting that these mechanisms may also be important in opioid enhancement of surgical pain. Therefore, we examined the roles of these mechanisms in sensory hypersensitivity produced by acute fentanyl administration in rats not undergoing surgical incision and in rats undergoing plantar incision. In non-operated rats, fentanyl induced analgesia followed by immediate and long-lasting sensory hypersensitivity, as previously described. Fentanyl also enhanced pain sensitivity induced by plantar incision. Ablation of NK-1-expressing spinal neurons by pre-treatment with substance P-Saporin reduced sensory hypersensitivity in fentanyl-treated rats and, to a lesser extent, in fentanyl-treated rats with a surgical incision. Microinjection of lidocaine into the RVM completely reversed fentanyl-induced sensory hypersensitivity and fentanyl enhancement of incision-induced sensory hypersensitivity. RVM lidocaine injection resulted in a slight reduction of incision-induced sensory hypersensitivity in the absence of fentanyl pre-treatment. Spinal dynorphin content increased by 30 +/- 7% and 66 +/- 17% in fentanyl- and fentanyl/incision-treated rats. Spinal administration of antiserum to dynorphin attenuated sensory hypersensitivity in fentanyl-treated rats. These data support a partial role of NK-1 receptor-containing ascending pathways and a crucial role of descending facilitatory pathways in fentanyl-induced hyperalgesia and in the enhanced hyperalgesia produced by fentanyl treatment following surgical incision.


Asunto(s)
Bulbo Raquídeo/fisiopatología , Neuronas/metabolismo , Dolor Postoperatorio/fisiopatología , Receptores de Neuroquinina-1/metabolismo , Médula Espinal/fisiopatología , Analgésicos Opioides/farmacología , Anestésicos Locales/farmacología , Animales , Modelos Animales de Enfermedad , Dinorfinas/metabolismo , Fentanilo/farmacología , Inmunohistoquímica , Lidocaína/farmacología , Masculino , Vías Nerviosas/fisiopatología , Umbral del Dolor/efectos de los fármacos , Dolor Postoperatorio/inducido químicamente , Dolor Postoperatorio/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley
3.
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
4.
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
5.
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
6.
J Mol Biol ; 180(4): 881-909, 1984 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-6098691

RESUMEN

CRP-cAMP was shown to activate transcription initiation at the Escherichia coli lac promoter in vitro as a result of two separate effects. An indirect component of the activation resulted from an enhancement of the fraction of promoters productively bound by RNA polymerase. This effect was due largely to CRP-cAMP repression of RNA polymerase binding to an overlapping site (lac P2) within the promoter region. In addition, a direct enhancement of RNA polymerase binding at the principal lac promoter (lac P1) was found. The combination of indirect and direct activation by CRP-cAMP was suggested to be responsible for the large activation observed in vivo. Promoter strength parameters were also determined for the L8, UV5 and Ps promoters. The effect of CRP-cAMP on these mutant promoters was shown to be consistent with the activation mechanism deduced for the lac wild-type promoter. DNA supercoiling enhanced the promoter strength of the lac wild-type and UV5 promoters. The combination of supercoiling and CRP-cAMP was necessary for optimal promoter strength for the lac wild-type promoter.


Asunto(s)
AMP Cíclico , Escherichia coli/genética , Operón Lac , Receptores de AMP Cíclico , Transcripción Genética , ADN Superhelicoidal , ARN Polimerasas Dirigidas por ADN/metabolismo , Cinética , Modelos Genéticos , Moldes Genéticos
7.
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
8.
Pain ; 79(2-3): 127-33, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10068158

RESUMEN

Tactile allodynia and thermal hyperalgesia, two robust signs of neuropathic pain associated with experimental nerve injury, have been hypothesized to be mechanistically distinguished based on (a) fiber types which may be involved in the afferent input, (b) participation of spinal and supraspinal circuitry in these responses, and (c) sensitivity of these endpoints to pharmacological agents. Here, the possibility that nerve-injury induced tactile allodynia and thermal hyperalgesia may be mediated via different afferent fiber input was tested by evaluating these responses in sham-operated or nerve-injured (L5/L6) rats before or after a single systemic injection of resiniferatoxin (RTX), an ultrapotent analogue of the C-fiber specific neurotoxin, capsaicin. Tactile allodynia, and three measures of thermal nociception, tail-flick, paw-flick and hot-plate responses, were determined before and at various intervals for at least 40 days after RTX injection. Nerve-injured, but not sham-operated, rats showed a long-lasting tactile allodynia and thermal hyperalgesia (paw-flick) within 2-3 days after surgery; responses to other noxious thermal stimuli (i.e., tail-flick and hot-plate tests) did not distinguish the two groups at the stimulus intensities employed. RTX treatment resulted in a significant and long-lasting (i.e. essentially irreversible) decrease in sensitivity to thermal noxious stimuli in both sham-operated and nerve-injured rats; thermal hyperalgesia was abolished and antinociception produced by RTX. In contrast, RTX treatment did not affect the tactile allodynia seen in the same nerve-injured rats. These data support the concept that thermal hyperalgesia seen after nerve ligation, as well as noxious thermal stimuli, are likely to be mediated by capsaicin-sensitive C-fiber afferents. In contrast, nerve-injury related tactile allodynia is insensitive to RTX treatment which clearly desensitizes C-fibers and, therefore such responses are not likely to be mediated through C-fiber afferents. The hypothesis that tactile allodynia may be due to inputs from large (i.e. A beta) afferents offers a mechanistic basis for the observed insensitivity of this endpoint to intrathecal morphine in this nerve-injury model. Further, these data suggest that clinical treatment of neuropathic pains with C-fiber specific agents such as capsaicin are unlikely to offer significant therapeutic benefit against mechanical allodynia.


Asunto(s)
Capsaicina/farmacología , Neuronas Aferentes , Dolor/fisiopatología , Sistema Nervioso Periférico/lesiones , Sistema Nervioso Periférico/fisiopatología , Piel/fisiopatología , Animales , Diterpenos/farmacología , Hiperalgesia/fisiopatología , Ligadura , Masculino , Neuronas Aferentes/efectos de los fármacos , Dimensión del Dolor/efectos de los fármacos , Estimulación Física , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Piel/inervación , Factores de Tiempo
9.
Pain ; 71(1): 57-64, 1997 May.
Artículo en Inglés | MEDLINE | ID: mdl-9200174

RESUMEN

Nerve ligation injury in rats results in reduced nociceptive and non-nociceptive thresholds, similar to some aspects of clinical conditions of neuropathic pain. Since underlying mechanisms of hyperalgesia and allodynia may differ, the present study investigated the pharmacology of morphine and MK-801 in rats subjected to a tight ligation of the L5 and L6 nerve roots or to a sham-operation procedure. Response to acute nociception was measured by (a) withdrawal of a hindpaw from a radiant heat source, (b) withdrawal of the tail from a radiant heat source or (c) the latency to a rapid flick of the tail following immersion in water at different noxious temperatures. Mechanical thresholds were determined by measuring response threshold to probing the hindpaw with von Frey filaments. Nerve ligation produced a significant, stable and long-lasting decrease in threshold to mechanical stimulation (i.e., tactile allodynia) when compared to sham-operated controls. Standardization of the diameter of the filaments (to that of the largest filament) did not alter the response threshold in nerve-injured animals. Nerve ligation produced decreased response latency of the ipsilateral paw (i.e., hyperalgesia) when compared to that of sham-operated rats. Tail-flick latencies to thermal stimuli induced by water at constant temperatures (48 degrees, 52 degrees or 55 degrees C) or by radiant heat were not significantly different between nerve-injured and sham-operated groups. At doses which were not behaviorally toxic, MK-801 had no effect on tactile allodynia. At these doses, MK-801 blocked decreased paw withdrawal latency to radiant heat in nerve-injured rats, but did not significantly elevate the response threshold of sham-operated rats. Systemic (i.p.) or intracerebroventricular (i.c.v.) doses of morphine previously shown to be antiallodynic in nerve-ligated rats did not affect the response to probing with von Frey filaments in sham-operated controls. Intrathecal (i.t.) morphine did not change paw withdrawal thresholds elicited by von Frey filaments of either nerve-ligated rats (as previously reported) or of sham-operated rats at doses maximally effective against thermal stimuli applied to the tail or foot. Spinal morphine produced dose-dependent antinociception in both nerve-injured and sham-operated groups in the foot-flick test but was less potent in the nerve-injured group. Presuppression of hyperalgesia of the foot with i.t. MK-801 in nerve-injured animals did not alter the potency of i.t. morphine. I.t. morphine was also active in the tail-flick tests with decreased potency in nerve-injured animals and, at some stimulus intensities, with a decreased efficacy as well. These data emphasize the distinction between the inactivity of morphine to suppress mechanical withdrawal thresholds (as elicited by von Frey filaments) and the activity of this compound to block the response to an acute thermal nociceptive stimulus in sham-operated or nerve-injured rats. It appears that nerve ligation injury produces a thermal allodynia/hyperalgesia which is likely dependent upon opioid-sensitive small-diameter primary afferent fibers and a mechanical allodynia which may be largely independent of small-fiber input.


Asunto(s)
Analgésicos Opioides/farmacología , Maleato de Dizocilpina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Hiperalgesia/tratamiento farmacológico , Morfina/farmacología , Traumatismos de los Nervios Periféricos , Analgésicos Opioides/administración & dosificación , Animales , Maleato de Dizocilpina/administración & dosificación , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Calor , Hiperalgesia/etiología , Hiperalgesia/fisiopatología , Masculino , Morfina/administración & dosificación , Dimensión del Dolor/efectos de los fármacos , Estimulación Física , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Factores de Tiempo
10.
Pain ; 68(2-3): 275-81, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9121815

RESUMEN

Neuropathic pain states are accompanied by increased sensitivity to both noxious and non-noxious sensory stimuli, characterized as hyperalgesia and allodynia, respectively. In animal models of neuropathic pain, the presence of hyperalgesia and allodynia are accompanied by neuroplastic changes including increased spinal levels of substance P, cholecystokinin (CCK), and dynorphin. N-Methyl-D-aspartate (NMDA) receptors appear to be involved in maintaining the central sensitivity which contributes to neuropathic pain. In addition to its opioid activities, dynorphin has been suggested to act at the NMDA receptor complex. In an attempt to mimic the increased levels of spinal dynorphin seen in animal models of neuropathic pain, rats received a single intrathecal (i.t.) injection of dynorphin A(1-17), dynorphin A(1-13), dynorphin A(2-17) or dynorphin A(2-13) through indwelling catheters. Tactile allodynia was determined by measuring response threshold to probing with von Frey filaments. Dynorphin A(1-17) administration evoked significant and long-lasting tactile allodynia (i.e. > 60 days). Likewise, the i.t. administration of dynorphin A(1-13) or dynorphin A(2-17) or dynorphin A(2-13) also produced long-lasting tactile allodynia. Intrathecal pretreatment, but not post-treatment, with MK-801 prevented dynorphin A(1-17)-induced development of allodynia; i.t. administration of MK-801 alone had no effect on responses to tactile stimuli. In contrast, i.t. pretreatment with naloxone did not affect the development of tactile allodynia induced by dynorphin A(1-17) or alter sensory threshold when given alone. These results demonstrate that a single dose of dynorphin A, or its des-Tyr fragments, produces long-lasting allodynia which may be irreversible in the rat. Further, this effect appears to be mediated through activation of NMDA, rather than opioid, receptors. While the precise mechanisms underlying the development and maintenance of the allodynia is unclear, it seems possible that dynorphin may produce changes in the spinal cord, which may contribute to the development of signs reminiscent of a "neuropathic' state. Given that levels of dynorphin are elevated following nerve injury, it seems reasonable to speculate that dynorphin may have a pathologically relevant role in neuropathic pain states.


Asunto(s)
Dinorfinas/antagonistas & inhibidores , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Fragmentos de Péptidos/antagonistas & inhibidores , Umbral Sensorial/efectos de los fármacos , Animales , Enfermedad Crónica , Maleato de Dizocilpina/uso terapéutico , Inyecciones Espinales , Masculino , Naloxona/uso terapéutico , Dolor/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley
11.
Pain ; 86(1-2): 185-94, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10779675

RESUMEN

Neuropathic pain is often associated with the appearance of pain in regions not related to the injured nerve. One mechanism that may underlie neuropathic pain is abnormal, spontaneous afferent drive which may contribute to NMDA-mediated central sensitization by the actions of glutamate and by the non-opioid actions of spinal dynorphin. In the present study, injuries to lumbar or sacral spinal nerves elicited elevation in spinal dynorphin content which correlated temporally and spatially with signs of neuropathic pain. The increase in spinal dynorphin content was coincident with the onset of tactile allodynia and thermal hyperalgesia. Injury to the lumbar (L(5)/L(6)) spinal nerves produced elevated spinal dynorphin content in the ipsilateral dorsal spinal quadrant at the L(5) and L(6) spinal segments and in the segments immediately adjacent. Lumbar nerve injury elicited ipsilateral tactile allodynia and thermal hyperalgesia of the hindpaw. In contrast, S(2) spinal nerve ligation elicited elevated dynorphin content in sacral spinal segments and bilaterally in the caudal lumbar spinal cord. The behavioral consequences of S(2) spinal nerve ligation were also bilateral, with tactile allodynia and thermal hyperalgesia seen in both hindpaws. Application of lidocaine to the site of S(2) ligation blocked thermal hyperalgesia and tactile allodynia of the hindpaws suggesting that afferent drive was critical to maintenance of the pain state. Spinal injection of antiserum to dynorphin A((1-17)) and of MK-801 both blocked thermal hyperalgesia, but not tactile allodynia, of the hindpaw after S(2) ligation. These data suggest that the elevated spinal dynorphin content consequent to peripheral nerve injury may drive sensitization of the spinal cord, in part through dynorphin acting directly or indirectly on the NMDA receptor complex. Furthermore, extrasegmental increases in spinal dynorphin content may partly underlie the development of extraterritorial neuropathic pain.


Asunto(s)
Dinorfinas/metabolismo , Dolor/metabolismo , Médula Espinal/metabolismo , Nervios Espinales/lesiones , Anestésicos Locales/administración & dosificación , Anestésicos Locales/farmacología , Animales , Anticuerpos Bloqueadores/farmacología , Maleato de Dizocilpina/farmacología , Dinorfinas/antagonistas & inhibidores , Dinorfinas/inmunología , Antagonistas de Aminoácidos Excitadores/farmacología , Calor , Hiperalgesia/metabolismo , Inmunoensayo , Lidocaína/administración & dosificación , Lidocaína/farmacología , Ligadura , Masculino , Dolor/etiología , Dimensión del Dolor/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Médula Espinal/efectos de los fármacos , Nervios Espinales/metabolismo
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.
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
14.
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
15.
Drugs ; 61(15): 2155-62, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11772127

RESUMEN

The inhaled anaesthetic sevoflurane is metabolised into two products that have the potential to produce renal injury. Fluoride ions are produced by oxidative defluorination of sevoflurane by the cytochrome P450 system in the liver. Until recently, inorganic fluoride has been thought to be the aetiological agent responsible for fluorinated anaesthetic nephrotoxicity, with a toxic concentration threshold of 50 micromol/L in serum. However, studies of sevoflurane administration in animals and humans have not shown evidence of fluoride-induced nephrotoxicity, despite serum fluoride concentrations in this range. Compound A (fluoromethyl-2,2-difluoro-1-[trifluoromethyl] vinyl ether) is a breakdown product of sevoflurane produced by its interaction with carbon dioxide absorbents in the anaesthesia machine. The patient then inhales compound A. Compound A produces evidence of transient renal injury in rats. The mechanism of compound A renal toxicity is controversial, with the debate focused on the role of the renal cysteine conjugate beta-lyase pathway in the biotransformation of compound A. The significance of this debate centres on the fact that the beta-lyase pathway is 10- to 30-fold less active in humans than in rats. Therefore, if biotransformation by this pathway is responsible for the production of nephrotoxic metabolites of compound A, humans may be less susceptible to compound A renal toxicity than are rats. In three studies in human volunteers and one in surgical patients, prolonged (8-hour) sevoflurane exposures and low fresh gas flow rates resulted in significant exposures to compound A. Transient abnormalities were found in biochemical markers of renal injury measured in urine. These studies suggested that sevoflurane can result in renal toxicity, mediated by compound A, under specific circumstances. However, other studies using prolonged sevoflurane administration at low flow rates did not find evidence of renal injury. Finally, there are substantial data to document the safety of sevoflurane administered for shorter durations or at higher fresh gas flow rates. Therefore, the United States Food and Drug Administration recommends the use of sevoflurane with fresh gas flow rates at least 1 L/min for exposures up to 1 hour and at least 2 L/min for exposures greater than 1 hour. We believe this is a rational, cautious approach based on available data. However, it is important to note that other countries have not recommended such limitations on the clinical use of sevoflurane and problems have not been noted.


Asunto(s)
Anestésicos por Inhalación/efectos adversos , Éteres/efectos adversos , Hidrocarburos Fluorados/efectos adversos , Enfermedades Renales/inducido químicamente , Éteres Metílicos/efectos adversos , Movimientos del Aire , Anestésicos por Inhalación/administración & dosificación , Anestésicos por Inhalación/metabolismo , Interacciones Farmacológicas , Éteres/administración & dosificación , Gases , Humanos , Hidrocarburos Fluorados/administración & dosificación , Riñón/efectos de los fármacos , Riñón/patología , Éteres Metílicos/administración & dosificación , Éteres Metílicos/metabolismo , Sevoflurano
16.
Ann N Y Acad Sci ; 909: 12-24, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10911921

RESUMEN

Neuropathic pain is associated with abnormal tactile and thermal responses that may be extraterritorial to the injured nerve. Importantly, tactile allodynia and thermal hyperalgesia may involve separate pathways, since complete and partial spinal cord lesions have blocked allodynia, but not hyperalgesia, after spinal nerve ligation (SNL). Furthermore, lesions of the dorsal column, and lidocaine microinjected into dorsal column nuclei block only tactile allodynia. Conversely, thermal hyperalgesia, but not tactile allodynia was blocked by desensitization of C-fibers with resiniferotoxin. Therefore, it seems that tactile allodynia is likely to be mediated by large diameter A beta fibers, and not susceptible to modulation by spinal opioids, whereas hyperalgesia is mediated by unmyelinated C-fibers, and is sensitive to blockade by spinal opioids. Additionally, abnormal, spontaneous afferent drive in neuropathic pain may contribute to NMDA-mediated central sensitization by glutamate and by non-opioid actions of spinal dynorphin. Correspondingly, SNL elicited elevation in spinal dynorphin content in spinal segments at and adjacent to the zone of entry of the injured nerve along with signs of neuropathic pain. Antiserum to dynorphin A(1-17) or MK-801 given spinally blocked thermal hyperalgesia, but not tactile allodynia, after SNL, and also restored diminished morphine antinociception. Finally, afferent drive may induce descending facilitation from the rostroventromedial medulla (RVM). Blocking afferent drive with bupivicaine also restored lost potency of PAG morphine, as did CCK antagonists in the RVM. This observation is consistent with afferent drive activating descending facilitation from the RVM, and thus diminishing opioid activity, and may underlie the clinical observation of limited responsiveness of neuropathic pain to opioids.


Asunto(s)
Dolor/etiología , Médula Espinal/fisiología , Animales , Maleato de Dizocilpina/farmacología , Dinorfinas/fisiología , Humanos , Hiperalgesia/etiología , Lidocaína/farmacología , Morfina/farmacología
17.
Brain Res ; 831(1-2): 55-63, 1999 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-10411983

RESUMEN

The co-administration of morphine at spinal (i.th.) and supraspinal (i.c.v.) sites to the same rat produces antinociceptive synergy, a phenomenon which may underlie the clinical analgesic utility of this drug. In animals with peripheral nerve injury, however, the antinociceptive potency and efficacy of i.th. morphine is significantly decreased. Here, the possible loss of spinal/supraspinal morphine antinociceptive synergy and relationship to elevation of spinal dynorphin content was studied. Ligation of lumbar spinal nerves resulted in elevated dynorphin in the ipsilateral lumbar and sacral spinal cord. In sham-operated rats supraspinal/spinal co-administration of morphine produced synergistic antinociception which was unaffected by i.th. MK-801 or dynorphin A((1-17)) antiserum. In nerve-injured rats, i.th. morphine was inactive against tactile allodynia and showed diminished in potency against acute nociception without supraspinal/spinal antinociceptive synergy. Antiserum to dynorphin A((1-17)) or the non-competitive NMDA antagonist MK-801 increased the antinociceptive potency of i.th. morphine, restored supraspinal/spinal morphine antinociceptive synergy and elicited a dose-related i.th. morphine antiallodynic action. These agents did not demonstrate antinociceptive or antiallodynic activity alone and did not alter morphine actions in sham-operated animals. The loss of spinal/supraspinal antinociceptive synergy and lack of antiallodynic activity of spinal morphine appear to be due to the elevation across multiple spinal segments of dynorphin following nerve injury. Pathological actions of elevated dynorphin may directly or indirectly modulate the NMDA receptor, result in a loss of supraspinal/spinal morphine synergy and may thus account for the decreased clinical analgesic efficacy of morphine in peripheral neuropathies.


Asunto(s)
Analgésicos Opioides/farmacología , Maleato de Dizocilpina/uso terapéutico , Dinorfinas/inmunología , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Morfina/farmacología , Neuralgia/tratamiento farmacológico , Animales , Sinergismo Farmacológico , Sueros Inmunes , Inyecciones Intraventriculares , Inyecciones Espinales , Masculino , Dimensión del Dolor , Traumatismos de los Nervios Periféricos , Ratas , Ratas Sprague-Dawley , Tacto/fisiología
18.
J Pain ; 2(4): 241-9, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-14622822

RESUMEN

Inhibition of spinal Fos expression increases formalin-induced nociception and decreases spinal prodynorphin messenger ribonucleic acid (mRNA), suggesting that Fos modulates nociception by inducing dynorphin synthesis. This study tests the hypothesis that Fos modulates sensitivity to other somatic stimuli, such that inhibition of Fos expression will result in tactile allodynia and thermal hyperalgesia. In addition, it correlates the somatosensory effects of inhibition of Fos expression with spinal dynorphin content. Antisense oligodeoxynucleotide (ODN) to c-fos mRNA was administered by intrathecal infusion. Tactile sensitivity was tested by probing the hindpaw with von Frey filaments. Thermal sensitivity was quantitated by using withdrawal latency to radiant heat. Two percent formalin was injected into the dorsal hindpaw, and flinches were quantitated. Fos was quantitated by counting immunoreactive cells. Dynorphin was measured by immunoassay. Intrathecal antisense, but not mismatch, ODN resulted in tactile allodynia, thermal hyperalgesia, and hyperalgesia to formalin-induced nociception. Antisense ODN decreased Fos-like immunoreactivity after formalin injection but did not alter Jun-like immunoreactivity. Antisense ODN had differing effects on spinal dynorphin content, depending on the method of administration. These experiments show a role of Fos in modulating somatosensory sensitivity and suggest that induction of dynorphin synthesis is not the sole mechanism by which Fos does so.

19.
Neurosci Lett ; 215(3): 161-4, 1996 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-8899738

RESUMEN

Cholecystokinin (CCK) may act as an endogenous anti-opioid and blockade of CCK receptors can enhance the potency and efficacy of morphine. This effect is blocked by opioid delta (delta) receptor antagonists, suggesting a tonic inhibitory action of CCK to diminish the release and/or availability of endogenous enkephalins. The present studies have further evaluated this possibility by studying the antiallodynic actions of a CCKB antagonist (L365,260) alone, or in the presence of thiorphan (a neutral endopeptidase inhibitor) in a model of peripheral neuropathy. Animals subjected to nerve injury, but not sham controls, exhibited long lasting, stable mechanical allodynia. Intrathecal (i.t.) administration of L365,260 or thiorphan alone did not alter allodynia. However, co-administration of these compounds produced a significant antiallodynic action which was antagonized by receptor selective doses of naltrindole, an opioid delta receptor antagonist. In addition, antisera to [Leu5]enkephalin, but not to [Met5]enkephalin, also blocked the antiallodynic action of thiorphan plus L365,260. These data suggest that blockade of CCKB receptors may enhance the actions or availability of endogenous [Leu5]enkephalin or a like substance which can elicit a significant antiallodynic action via opioid delta receptors when its degradation is by inhibited by thiorphan. The data suggest that delta opioids are involved in regulation of some aspects of nerve-injury induced pain.


Asunto(s)
Dolor/tratamiento farmacológico , Sistema Nervioso Periférico/efectos de los fármacos , Receptores de Colecistoquinina/antagonistas & inhibidores , Tiorfan/farmacología , Animales , Masculino , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Ratas , Ratas Sprague-Dawley
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA